Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is an intrinsically fluorescent neurotransmitter found in organisms spanning a wide evolutionary range. Serotonin exerts its diverse actions by binding to distinct cell membrane receptors which are classified into many groups. Serotonin receptors are involved in regulating a diverse array of physiological signaling pathways and belong to the family of either G protein-coupled receptors (GPCRs) or ligand-gated ion channels. Serotonergic signaling appears to play a key role in the generation and modulation of various cognitive and behavioral functions such as sleep, mood, pain, anxiety, depression, aggression, and learning. Serotonin receptors act as drug targets for a number of diseases, particularly neuropsychiatric disorders. The signaling mechanism and efficiency of serotonin receptors depend on their amazing ability to rapidly access multiple conformational states. This conformational plasticity, necessary for the wide variety of functions displayed by serotonin receptors, is regulated by binding to various ligands. In this review, we provide a succinct overview of recent developments in generating and analyzing high-resolution structures of serotonin receptors obtained using crystallography and cryo-electron microscopy. Capturing structures of distinct conformational states is crucial for understanding the mechanism of action of these receptors, which could provide important insight for rational drug design targeting serotonin receptors. We further provide emerging information and insight from studies on interactions of membrane lipids (such as cholesterol) with serotonin receptors. We envision that a judicious combination of analysis of high-resolution structures and receptor-lipid interaction would allow a comprehensive understanding of GPCR structure, function and dynamics, thereby leading to efficient drug discovery.
Keywords: Serotonin (5-HT) receptors, Ligand binding site, Active and inactive states, Conserved microswitches, Conformational plasticity, Lipid-receptor interaction
Serotonin: an ancient neurotransmitter with wide-ranging physiology
Serotonin (5-hydroxytryptamine, 5-HT), an intrinsically fluorescent biogenic amine that acts as a neurotransmitter (see Fig. 1a), was discovered as a component of bovine serum ~ 70 years ago in a rather serendipitous manner by Irvine Page and co-workers at Cleveland Clinic (Rapport et al. 1948a, see Whitaker-Azmitia 1999 for a lucid account of the discovery of serotonin). However, the neurobiological relevance of serotonin was established a few years later when it was shown by Betty Twarog that serotonin is a crucial component of the central nervous system (Twarog and Page 1953; apparently, Irvine Page had initial reservations about finding serotonin in mammalian brain (Whitaker-Azmitia 1999)). Serotonin was crystallized (Rapport et al. 1948b) and the chemical structure was determined as 5-hydroxytryptamine (Rapport 1949) by Maurice Rapport.
Fig. 1.
Classification and molecular architecture of human serotonin (5-HT) receptors. a Chemical structure of serotonin (5-hydroxytryptamine). b Over the course of their long evolutionary history, serotonin receptors have differentiated into several subtypes. With the exception of serotonin3 (5-HT3) receptors (which are ligand-gated ion channels), all subtypes of serotonin receptors belong to the GPCR superfamily. The protein sequences of human serotonin receptors (serotonin1A (P08908), serotonin1B (P28222), serotonin1D (P28221), serotonin1E (P28566), serotonin1F (P30939), serotonin2A (P28223), serotonin2B (P41595), serotonin2C (P28335), serotonin3A (P46098), serotonin3B (O95264), serotonin3C (Q8WXA8), serotonin3D (Q70Z44), serotonin3E (A5X5Y0), serotonin4 (Q13639), serotonin5A (P47898), serotonin6 (P50406), and serotonin7 (P34969)) were retrieved from the UniProt database (The UniProt Consortium 2019) and the phylogenetic tree was constructed using the MEGA software (Kumar et al. 2018). Distinct families of serotonin (5-HT) receptors are color coded. General topology of serotonin receptors belonging to the (c) GPCR family, and (d) serotonin3 (5-HT3) receptors which are ion channels. Serotonin receptors that belong to the GPCR family contain seven transmembrane α-helices, whereas serotonin3 (5-HT3) receptors have three distinct domains: a large N-terminal extracellular domain (ECD), a transmembrane domain (TMD) comprising of four α-helices and a large intracellular domain (ICD) between transmembrane helices 3 and 4
Serotonin is found in a wide variety of life forms ranging from primitive single-celled eukaryotes paramecium to humans, and is distributed across a range of tissues, both neural and non-neural (Hen 1992; Nichols and Nichols 2008; Berger et al. 2009). Serotonergic signaling plays a crucial role in the generation and modulation of various cognitive and behavioral functions such as learning, mood, stress, pain, sleep, aggression, depression, anxiety, cognition, and sexual behavior (Sodhi and Sanders-Bush 2004; Olivier 2015). Essentially, serotonin appears to modulate virtually the entire spectrum of human behavioral processes (Berger et al. 2009). This is rather surprising, in view of the fact that majority (~ 95%) of total body serotonin is localized outside the central nervous system (Gershon and Tack 2007) and about one in a million neurons in the central nervous system synthesize serotonin (Berger et al. 2009). Yet, all regions of the brain express multiple serotonin receptors in a tissue-dependent fashion (Nichols and Nichols 2008). As a result, drugs that target serotonin receptors are useful in the treatment of a variety of disorders (Fiorino et al. 2014). This makes drug development involving serotonin receptors an exciting area of research with tremendous opportunity.
Serotonin is intrinsically fluorescent and is a derivative of the naturally occurring fluorescent amino acid tryptophan. Although the intrinsic fluorescence of serotonin was observed a long time back (Udenfriend et al. 1955), a comprehensive understanding of serotonin fluorescence in terms of its excited state molecular properties was reported much later (Chattopadhyay et al. 1996; Maiti et al. 1997; Sarkar et al. 2018).
Serotonin receptors
In evolutionary terms, the serotonin receptors go back to 700–800 million years (Peroutka and Howell 1994). The diverse cellular activity of serotonin in humans is mediated by a total of seventeen receptors that belong to seven distinct subfamilies (5-HT1–7). Out of these, members of six subfamilies (5-HT1–2 and 5-HT4–7) belong to the G protein-coupled receptor (GPCR) family, whereas members of the serotonin3 (5-HT3) receptor subfamily are ligand-gated ion channels (see Fig. 1). Serotonin receptors constitute the largest class of neurotransmitter GPCRs and non-odorant GPCRs (Chattopadhyay 2007; Nichols and Nichols 2008).
Serotonin receptors are instrumental in the generation and modulation of cognitive and behavioral functions such as sleep, mood, pain, addiction, sexual activity, depression, anxiety, alcohol abuse, aggression, learning, and memory (Lin et al. 2014; Cowen and Browning 2015; Stiedl et al. 2015; Carhart-Harris and Nutt 2017; Yohn et al. 2017; Underwood et al. 2018). Malfunctioning of serotonin receptors has been implicated in the etiology of mental disorders such as schizophrenia, depression, suicidal behavior, infantile autism, and obsessive compulsive disorder. For this reason, members of the serotonin receptor family represent a major drug target in all clinical areas (Chattopadhyay 2014; Fiorino et al. 2014; McCorvy and Roth 2015). More importantly, the design of novel drugs that would target specific serotonin receptors with improved selectivity and lower side effects represents an exciting and challenging area of drug development. This would require the availability of high-resolution structures of target serotonin receptors, both in apo and ligand-bound states and knowledge of detailed conformational change(s) upon ligand binding (McCorvy and Roth 2015).
Structural biology of serotonin receptors
Structural insights contribute to our understanding of ligand binding, receptor activation and inactivation mechanisms, conformational flexibility, and other crucial aspects of receptor function. Structure determination of a functionally active, full-length GPCR in native form is challenging due to its inherent conformational dynamics (Manglik et al. 2015; Latorraca et al. 2017; Wingler and Lefkowitz 2020) that poses a problem for X-ray crystallography. As a consequence, available crystal structures of GPCRs are engineered with truncations of N- and/or C-termini and flexible intracellular loops, since these regions interfere in the crystallization process (Wang et al. 2013; Yin et al. 2018; Pal and Chattopadhyay 2019). In addition, incorporation of thermostable mutation(s) in the native sequence of GPCRs and treatment with ligands that significantly help to stabilize the receptor (in most cases inverse agonists) were strategically utilized in almost all the available structures (Lebon et al. 2012; Zhang et al. 2015a). Interestingly, in addition to ligands, GPCR structures are often stabilized in active conformations by replacing ICL3 with T4 lysozyme, apocytochrome b562 (BRIL) or by addition of G-proteins, β-arrestins, and fragment antibodies/nanobodies with affinities for the active conformation (Ghosh et al. 2015).
Most of the structures of serotonin receptors, including serotonin1B (Wang et al. 2013; Yin et al. 2018; Miyagi et al. 2020), serotonin2A (Kimura et al. 2019; Kim et al. 2020), serotonin2B (Liu et al. 2013; Wacker et al. 2013, 2017a; McCorvy et al. 2018), serotonin2C (Peng et al. 2018), and serotonin3A (Hassaine et al. 2014) receptors, have been solved by X-ray crystallography. With the recent resolution revolution using cryo-electron microscopy (cryo-EM) (Safdari et al. 2018), the high-resolution structures of serotonin1B (García-Nafría et al. 2018), serotonin2A (Kim et al. 2020), and serotonin3A (Basak et al. 2018a, b, 2019; Polovinkin et al. 2018; Zarkadas et al. 2020) receptors have been solved. High-field nuclear magnetic resonance (NMR) spectroscopy has emerged as a powerful tool for determination of structure of membrane proteins without any perturbation to the whole protein (Arora and Tamm 2001; Opella 2013). An additional advantage of NMR spectroscopy is that it provides insight into protein dynamics, in addition to structural details. However, no NMR structure of any serotonin receptor is available yet although NMR structure of a chemokine GPCR (CXCR-1) is available without sequence alteration (Park et al. 2012). Compared to X-ray crystallography and NMR spectroscopy, cryo-EM appears to be more advantageous for structural characterization of full-length membrane proteins and protein complexes (such as serotonin receptors coupled with heterotrimeric G-proteins). A unique advantage of cryo-EM is its ability to capture structural details of molecular assemblies (complexes) in physiologically relevant environment (García-Nafría et al. 2018; Basak et al. 2018a, b; Kim et al. 2020).
We discuss below important characteristics of serotonin receptors provided by recently published high-resolution structures, thereby highlighting structural conservation as well as diversity across the receptor subtypes. In addition, we will outline areas that need to be explored further for an in-depth understanding of the molecular mechanisms of activation and desensitization of serotonin receptors and resulting consequence in signaling cascades.
Recent advances in structural biology of serotonin receptors
High-resolution structure determination of membrane proteins involves several challenges that include poor expression of recombinant protein, difficulties in efficient solubilization and low purification yields (Arora and Tamm 2001; Kalipatnapu and Chattopadhyay 2005; Carpenter et al. 2008; Skretas and Georgiou 2008; Chattopadhyay et al. 2015; Wiseman et al. 2020). However, recent improvements in the areas of membrane protein molecular biology and biochemistry along with technical advancements in crystallography, NMR spectroscopy, and cryo-EM have tremendously helped to overcome these challenges toward determination of high-resolution structures of membrane proteins, including serotonin receptors (Lacapère et al. 2007; Bill et al. 2011; Opella 2013; Ishchenko et al. 2017a; Sjöstrand et al. 2017; Smith 2017; Huang et al. 2018; Mandala et al. 2018).
No high-resolution structure of any member of the serotonin receptor family was available until 2013, although by that time, structures of many GPCRs were available. By early 2013, the first structures of serotonin1B receptor in complex with ergotamine (PDB ID: 4IAR, Fig. 2a) and dihydroergotamine (PDB ID: 4IAQ) (Wang et al. 2013) and serotonin2B receptor in complex with ergotamine (PDB ID: 4IB4, Fig. 2c) (Wacker et al. 2013) were solved by X-ray crystallography. From 2013 to 2020, 30 structures from five serotonin receptor subtypes were experimentally determined (see Table 1). More than 60% of the structures of serotonin receptors were GPCRs, including serotonin1B, serotonin2A, serotonin2B, and serotonin2C in complex with a diverse array of ligands. On the other hand, ten structures of the ion channel serotonin3A receptor are available till date and seven of them were reported in 2018 alone. Most of the available structures of serotonin receptors are truncated variants of the full receptor and therefore lack structural information from the N- and C-termini and loop regions (Fig. 2, marked as dashed lines), often necessary for proper functioning of the receptor (Pal and Chattopadhyay 2019). These truncations in the native serotonin receptor sequence were necessary to overcome the inherent receptor dynamics, thereby enabling crystallization (Chattopadhyay 2014). In addition, most of the crystal structures of serotonin receptors belonging to the GPCR family have one or more thermostabilizing mutations in their native sequence (Wacker et al. 2013, 2017a; Liu et al. 2013; Wang et al. 2013; Ishchenko et al. 2017b; McCorvy et al. 2018; Peng et al. 2018; Yin et al. 2018; Kimura et al. 2019). These modifications reduce dynamics of the receptor and help the crystallization process. However, such mutations could perturb the native conformation of the receptor which is likely to alter physiological function.
Fig. 2.
Structural organization of serotonin receptors. Crystal structures of (a) serotonin1B (PDB ID: 4IAR) (Wang et al. 2013), (b) serotonin2A (PDB ID: 6A93) (Kimura et al. 2019), (c) serotonin2B (PDB ID: 4IB4) (Wacker et al. 2013), (d) serotonin2C (PDB ID: 6BQH) (Peng et al. 2018), and (e) serotonin3A (PDB ID: 4PIR) (Hassaine et al. 2014) receptors. The domain organization of serotonin receptors belonging to (f) GPCR family and (g) ligand-gated ion channels. Serotonin receptors belonging to the GPCR family contain a well conserved class A GPCR architecture with an extracellular N-terminus, followed by 7 transmembrane (TM) α-helices (TM1–7) connected by 3 intracellular (ICL1–3) and 3 extracellular loops (ECL1–3), followed by an amphipathic helix 8 (H8) and an intracellular C-terminus. In contrast to other serotonin receptors, serotonin3 receptors are structurally characterized by a twisted β-sandwich N-terminal extracellular domain (ECD), 4 helical transmembrane domains (M1–4), and a large intracellular domain (ICD). The horizontal cylinders represent α-helices, solid lines denote extramembranous linker regions, and dashed lines depict regions where structural information is missing. Structures of GPCRs were generated from their respective PDB IDs using PyMOL Molecular Graphics System (Schrödinger, LLC). See text for more details
Table 1.
Available structures of various serotonin receptors till date
| Receptor | # | Experimental method | PDB ID | Resolution (Å) | Bound ligand/effector | Missing structural region(s) | Reference |
|---|---|---|---|---|---|---|---|
| Serotonin1B | 5 | X-ray | 4IAQ | 2.8 | Dihydroergotamine | N, ICL3 | Wang et al. 2013 |
| X-ray | 4IAR | 2.7 | Ergotamine | N, ICL3 | Wang et al. 2013 | ||
| X-ray | 5V54 | 3.9 | Methiothepin | N, ICL3 | Yin et al. 2018 | ||
| Cryo-EM | 6G79 | 3.78 | EP5/Go1β1γ2 | N | García-Nafría et al. 2018 | ||
| X-ray | 7C61 | 3.0 | Ergotamine | N, ICL3 | Miyagi et al. 2020 | ||
| Serotonin2A | 5 | X-ray | 6A93 | 3.0 | Risperidone | N, C, ICL3 | Kimura et al. 2019 |
| X-ray | 6A94 | 2.9 | Zotepine | N, C, ICL3 | Kimura et al. 2019 | ||
| X-ray | 6WGT | 3.4 | LSD | N, C, ICL3 | Kim et al. 2020 | ||
| X-ray | 6WH4 | 3.4 | Methiothepin | N, C, ICL3 | Kim et al. 2020 | ||
| Cryo-EM | 6WHA | 3.36 | 25-CN-NBOH/Gq/Gβ/Gγ | N, C | Kim et al. 2020 | ||
| Serotonin2B | 8 | X-ray | 4IB4 | 2.7 | Ergotamine | N, C, ICL3 | Wacker et al. 2013 |
| X-ray | 4NC3 | 2.8 | Ergotamine | N, C, ICL3 | Liu et al. 2013 | ||
| X-ray | 5TUD | 3.0 | Ergotamine | N, C, ICL3 | Ishchenko et al. 2017b | ||
| X-ray | 5TVN | 2.9 | LSD | N, C, ICL3 | Wacker et al. 2017a | ||
| X-ray | 6DRX | 3.1 | H8G | N, C, ICL3 | McCorvy et al. 2018 | ||
| X-ray | 6DRY | 2.92 | H8D | N, C, ICL3 | McCorvy et al. 2018 | ||
| X-ray | 6DRZ | 3.1 | H8J | N, C, ICL3 | McCorvy et al. 2018 | ||
| X-ray | 6DS0 | 3.19 | H8M | N, C, ICL3 | McCorvy et al. 2018 | ||
| Serotonin2C | 2 | X-ray | 6BQG | 3.0 | Ergotamine | N, C, ICL3 | Peng et al. 2018 |
| X-ray | 6BQH | 2.7 | Ritanserin | N, C, ICL3 | Peng et al. 2018 | ||
| Serotonin3A | 10 | X-ray | 4PIR | 3.5 | – | – | Hassaine et al. 2014 |
| Cryo-EM | 6BE1 | 4.31 | – | – | Basak et al. 2018b | ||
| Cryo-EM | 6DG7 | 3.32 | Serotonin | – | Basak et al. 2018a | ||
| Cryo-EM | 6DG8 | 3.89 | Serotonin | – | Basak et al. 2018a | ||
| Cryo-EM | 6HIN | 4.1 | Serotonin | – | Polovinkin et al. 2018 | ||
| Cryo-EM | 6HIO | 4.2 | Serotonin | – | Polovinkin et al. 2018 | ||
| Cryo-EM | 6HIQ | 3.2 | Serotonin | – | Polovinkin et al. 2018 | ||
| Cryo-EM | 6HIS | 4.5 | Tropisetron | – | Polovinkin et al. 2018 | ||
| Cryo-EM | 6NP0 | 2.92 | Granisetron | – | Basak et al. 2019 | ||
| Cryo-EM | 6Y1Z | 2.82 | Palonosetron | – | Zarkadas et al. 2020 |
#, number of available structures; N, N-terminus; C, C-terminus; ICL3, intracellular loop 3; cryo-EM, cryo-electron microscopy; X-ray, X-ray diffraction; EP5, 2-[5-[2-[4-(4-cyanophenyl)piperazin-1-yl]-2-oxidanylidene-ethoxy]-1~{H}-indol-3-yl]ethylazanium; LSD, lysergic acid diethylamide; 25-CN-NBOH, (2S,6S)-2-(2,5-dimethoxy-4-bromobenzyl)-6-(2-methoxyphenyl)piperidine; H8G, N,N-diethyl-N′-[(8α)-6-methyl-9,10-didehydroergolin-8-yl]urea; H8D, (8β)-N-[(2S)-1-hydroxybutan-2-yl]-6-methyl-9,10-didehydroergoline-8-carboxamide; H8J, (8α)-N-[(2S)-1-hydroxybutan-2-yl]-1,6-dimethyl-9,10-didehydroergoline-8-carboxamide; H8M, (1S)-1-[(2-chloro-3,4-dimethoxyphenyl)methyl]-6-methyl-2,3,4,9-tetrahydro-1H-β-carboline
In contrast to the available crystal structures of serotonin receptors, the structure of the intracellular loop 3 (ICL3) of serotonin1A receptor was independently solved by NMR spectroscopy (Chen et al. 2011), in which part of the ICL3 showed α-helical conformation, similar to the ICL3 structure of another GPCR, the vasopressin V2 receptor (Bellot et al. 2009). Importantly, the third intracellular loop of the serotonin1A receptor has been shown to be crucial in coupling to and activation of G-proteins (Varrault et al. 1994; Raymond et al. 1999). In addition, it has been previously reported using site-directed mutagenesis that mutations in ICL3 of the serotonin1A receptor alters G-protein coupling from Gi to Gs in a ligand-dependent manner (Malmberg and Strange 2000). Interestingly, in a study using the intrinsic fluorescence of the sole tryptophan residue of ICL3, it was shown that the tryptophan is localized in a restricted environment, probably due to the constraints induced by the peptide secondary structure (Pal et al. 2018). Despite the presence of a binding partner (G-protein), information on ICL3 electron density was found to be absent in the cryo-EM structure of serotonin1B coupled to heterotrimeric Go protein, thereby indicating that such secondary structural elements in ICL3 could be rather transient and/or flexible (García-Nafría et al. 2018). The structural characterization of the ICL3 region of serotonin receptors in the GPCR family is an emerging area and determining its structure could help our understanding of signaling mechanism of serotonin receptors as well as other GPCRs.
General structural features of serotonin receptors
As mentioned above, serotonin receptors are classified into seven subfamilies of receptors (serotonin1–7, see Fig. 1b). Except for the serotonin3 receptor which is a ligand-gated ion channel, the remaining six members are GPCRs which mediate canonical signaling through coupling with one of the three G-proteins (Gαi/o, Gαq/11 and Gαs) (Giulietti et al. 2014). Serotonin receptors belonging to the GPCR family contain a well conserved class A GPCR structural design with an extracellular N-terminus, followed by 7 transmembrane (TM) α-helices (TM1–7) connected by 3 intracellular (ICL1–3) and 3 extracellular (ECL1–3) loops, and an amphipathic helix 8 (H8) and an intracellular C-terminus (Fig. 1c and Fig. 2f). The receptors adopt a tertiary structure resembling a barrel, with the ligand binding site within the membrane. All serotonin receptors in the GPCR family share relatively similar TM domains, except for slight differences in helix tilt and twist, which is probably due to the sequence conservation in the TM regions across serotonin receptors belonging to the GPCR family (McCorvy and Roth 2015; Zhang et al. 2015b; Cvicek et al. 2016). The conserved residues are assigned according to the well-established Ballesteros-Weinstein numbering system (Ballesteros and Weinstein 1995; Isberg et al. 2015), which is a sequence-based generic GPCR residue numbering scheme. It involves a numbering scheme based on the most conserved amino acid residue within each helix of the TM region being assigned as X.50, where X is the helix number denoted serially by 1–7 from N-terminus to C-terminus. The residues toward the C and N-termini are numbered in increasing and decreasing order, respectively. Not surprisingly, these conserved residues designated as X.50 have notable functional importance (Fig. 3). For example, N1.50 in TM1 and D2.50 in TM2 form the sodium ion binding site (Fenalti et al. 2014; McCorvy and Roth 2015), R3.50 with D/E3.49 and Y3.51 form the conserved D(E)RY motif responsible for receptor activation and inactivation (Wacker et al. 2013), W4.50 stabilizes the helical packing of TM2, 3 and 4 (McCorvy and Roth 2015), P5.50 forms the conserved P-I-F motif together with I3.40 and F6.44 and regulates receptor activation (Rasmussen et al. 2011; Wacker et al. 2013), P6.50 probably allows the movement of helices toward the intracellular side (McCorvy and Roth 2015), and P7.50 together with N7.49, Y7.53 and two random amino acids at 7.51 and 7.52 positions form the NPxxY conserved motif, which controls the dynamics of TM7 toward GPCR signaling (Wacker et al. 2013). In addition to the TM domains, serotonin receptors in the GPCR family exhibit conserved structural features in the loop regions. Importantly, a conserved disulfide bridge is formed between C3.25 and CECL2 that connects ECL2 with TM3 and regulates the dynamics of the loop in majority of GPCRs (Unal et al. 2010; Wheatley et al. 2012).
Fig. 3.
A schematic representation of serotonin receptors belonging to the GPCR family showing functionally important residues. The most conserved residues are colored red, whereas, residues with functional significance (specific functionality identified by various studies) are colored black. N1.50 in TM1 and D2.50 in TM2 form the sodium ion binding site (Fenalti et al. 2014; McCorvy and Roth 2015), R3.50 with D/E3.49 and Y3.51 form the conserved D(E)RY motif responsible for receptor activation and inactivation (Wacker et al. 2013). The residue W4.50 stabilizes the helical packing (McCorvy and Roth 2015), P5.50 forms the conserved P-I-F motif together with I3.40 and F6.44 and regulates receptor activation (Rasmussen et al. 2011; Wacker et al. 2013) and P6.50 likely allows the movement of helices (McCorvy and Roth 2015), and P7.50 together with N7.49, Y7.53 and two random amino acids at 7.51 and 7.52 positions forms the NPxxY conserved motif which controls the dynamics of TM7 toward GPCR signaling (Wacker et al. 2013). In addition to the TM domains, serotonin receptors exhibit conserved structural features in the loop regions. Importantly, a conserved disulfide bridge is formed between C3.25 and CECL2 that connects ECL2 with TM3 and regulates the dynamics of the loop in the vast majority of GPCRs
In contrast to all other serotonin receptors, serotonin3 receptors are structurally characterized by a twisted β-sandwich N-terminal extracellular domain (ECD), four helical transmembrane domains (TMD), and a large intracellular domain (ICD, Figs. 1d and 2g). The ligand-gated ion channel is either homopentameric composed of five identical serotonin3A subunits or heteropentameric consisting of a mixture of serotonin3A and one member from any of the other four (serotonin3B, serotonin3C, serotonin3D, and serotonin3E) subunits (Niesler et al. 2003; O’Leary and Cryan 2010; Hensler 2012).
Structural diversity across serotonin receptor subtypes
Structural variations are exhibited not only between serotonin receptor subfamilies and subtypes, but also within a subtype member upon binding to specific type of ligands. Ligands are classified in terms of the downstream signaling responses generated upon binding to the receptor. Ligands that activate their cognate GPCRs are termed agonists, whereas antagonists are ligands that reduce agonist-mediated receptor activity to basal level. In contrast, inverse agonists inhibit even the basal activity of a receptor (Tate 2012; Wacker et al. 2017b). In addition, ligands can modulate signaling upon binding to sites other than the orthosteric binding site (allosteric sites), or can interact with both orthosteric and allosteric sites (bitopic ligands) (Wacker et al. 2017b). Crystal structures of serotonin1B bound to agonist ergotamine (Wang et al. 2013; Fig. 4a–c) and inverse agonist methiothepin (Yin et al. 2018; Fig. 4d–f) display significant conformational perturbations. Compared to the methiothepin-bound inactive structure, the most striking difference in the agonist-bound (active) structure is the outward movement of the intracellular end of TM6, a hallmark of GPCR activation, that expand the cytoplasmic pocket of the TM bundle for coupling downstream signaling effectors (Yin et al. 2018). In addition, the side-chains of the residues in the conserved motifs (sometimes denoted as microswitches) such as D(E)RY, P-I-F, and NPxxY motifs exhibit significant positional and/or rotational displacements between ergotamine and methiothepin-bound structures (Fig. 4a–f). Similar conformational excursions have been found in agonist ergotamine and inverse agonist ritanserin-bound structures of serotonin2C receptor, which represent the active and inactive states of the receptor, respectively (Peng et al. 2018). Molecular modeling studies revealed that while specific ligands could capture the receptor in a specific conformational state, ligand-free model of the serotonin2A receptor shows clear distinction in the extracellular and intracellular end of transmembrane helices, and conserved microswitches from the ligand-bound structures (Mozumder et al. 2019). On the other hand, serotonin3A receptor shows various conformational states not only upon binding to different ligands such as tropisetron and serotonin (Polovinkin et al. 2018), but also upon binding to the same ligand serotonin (Basak et al. 2018a). Structural differences upon ligand binding are largely restricted to the TMD of the serotonin3A receptor, leading to conformational modulation of the pore.
Fig. 4.
Conformational diversity among serotonin receptors belonging to the GPCR family. Conformational states of (a, d, and g) ionic lock, (b, e, and h) P-I-F motif, and (c, f, and i) Y7.53 of NPxxY motif in ergotamine-bound serotonin1B (PDB ID: 4IAR (Wang et al. 2013), cyan), methiothepin-bound serotonin1B (PDB ID: 5V54 (Yin et al. 2018), orange), and ergotamine-bound serotonin2B (PDB ID: 4IB4 (Wacker et al. 2013), magenta) receptors. Whereas the ergotamine-bound serotonin1B adopts a classical agonist-induced active-like state that modulates the canonical signaling pathway, ergotamine-bound serotonin2B receptor structure exhibits conformational characteristics of both the active and inactive states which display strong functional selectivity for β-arrestin signaling. Structures of GPCRs were generated from their respective PDB IDs using PyMOL Molecular Graphics System (Schrödinger, LLC). See text for more details
In contrast to the same serotonin receptor subtypes, receptors from different subfamilies adopt different conformational states upon binding to the same ligand. Crystal structures of ergotamine-bound serotonin1B (Wang et al. 2013) and serotonin2B (Wacker et al. 2013) receptors show structural distinction over intracellular segment of TM5 as well as D(E)RY, P-I-F, and NPxxY motifs (Fig. 4a–c and Fig. 4g–i). Whereas ergotamine-bound serotonin1B receptor adopts a classical agonist-induced active-like state that modulates the canonical signaling pathway, ergotamine-bound serotonin2B receptor structure exhibits conformational characteristics of both the active and inactive states which display strong functional selectivity for β-arrestin signaling (Wacker et al. 2013). Beyond the ligand-bound structures, the structure of serotonin1B receptor bound to the agonist donitriptan and coupled to an engineered Gαo heterotrimer (PDB ID: 6G79) has been solved by cryo-EM (see Fig. 5, García-Nafría et al. 2018), indicating an active state conformation of the intracellular domain.
Fig. 5.
High-resolution cryo-EM structure of serotonin1B receptor coupled to heterotrimeric G-protein. a Surface representation of the receptor (EMDB ID: 4358 (García-Nafría et al. 2018)). b Cartoon representations of serotonin1B receptor (in green), Gαo (in red), Gβ (in cyan), and Gγ (in yellow) molecular models (PDB ID: 6G79 (García-Nafría et al. 2018)) fitted into the cryo-EM density map (semi-transparent blue surface). c The orthosteric binding site in serotonin1B receptor (blue mesh with the molecular model (green) in cartoon) bound to donitriptan (shown as red stick). The blue mesh is derived from high-resolution cryo-EM structure of serotonin1B receptor coupled to heterotrimeric G-protein generated from EMDB ID: 4358 (García-Nafría et al. 2018). Structures of GPCRs were generated from their respective EMDB and PDB IDs using UCSF Chimera (https://www.cgl.ucsf.edu/chimera) and PyMOL Molecular Graphics System (Schrödinger, LLC)
Ligand binding sites in serotonin receptors
Ligand binding sites in GPCRs belonging to the serotonin receptor family are mainly formed by TM3, TM5, TM6, TM7, and ECL2. They are strikingly similar in their overall architecture with subtle divergence exhibited in the binding residues (Fig. 6). It is found that the orthosteric site is embedded deep in the transmembrane core, encompassing helices TM3, TM5, TM6, and TM7, whereas the extended binding site occupies the extracellular portions of TM3, TM5, TM6, and TM7 as well as ECL2. While the former binding site contributes to the ligand binding affinity, the latter site determines ligand selectivity (Wacker et al. 2013; Wang et al. 2013).
Fig. 6.
Comparison of ligand-receptor interactions in serotonin receptors belonging to the GPCR family. Crystal structures showing binding of (a) agonist ergotamine with serotonin1B (PDB ID: 4IAR) (Wang et al. 2013) and (c) inverse agonist risperidone with serotonin2A (PDB ID: 6A93) (Kimura et al. 2019) receptor. Close-up views of the ligand binding site of (b) serotonin1B and (d) serotonin2A receptors in two different orientations are shown. Residues involved in ligand binding are labeled and the most conserved residues at the binding site are highlighted with boxes. Structures of GPCRs were generated from their respective PDB IDs using PyMOL Molecular Graphics System (Schrödinger, LLC). See text for more details
From recent crystal structures of serotonin1B (Wang et al. 2013), serotonin2A (Kimura et al. 2019), serotonin2B (Wacker et al. 2013), and serotonin2C (Peng et al. 2018) receptors bound to agonists or inverse agonists, it is evident that amine-containing ligands mainly interact with the ligand binding pocket through salt bridge between positively charged nitrogen and the carboxyl group of the conserved D3.32 residue (Fig. 6). Mutation of D3.32 significantly disrupts the binding of amine-containing ligand to the serotonin1B receptor (Yin et al. 2018) and the serotonin4 receptor (Claeysen et al. 2003). The residue 3.36 (serine in serotonin2 receptor, threonine in serotonin4 receptor and cysteine in all other serotonin receptors) is localized just a turn below the residue D3.32 and faces the ligand binding pocket. In the serotonin2A receptor, mutation of S3.36 to alanine significantly affects the affinity of primary amine ligands (such as serotonin and tryptamine), but not the affinity of secondary or tertiary amine ligands (such as N,N-dimethyl tryptamine (DMT) and 5-methoxy-DMT) (Almaula et al. 1996). In addition, W6.48, F6.51, F6.52, and Y7.43 are conserved aromatic residues in the binding pocket of all GPCRs belonging to the serotonin receptor family and could form a narrow hydrophobic cleft for important π-π stacking interactions with the ligand molecules containing aromatic ring systems (Fig. 6b, d). Mutations in F6.51, F6.52, and Y7.43 reduce the ligand binding affinity in serotonin2A receptor (Kimura et al. 2019), indicating significant loss of π-π aromatic interactions between receptor and ligand molecules. Importantly, various point mutations in the transmembrane domains of the serotonin1A receptor have helped to identify key residues required for binding of the natural ligand serotonin. Mutations in Asp82 to Asn, Asp116 to Asn, and Ser199 to Ala resulted in a marked reduction in affinity for serotonin, whereas the binding characteristics of the antagonist pindolol remained unaffected (Ho et al. 1992). Interestingly, these mutations did not alter the ability of the receptor to induce GTP hydrolysis upon addition of excess serotonin indicating that the mutants were competent in terms of their interaction with G-proteins.
Compared to the orthosteric site, ligand binding residues in the extended binding sites are more diverse among serotonin receptors in the GPCR family. In serotonin1B receptor, the extended binding pocket is much broader than that of serotonin2B receptor due to the 3.0 Å outward shift of the top of TM5 (Fig. 6a) and the presence of T2095.39 in the corresponding position of the bulky M2185.39 in serotonin2B receptor (Wang et al. 2013). It is important to note that the overall size of the extended binding pocket can significantly change depending upon ligand types. In the serotonin2B receptor, the extended binding site shows 28.6% reduction in overall volume upon binding to lysergic acid diethylamide (LSD) relative to ergotamine (Wacker et al. 2017a). Many single point mutations, including the conserved D3.32, within the orthosteric binding pocket reduce or abolish the binding of LSD at both serotonin1B serotonin2B receptors (Wang et al. 2013).
In contrast to the serotonin receptors belonging to the GPCR family, the ligand binding site of the ion channel serotonin3 receptor is located at the interface of two adjacent subunits at the N-terminal ECD and is formed by three loops from one (the principal) subunit and portions of β-strands and four loops from the adjacent or complementary subunit (Fig. 7). Recent cryo-EM structures of mouse serotonin3A bound to agonist serotonin (Basak et al. 2018a) and antagonist granisetron (Basak et al. 2019) show a well conserved binding pocket nestled by W156 and Y207 of principal subunit and W63, R65, and Y126 of complementary subunit (Fig. 7c). However, W168 experiences a large orientational difference upon binding to agonist or antagonist.
Fig. 7.
Cryo-EM structure of serotonin-bound serotonin3A receptor. a Surface representation shows electron density map of the receptor (EMDB ID: 7882 (Basak et al. 2018a)); b superimposition of the cartoon structure (light blue) of the receptor (PDB ID: 6DG7 (Basak et al. 2018a)) into the cryo-EM density map (semi-transparent purple surface), with the serotonin molecule represented as a magenta stick; c close-up view of the serotonin-binding site. The residue labels on the principal and complementary subunits are marked in black and red, respectively. Structures of GPCRs were generated from their respective EMDB and PDB IDs using UCSF Chimera (https://www.cgl.ucsf.edu/chimera) and PyMOL Molecular Graphics System (Schrödinger, LLC). See text for more details
Ligand-induced activation-inactivation switch in serotonin receptors
The activation or inactivation of all serotonin receptors belonging to the GPCR family is similar to class A GPCRs and is characterized by the rearrangement of the amino acid side-chains in the conserved microswitches (Katritch et al. 2013; McCorvy and Roth 2015; Filipek 2019). The inactive and active states of serotonin receptors are originally thought to be classified based on the presence and absence, respectively, of a salt bridge between D/E3.49 and R3.50 in the D(E)RY motif. However, recent crystal structures of agonist (ergotamine)-bound serotonin1B (Wang et al. 2013) and serotonin2C receptors (Peng et al. 2018), as well as inverse agonist (risperidone)-bound serotonin2A (Kimura et al. 2019) and antagonist (ritanserin)-bound serotonin2C (Peng et al. 2018) receptors, show broken ionic lock in their structures, whereas the ionic lock is preserved in ergotamine-bound serotonin2B receptor structures (Wacker et al. 2013; McCorvy et al. 2018; see Fig. 4a and g). Therefore, the salt bridge interaction in the ionic lock may not be a true indicator of the active and inactive states of the receptor. It is important to note that the intact salt bridge in serotonin2B receptor displays strong functional selectivity of the receptor for β-arrestin signaling (Wacker et al. 2013). This implies that the salt bridge interaction could be important to determine both types of downstream signaling pathways, either canonical signaling via G-proteins or non-canonical signaling via β-arrestins.
In contrast to the D/E3.49 and R3.50 interaction, a conserved salt bridge between R3.50 and E6.30 is crucial to distinguish between active and inactive states. In inactive conformation the interaction between R3.50 and E6.30 remains intact, whereas upon receptor activation, the salt bridge is broken (Wacker et al. 2013; Wang et al. 2013; McCorvy et al. 2018; Peng et al. 2018). Importantly, the inverse agonist (ritanserin)-bound and agonist (ergotamine)-bound serotonin2C receptor structures exhibit presence and absence of the salt bridge, respectively (Peng et al. 2018). Taken together, the salt bridges in the D(E)RY motif play an important role in signaling of serotonin receptors by regulating the movement of the intracellular segment of TM3 and TM6. Importantly, mutagenesis studies further supported the role of the D(E)RY motif to stabilize the ground (inactive) state of the receptor because mutations of this motif in the serotonin2A receptor usually result in constitutive activity (Shapiro et al. 2002).
The conserved P-I-F motif appears to be a critical trigger for receptor activation and inactivation, especially in the light of crystal structures of agonist-bound serotonin1B (Wang et al. 2013), serotonin2B (Wacker et al. 2013; McCorvy et al. 2018) and serotonin2C (Peng et al. 2018) receptors, and inverse agonist-bound serotonin2A (Kimura et al. 2019) and serotonin2C (Peng et al. 2018) receptors. Comparison of active vs. inactive structures reveals inward shifts of P5.50 and F6.44 as well as rotamer switch of I3.40 upon receptor activation, resulting in a rotation and overall movement of TM6 (see Fig. 4b, e). The conserved Y7.53 in the NPxxY motif functions as a “gatekeeper” to allow an opening for a water channel to flow through upon receptor activation (Yuan et al. 2014). However, recent crystal structures show that the NPxxY motif is crucial for receptor signaling either via G-protein or β-arrestin (McCorvy and Roth 2015). While Y7.53 moves and rotates toward the helical bundle upon receptor activation in the canonical pathway, Y7.53 shows more pronounced inward rotation in a β-arrestin biased state (see Fig. 4c–i). Interestingly, in serotonin2C receptor, substituting Ala or Cys for Y7.53 resulted in a marked increase in the basal level of cellular signaling and makes these mutants constitutively active (Rosendorff et al. 2000). The P-I-F and NPxxY motifs in ligand-free ground state structure are believed to lie close to the inactive state conformation.
On the other hand, serotonin3 receptor exhibits significant structural difference in apo, agonist (serotonin)-bound, and antagonist (granisetron)-bound states. Conserved residues surrounding the binding site including W63, R65, Y126, W156, F199, and Y207 undergo rotameric reorientations over apo, serotonin-bound, and granisetron-bound structures. In addition, the loop C and TMD conformations of antagonist-bound serotonin3 receptor are distinct and lie in between apo and agonist-bound conformations (Basak et al. 2019).
Significance of the conformational motions in receptor function
Serotonin receptors exhibit considerable flexibility around the loops as well as the transmembrane helices which are linked to their different signaling functions. While majority of the crystal structures of serotonin receptors provide a high-resolution yet static picture of the lowest energy state, multiple conformational states in ligand-free and -bound conditions would throw light onto the conformational plasticity of the receptor. The ligand binding pocket of serotonin receptors differs from one conformation to another depending upon the nature of the ligand. Although the binding of agonist and antagonist shares some common conserved residues in the binding pocket, the conformational arrangement of these residues significantly differs among agonist- and antagonist-bound states (Fig. 6b, d). In addition, the specificity of orthosteric ligand binding could be provided by allosteric modulators, thereby providing ligand specificity to the receptors (Wootten et al. 2013; Gentry et al. 2015). In some cases, allosteric modulators could also influence receptor responses without the binding of orthosteric ligands (van der Westhuizen et al. 2015). Importantly, information on binding pocket dynamics is important to design selective ligand molecules.
In addition to the ligand binding pocket, the transmembrane helices and the intracellular and extracellular loops exhibit substantial flexibility which is significantly modulated upon ligand binding. Extensive flexibility of the intracellular loops is crucial to accommodate a diverse range of effector molecules (such as G-protein and β-arrestin) (Latorraca et al. 2017). For serotonin receptors, ECL2 is usually the longest extracellular loop and its flexibility is essential to allow a variety of ligand molecules into the binding pocket (Wheatley et al. 2012; Kimura et al. 2019). On the other hand, intracellular loops, mainly ICL2 and ICL3, form the binding cavity for G-protein and therefore conformational dynamics of ICL2 and ICL3 are crucial for signaling by serotonin receptors (Prossnitz et al. 1993; Arora et al. 1995; Gáborik et al. 2003; Chakir et al. 2003; Pydi et al. 2014). Although the dynamic information of ICL3 is missing in available structures due to truncation of the loop region, estimates of motional constraint are available for serotonin1A (Pal et al. 2018) and serotonin2A (Mozumder et al. 2019) receptors. The extracellular and intracellular segments of transmembrane helices, in particular TM3, TM5, and TM6, undergo conformational transition due to different types of ligand binding. A specific conformational state of transmembrane helices is responsible for a specific receptor response over another.
Interestingly, superimposition of X-ray and cryo-EM structures of several serotonin receptors did not exhibit a significant difference in the overall secondary structural fold (see Fig. 8). However, cryo-EM structures of serotonin receptors belonging to the GPCR family showed conformational perturbations in the intracellular region of the receptor relative to the corresponding X-ray structures. This perturbation could be due to the binding of G-protein in the intracellular part of the receptor in the cryo-EM structures. This is supported by the absence of structural difference between X-ray and cryo-EM structures of serotonin3A receptor, which is an ion channel and not coupled to G-proteins. Most of the perturbations in the serotonin3A receptor was largely restricted to the loop regions of the receptor.
Fig. 8.
Overview of X-ray (in red) and cryo-EM (in blue) structures of serotonin receptors. Cartoon representation of (a) serotonin1B (PDB IDs: 4IAQ (red) and 6G79 (blue)), (b) serotonin2A (PDB IDs: 6A93 (red) and 6WHA (blue)) and (c) serotonin3A (PDB IDs: 4PIR (red) and 6HIQ (blue)) receptors. Structural alignment of X-ray and cryo-EM structures exhibits root-mean-square deviation of 0.59 Å, 1.68 Å, and 1.15 Å for serotonin1B, serotonin2A and serotonin3A receptors, respectively. Structures of serotonin receptors were generated from their respective PDB IDs using PyMOL Molecular Graphics System (Schrödinger, LLC). See text for more details
Lipid interactions of serotonin receptors: implications in function, dynamics and oligomerization
A hallmark of membrane proteins and receptors is their interaction with membrane lipids. This is due to the fact that a considerable portion of integral membrane proteins (receptors) lie buried in the membrane bilayer, thereby facilitating their interaction with proximal membrane lipids. For example, molecular dynamics simulations show that the lipid-receptor interface for rhodopsin, a representative GPCR, corresponds to ~ 38% of the total surface area of the receptor (Huber et al. 2004). This raises the obvious possibility that the membrane lipids could be an important modulator of receptor structure and function (Lee 2004). The importance of membrane lipids in the function of GPCRs was apparent even in early days of GPCR research from the adverse effects of delipidation on receptor function (Cerione et al. 1983; Kirilovsky and Schramm 1983). Although the interaction with lipids is important for the function of membrane proteins, it offers a unique challenge to structural biology of membrane proteins and receptors.
Fortunately, the most extensive literature on lipid-GPCR interaction is available for a member of the serotonin receptor family, namely, the serotonin1A receptor (reviewed in Pucadyil and Chattopadhyay 2006; Paila and Chattopadhyay 2010; Jafurulla and Chattopadhyay 2013; Sengupta and Chattopadhyay 2015; Sengupta et al. 2017, 2018). Using a combination of biophysical, biochemical, cell biological, and molecular dynamics simulation approaches, it has been established that membrane lipids, such as cholesterol and sphingolipids, play a crucial role in ligand binding, G-protein coupling, signaling, and oligomerization of the serotonin1A receptor. In addition, in a recent work, it has been shown that the endocytic pathway and intracellular trafficking of the serotonin1A receptor is modulated by cholesterol (Kumar et al. 2019; Kumar and Chattopadhyay 2020). There are a few reports on the sensitivity of the serotonin7 receptor to cholesterol and sphingolipids (Sjögren and Svenningsson 2007a, b; Sjögren et al. 2006). In addition, membrane cholesterol has been reported to play an important role in the oligomerization of the serotonin2a and serotonin2C receptors (Shan et al. 2012; Massaccesi et al. 2020). In contrast to the rich literature on cholesterol sensitivity of serotonin receptors belonging to the GPCR family, there is scant literature on cholesterol sensitivity of serotonin receptors which act as ion channels (Nothdurfter et al. 2010). Over a period of time, lipid (cholesterol) sensitivity of other GPCRs such as the β2-adrenergic, cannabinoid (CB1), bitter taste (T2R4), cholecystokinin (CCK), opioid, oxytocin, galanin, and chemokine receptors has been reported (this list is not exhaustive, only representative; reviewed in Gimpl 2016; Oates and Watts 2011; Jafurulla et al. 2019). In spite of this existing literature, the exact mechanism behind cholesterol sensitivity of GPCR function remains elusive. A school of thought has developed which proposes mechanisms based on direct interaction of cholesterol with GPCRs. Yet, mechanisms based on the ability of cholesterol to modulate GPCR microenvironment in the membrane (fluidity, hydrophobic thickness and dipole potential) have also been postulated. Of course, a combination of these two mechanisms represents another possibility (for a comprehensive discussion, see Jafurulla et al. 2019).
Interestingly, closely bound cholesterol molecules have been reported in crystal structures of the serotonin2A and serotonin2B receptors (Liu et al. 2013; Wacker et al. 2013; Wacker et al. 2017a; McCorvy et al. 2018; Kimura et al. 2019; Kim et al. 2020; see Fig. 9). Closely bound cholesterol molecules were first observed in the crystal structure of the β2-adrenergic receptor (Cherezov et al. 2007) and have become a hallmark for a number of crystal structures of many GPCRs (see Jafurulla et al. 2019 for a comprehensive list of GPCRs displaying bound cholesterol in their structures). In a cautionary fashion, we would like to point out that no clear link has been established yet between GPCRs displaying bound cholesterol in their structures and cholesterol sensitivity in GPCR function. The reason for this lies in the lack of experimental data demonstrating functional cholesterol sensitivity for these receptors. We believe that this link (or lack of it) would become more clear as more functional data become available for GPCRs exhibiting closely bound cholesterol in their structures.
Fig. 9.
Crystal structures of serotonin receptors with bound cholesterol molecules. Bound cholesterol molecules in crystal structures of serotonin receptors belonging to the GPCR family (the corresponding PDB IDs are indicated in parentheses): a serotonin2A receptor (6A93), b serotonin2A receptor (6WGT), c serotonin2B receptor (4IB4), d serotonin2B receptor (4NC3), e serotonin2B receptor (5TVN), and f serotonin2B receptor (6DRX). Snapshots of cholesterol-bound (cholesterol shown in green) structures of serotonin receptors (receptors shown in gray) were generated from their respective PDB structures using UCSF Chimera (https://www.cgl.ucsf.edu/chimera). See text for more details
Specific interaction of GPCRs with membrane cholesterol is often attributed to structural features of these receptors that allow their preferential association with cholesterol (Sarkar and Chattopadhyay 2020). In this context, cholesterol interaction motifs represent putative interaction sites in GPCRs that could induce cholesterol-sensitive function. A popular motif is the cholesterol recognition/interaction amino acid consensus (CRAC) motif characterized by the sequence -L/V-(X)1–5-Y-(X)1–5-R/K- (from N-terminus to C-terminus of the protein), where (X)1–5 represents between one and five residues of any amino acid (Epand 2006; Fantini and Barrantes 2013). The serotonin1A receptor, a prototypical GPCR displaying cholesterol sensitivity, has three CRAC motifs in TM2, 5, and 7 (Jafurulla et al. 2011; see Fig. 10). It should be noted here that mere presence of cholesterol interaction motifs does not necessarily indicate cholesterol-sensitive function of GPCRs. For example, the neurotensin type-1 and secretin receptors have cholesterol consensus motif (CCM) in their TM4 (Hanson et al. 2008). However, both these GPCRs fail to exhibit any appreciable change in downstream signaling response upon cholesterol depletion relative to untreated cells (Harikumar et al. 2005; Oates et al. 2012).
Fig. 10.
A schematic representation depicting the topological features and amino acid sequence of the human serotonin1A receptor embedded in a membrane bilayer. The putative positions of the transmembrane helices of the human serotonin1A receptor were predicted using the crystal structure of the human serotonin1B receptor (PDB ID: 6G79 (García-Nafría et al. 2018)) as a template and the amino acids in the receptor sequence are shown as circles. The receptor has seven transmembrane stretches depicted as putative α-helices. The exact boundary between the membrane and the aqueous phase is not known and therefore location of the amino acid residues relative to the membrane bilayer is putative. The serotonin1A receptor consists of three CRAC motifs in TM2 (CRAC I, shown in yellow), TM5 (CRAC II, shown in green), and TM7 (CRAC III, shown in blue). The palmitoylation at the C-terminus is shown as blue zigzag lines. Adapted and modified with permission from Sarkar and Chattopadhyay 2020, Copyright 2020 Wiley Periodicals, Inc.
Coarse-grain molecular dynamics simulations using the MARTINI force-field helped identify high cholesterol occupancy at the CRAC motif in TM5 of the serotonin1A receptor (Sengupta and Chattopadhyay 2012; see Fig. 11a). An additional advantage offered by simulations is that it provides information on receptor dynamics, generally not available from crystal structures (Torrens-Fontanals et al. 2020). Simulations further showed that these sites are characterized by dynamics associated with cholesterol, typically in the ns-μs timescale. The corresponding energy landscape for association of cholesterol with GPCRs can be viewed as a series of shallow minima, interconnected by low energy barriers (Fig. 11b) (Sengupta and Chattopadhyay 2015). Figure 12 depicts a schematic representation of the dynamics of the fast and slow exchange sites in the serotonin1A receptor. Taken together, the timescales of binding/unbinding of cholesterol at the CRAC sites are in the μs range, while the association/dissociation of cholesterol takes place at the ns timescale at most other sites.
Fig. 11.
Cholesterol interaction hot spots in the human serotonin1A receptor. a Residue-wise maximum occupancy of cholesterol bound to the serotonin1A receptor, obtained by coarse-grain molecular dynamics simulations. Maximum occupancy time (defined as the maximum time a given cholesterol molecule is found at a particular residue during the entire length of the simulation) of cholesterol at each amino acid of the serotonin1A receptor was averaged and normalized over simulations carried out at varying concentrations of cholesterol. The transmembrane helices are represented as gray bands, and CRAC motifs are highlighted with the same color coding as in Fig. 10. Adapted and modified with permission from Sengupta and Chattopadhyay (2012) Identification of cholesterol binding sites in the serotonin1A receptor. J Phys Chem B 116:12991–12,996 DOI: 10.1021/jp309888u), Copyright 2012 American Chemical Society. b A schematic energy landscape corresponding to cholesterol interaction sites in GPCRs. The energy landscape of cholesterol interaction is represented as a series of shallow minima interconnected by low energy barriers. The abscissa can be thought to correspond to individual occupancy sites represented by single residues or by a sub-space at the receptor surface (such as cholesterol consensus motif (CCM) or CRAC sites). The occupancy sites are most likely to be accessed via an exchange with the annular lipids and less often by direct site hopping of cholesterol. Note that the energy barriers and the minima could be modulated by other membrane lipids such as sphingolipids. Reprinted with permission from Sengupta and Chattopadhyay 2015, Copyright 2015 Elsevier B.V
Fig. 12.
Dynamics and diversity of cholesterol occupancy sites in GPCRs. The schematic shows weak, dynamic yet crucial cholesterol interaction sites on GPCRs, governed by cholesterol and receptor dynamics. A few sites show fast exchange with bulk membrane lipids and low occupancy at the receptor surface. Other sites characteristically exhibit slow exchange with bulk lipids and high occupancy at the GPCR surface. Fast ns timescale dynamics is observed in several sites. Site hopping in μs timescale between proximal sites is also observed. Reprinted with permission from Sengupta and Chattopadhyay 2015, Copyright 2015 Elsevier B.V and Sengupta et al. 2018 (https://pubs.acs.org/doi/10.1021/acs.jpcb.8b01657), Copyright 2018 American Chemical Society
Concluding remarks and the road ahead
Available structures of serotonin receptors in complex with a variety of ligands (e.g., agonists, inverse agonists, and antagonists) and effectors (e.g., G-proteins) outline the molecular mechanism of receptor activation and inactivation upon ligand binding in atomic details. Activation and inactivation mechanisms of serotonin receptors share several common yet distinct key features, including movement of helices and amino acid side-chains in the conserved microswitches. Interestingly, significant structural differences have been identified not only between active and inactive states of the receptors, but also among various active conformations, illustrating the process by which receptors activate a specific pathway over another. However, truncations and mutations in the available structures have limited our understanding of conformational dynamics of flexible loop regions that play a pivotal role in receptor activation and inactivation (Haldar et al. 2010; Huang et al., 2016; Pal et al. 2018; Pal and Chattopadhyay 2019; Dijkman et al. 2020). Determination of full-length receptor structures is clearly necessary in order to unravel the in-depth molecular mechanism of regulation of intracellular signaling pathways by serotonin receptors. This is becoming increasingly apparent as more reports appear elucidating the crucial role(s) of the extra- and intracellular loops in activation and signaling of GPCRs (Pal and Chattopadhyay 2019; Ma et al. 2020).
Serotonin receptors act as potential drug targets for a multitude of human diseases. However, drug molecules often interact with more than a single target through a process known as “polypharmacology” and the unintended interactions can lead to side effects. Interaction of a single drug with multiple serotonin receptors is increasingly evident for the treatment of obesity, drug abuse, and schizophrenia (Roth et al. 2004). Although development of ligands targeting selective serotonin receptors (prospective drug molecules) with a defined polypharmacological profile (the so called magic bullet approach) has proved to be extremely difficult, structure-based rational drug design approach has the potential to develop target-selective drugs with improved therapeutic profiles. However, it requires multitude of high-resolution receptor structures with diverse ligands to elucidate the mechanism of polypharmacology at multiple defined drug targets. The other approach to address this issue is to design non-selective drugs (sometimes termed as “magic shotguns”) that interact with several receptors as targets and could result in effective therapeutic intervention (Roth et al. 2004). Having said that, it may be noted that recent crystal and cryo-EM structures of serotonin receptors have helped to design novel drugs selective to specific serotonin receptors, including serotonin1A (Becker et al. 2016), serotonin1B (Tatarczyńska et al. 2004), serotonin2B (Kovács et al. 2003), serotonin2C (Kovács et al. 2003), and serotonin3 (Gan 2005; Theriot et al. 2020) receptors.
Notably, several mutations of serotonin receptors are reported to be associated with different psychiatric disorders (Drago et al. 2008; Banlaki et al. 2015; Xia et al. 2018). Mapping the structural tweaking caused by the important pathological mutants of serotonin to understand the functional traits of the mutant receptors relative to the wild-type receptor would be of immense relevance. We believe that recent advances in high-resolution structural biology of GPCRs could provide new insights in this area.
As mentioned above, serotonin receptors are ancient receptors from an evolutionary perspective and represent hubs for an array of signaling pathways that are associated with human emotion, happiness, anxiety, depression, and overall health. Their usefulness as drug targets go beyond neuropsychiatric disorders and extends up to cancer (Fiorino et al. 2014). The important role played by serotonin receptors in human health and happiness has contributed to their immense popularity. Nonetheless, serotonin receptors currently appear to be at a crossroad, since some of the key serotonin receptors (such as the serotonin1A receptor) has not lend itself to purification and structural analysis yet. Having said that, serotonin receptors (especially, the serotonin1A receptor) enjoy a special position among GPCRs in terms of their sensitivity to lipids in general and cholesterol in particular due to the breadth and depth of available information in this area. This would help in translating emerging structural details in terms of a functional membrane interaction paradigm. With tremendous advancements in GPCR structural biology in the recent past, it is envisioned that these challenges would be overcome soon, enabling us to carry out in-depth analysis of serotonin receptors at all levels allowing fine-tuned effective drug development against the backdrop of receptor-lipidome interaction.
Acknowledgments
S.M. and A.B. thank DST for Innovation in Science Pursuit for Inspired Research (INSPIRE) Fellowships. P.S. thanks CSIR for the award of Shyama Prasad Mukherjee Fellowship. A.C. is a Distinguished Visiting Professor at the Indian Institute of Technology Bombay (Mumbai), and Adjunct Professor at Tata Institute of Fundamental Research (Mumbai) and Indian Institute of Science Education and Research (Kolkata), and an Honorary Professor at the Jawaharlal Nehru Centre for Advanced Scientific Research (Bengaluru). We thank members of the Chattopadhyay laboratory for their comments and discussions.
Funding
This work was supported by Science and Engineering Research Board (SERB) Distinguished Fellowship grant to A.C., Council of Scientific and Industrial Research (CSIR) Network project UNSEEN (BSC0113) and Department of Science and Technology (DST) Ramanujan fellowship to Sujoy Mukherjee.
Compliance with ethical standards
Conflict of interest
The authors declare that they have no conflict of interest.
Footnotes
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Parijat Sarkar and Sukanya Mozumder contributed equally to this work.
References
- Almaula N, Ebersole BJ, Zhang D, et al. Mapping the binding site pocket of the serotonin 5-hydroxytryptamine2A receptor. Ser3.36(159) provides a second interaction site for the protonated amine of serotonin but not of lysergic acid diethylamide or bufotenin. J Biol Chem. 1996;271:14672–14675. doi: 10.1074/jbc.271.25.14672. [DOI] [PubMed] [Google Scholar]
- Arora A, Tamm LK. Biophysical approaches to membrane protein structure determination. Curr Opin Struct Biol. 2001;11:540–547. doi: 10.1016/s0959-440x(00)00246-3. [DOI] [PubMed] [Google Scholar]
- Arora KK, Sakai A, Catt KJ. Effects of second intracellular loop mutations on signal transduction and internalization of the gonadotropin-releasing hormone receptor. J Biol Chem. 1995;270:22820–22826. doi: 10.1074/jbc.270.39.22820. [DOI] [PubMed] [Google Scholar]
- Ballesteros JA, Weinstein H. Integrated methods for the construction of three-dimensional models and computational probing of structure-function relations in G protein-coupled receptors. Methods Neurosci. 1995;25:366–428. doi: 10.1016/S1043-9471(05)80049-7. [DOI] [Google Scholar]
- Banlaki Z, Elek Z, Nanasi T, et al. Polymorphism in the serotonin receptor 2a (HTR2A) gene as possible predisposal factor for aggressive traits. PLoS One. 2015;10:e0117792. doi: 10.1371/journal.pone.0117792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basak S, Gicheru Y, Rao S, et al. Cryo-EM reveals two distinct serotonin-bound conformations of full-length 5-HT3A receptor. Nature. 2018;563:270–274. doi: 10.1038/s41586-018-0660-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basak S, Gicheru Y, Samanta A, et al. Cryo-EM structure of 5-HT3A receptor in its resting conformation. Nat Commun. 2018;9:514. doi: 10.1038/s41467-018-02997-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basak S, Gicheru Y, Kapoor A, et al. Molecular mechanism of setron-mediated inhibition of full-length 5-HT3A receptor. Nat Commun. 2019;10:3225. doi: 10.1038/s41467-019-11142-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Becker G, Bolbos R, Costes N, et al. Selective serotonin 5-HT1A receptor biased agonists elicit distinct brain activation patterns: a pharmacoMRI study. Sci Rep. 2016;6:26633. doi: 10.1038/srep26633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bellot G, Granier S, Bourguet W, et al. Structure of the third intracellular loop of the vasopressin V2 receptor and conformational changes upon binding to gC1qR. J Mol Biol. 2009;388:491–507. doi: 10.1016/j.jmb.2009.02.065. [DOI] [PubMed] [Google Scholar]
- Berger M, Gray JA, Roth BL. The expanded biology of serotonin. Ann Rev Med. 2009;60:355–366. doi: 10.1146/annurev.med.60.042307.110802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bill RM, Henderson PJF, Iwata S, et al. Overcoming barriers to membrane protein structure determination. Nat Biotechnol. 2011;29:335–340. doi: 10.1038/nbt.1833. [DOI] [PubMed] [Google Scholar]
- Carhart-Harris RL, Nutt DJ. Serotonin and brain function: a tale of two receptors. J Psychopharmacol. 2017;31:1091–1120. doi: 10.1177/0269881117725915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carpenter EP, Beis K, Cameron AD, Iwata S. Overcoming the challenges of membrane protein crystallography. Curr Opin Struct Biol. 2008;18:581–586. doi: 10.1016/j.sbi.2008.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerione RA, Strulovici B, Benovic JL, et al. Reconstitution of β-adrenergic receptors in lipid vesicles: affinity chromatography-purified receptors confer catecholamine responsiveness on a heterologous adenylate cyclase system. Proc Natl Acad Sci U S A. 1983;80:4899–4903. doi: 10.1073/pnas.80.16.4899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chakir K, Xiang Y, Yang D, et al. The third intracellular loop and the carboxyl terminus of β2-adrenergic receptor confer spontaneous activity of the receptor. Mol Pharmacol. 2003;64:1048–1058. doi: 10.1124/mol.64.5.1048. [DOI] [PubMed] [Google Scholar]
- Chattopadhyay A. Serotonin receptors in neurobiology. In: Simon SA, MAL N, editors. New Frontiers in Neuroscience series. Boca Raton: CRC Press/Taylor & Francis; 2007. [Google Scholar]
- Chattopadhyay A. GPCRs: lipid-dependent membrane receptors that act as drug targets. Adv Biol. 2014;2014:143023. doi: 10.1155/2014/143023. [DOI] [Google Scholar]
- Chattopadhyay A, Rukmini R, Mukherjee S. Photophysics of a neurotransmitter: ionization and spectroscopic properties of serotonin. Biophys J. 1996;71:1952–1960. doi: 10.1016/S0006-3495(96)79393-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chattopadhyay A, Rao BD, Jafurulla M. Solubilization of G protein-coupled receptors: a convenient strategy to explore lipid-receptor interaction. Methods Enzymol. 2015;557:117–134. doi: 10.1016/bs.mie.2015.01.001. [DOI] [PubMed] [Google Scholar]
- Chen AS, Kim YM, Gayen S, et al. NMR structural study of the intracellular loop 3 of the serotonin 5-HT1A receptor and its interaction with calmodulin. Biochim Biophys Acta. 2011;1808:2224–2232. doi: 10.1016/j.bbamem.2011.05.014. [DOI] [PubMed] [Google Scholar]
- Cherezov V, Rosenbaum DM, Hanson MA, et al. High-resolution crystal structure of an engineered human β2-adrenergic G protein-coupled receptor. Science. 2007;318:1258–1265. doi: 10.1126/science.1150577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Claeysen S, Joubert L, Sebben M, Bockaert J, Dumuis A. A single mutation in the 5-HT4 receptor (5-HT4-R D100(3.32)A) generates a Gs-coupled receptor activated exclusively by synthetic ligands (RASSL) J Biol Chem. 2003;278:699–702. doi: 10.1074/jbc.C200588200. [DOI] [PubMed] [Google Scholar]
- Cowen PJ, Browning M. What has serotonin to do with depression? World Psychiatry. 2015;14:158–160. doi: 10.1002/wps.20229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cvicek V, Goddard WA, III, Abrol R. Structure-based sequence alignment of the transmembrane domains of all human GPCRs: phylogenetic, structural and functional implications. PLoS Comput Biol. 2016;12:e1004805. doi: 10.1371/journal.pcbi.1004805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dijkman PM, Muñoz-García JC, Lavington SR et al (2020) Conformational dynamics of a G protein-coupled receptor helix 8 in lipid membranes. Sci Adv 6:eaav8207. 10.1126/sciadv.aav8207 [DOI] [PMC free article] [PubMed]
- Drago A, De Ronchi D, Serretti A. 5-HT1A gene variants and psychiatric disorders: a review of current literature and selection of SNPs for future studies. Int J Neuropsychopharmacol. 2008;11:701–721. doi: 10.1017/S1461145707008218. [DOI] [PubMed] [Google Scholar]
- Epand RM. Cholesterol and the interaction of proteins with membrane domains. Prog Lipid Res. 2006;45:279–294. doi: 10.1016/j.plipres.2006.02.001. [DOI] [PubMed] [Google Scholar]
- Fantini J, Barrantes FJ. How cholesterol interacts with membrane proteins: an exploration of cholesterol-binding sites including CRAC, CARC, and tilted domains. Front Physiol. 2013;4:31. doi: 10.3389/fphys.2013.00031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fenalti G, Giguere PM, Katritch V, et al. Molecular control of δ-opioid receptor signalling. Nature. 2014;506:191–196. doi: 10.1038/nature12944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Filipek S. Molecular switches in GPCRs. Curr Opin Struct Biol. 2019;55:114–120. doi: 10.1016/j.sbi.2019.03.017. [DOI] [PubMed] [Google Scholar]
- Fiorino F, Severino B, Magli E, et al. 5-HT1A receptor: an old target as a new attractive tool in drug discovery from central nervous system to cancer. J Med Chem. 2014;57:4407–4426. doi: 10.1021/jm400533t. [DOI] [PubMed] [Google Scholar]
- Gáborik Z, Jagadeesh G, Zhang M, et al. The role of a conserved region of the second intracellular loop in AT1 angiotensin receptor activation and signaling. Endocrinology. 2003;144:2220–2228. doi: 10.1210/en.2002-0135. [DOI] [PubMed] [Google Scholar]
- Gan TJ. Selective serotonin 5-HT3 receptor antagonists for postoperative nausea and vomiting. Are they all the same? CNS Drugs. 2005;19:225–238. doi: 10.2165/00023210-200519030-00004. [DOI] [PubMed] [Google Scholar]
- García-Nafría J, Nehmé R, Edwards PC, Tate CG. Cryo-EM structure of the serotonin 5-HT1B receptor coupled to heterotrimeric Go. Nature. 2018;558:620–623. doi: 10.1038/s41586-018-0241-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gentry PR, Sexton PM, Christopoulos A. Novel allosteric modulators of G protein-coupled receptors. J Biol Chem. 2015;290:19478–19488. doi: 10.1074/jbc.R115.662759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gershon MD, Tack J. The serotonin signaling system: from basic understanding to drug development for functional GI disorders. Gastroenterology. 2007;132:397–414. doi: 10.1053/j.gastro.2006.11.002. [DOI] [PubMed] [Google Scholar]
- Ghosh E, Kumari P, Jaiman D, Shukla AK. Methodological advances: the unsung heroes of the GPCR structural revolution. Nat Rev Mol Cell Biol. 2015;16:69–81. doi: 10.1038/nrm3933. [DOI] [PubMed] [Google Scholar]
- Gimpl G. Interaction of G protein coupled receptors and cholesterol. Chem Phys Lipids. 2016;199:61–73. doi: 10.1016/j.chemphyslip.2016.04.006. [DOI] [PubMed] [Google Scholar]
- Giulietti M, Vivenzio V, Piva F, et al. How much do we know about the coupling of G-proteins to serotonin receptors? Mol Brain. 2014;7:49. doi: 10.1186/s13041-014-0049-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haldar S, Raghuraman H, Namani T, et al. Membrane interaction of the N-terminal domain of chemokine receptor CXCR1. Biochim Biophys Acta. 2010;1798:1056–1061. doi: 10.1016/j.bbamem.2010.02.02. [DOI] [PubMed] [Google Scholar]
- Hanson MA, Cherezov V, Griffith MT, et al. A specific cholesterol binding site is established by the 2.8 Å structure of the human β2-adrenergic receptor. Structure. 2008;16:897–905. doi: 10.1016/j.str.2008.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harikumar KG, Puri V, Singh RD, et al. Differential effects of modification of membrane cholesterol and sphingolipids on the conformation, function, and trafficking of the G protein-coupled cholecystokinin receptor. J Biol Chem. 2005;280:2176–2185. doi: 10.1074/jbc.M410385200. [DOI] [PubMed] [Google Scholar]
- Hassaine G, Deluz C, Grasso L, et al. X-ray structure of the mouse serotonin 5-HT3 receptor. Nature. 2014;512:276–281. doi: 10.1038/nature13552. [DOI] [PubMed] [Google Scholar]
- Hen R. Of mice and flies: commonalities among 5-HT receptors. Trends Pharmacol Sci. 1992;13:160–165. doi: 10.1016/0165-6147(92)90054-A. [DOI] [PubMed] [Google Scholar]
- Hensler JG (2012) Serotonin. In: Brady S, Siegel G, Albers RW, Price D (eds) Basic Neurochemistry, 8th edn. Academic Press, pp 300-322. 10.1016/B978-0-12-374947-5.00015-8
- Ho BY, Karschin A, Branchek T, Davidson N, Lester HA. The role of conserved aspartate and serine residues in ligand binding and in function of the 5-HT1A receptor: a site-directed mutation study. FEBS Lett. 1992;12:259–262. doi: 10.1016/0014-5793(92)80948-g. [DOI] [PubMed] [Google Scholar]
- Huang J, Lakkaraju SK, Coop A, MacKerell AD., Jr Conformational heterogeneity of intracellular loop 3 of the μ-opioid G-protein coupled receptor. J Phys Chem B. 2016;120:11897–11904. doi: 10.1021/acs.jpcb.6b09351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang C-Y, Olieric V, Howe N, et al. In situ serial crystallography for rapid de novo membrane protein structure determination. Commun Biol. 2018;1:124. doi: 10.1038/s42003-018-0123-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huber T, Botelho AV, Beyer K, Brown MF. Membrane model for the G-protein-coupled receptor rhodopsin: hydrophobic interface and dynamical structure. Biophys J. 2004;86:2078–2100. doi: 10.1016/S0006-3495(04)74268-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Isberg V, de Graaf C, Bortolato A, et al. Generic GPCR residue numbers—aligning topology maps while minding the gaps. Trends Pharmacol Sci. 2015;36:22–31. doi: 10.1016/j.tips.2014.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishchenko A, Abola EE, Cherezov V. Crystallization of membrane proteins: an overview. Methods Mol Biol. 2017;1607:117–141. doi: 10.1007/978-1-4939-7000-1_5. [DOI] [PubMed] [Google Scholar]
- Ishchenko A, Wacker D, Kapoor M, et al. Structural insights into the extracellular recognition of the human serotonin 2B receptor by an antibody. Proc Natl Acad Sci U S A. 2017;114:8223–8228. doi: 10.1073/pnas.1700891114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jafurulla M, Chattopadhyay A. Membrane lipids in the function of serotonin and adrenergic receptors. Curr Med Chem. 2013;20:47–55. doi: 10.2174/09298673130106. [DOI] [PubMed] [Google Scholar]
- Jafurulla M, Tiwari S, Chattopadhyay A. Identification of cholesterol recognition amino acid consensus (CRAC) motif in G-protein coupled receptors. Biochem Biophys Res Commun. 2011;404:569–573. doi: 10.1016/j.bbrc.2010.12.031. [DOI] [PubMed] [Google Scholar]
- Jafurulla M, Kumar GA, Rao BD, Chattopadhyay A. A critical analysis of molecular mechanisms underlying membrane cholesterol sensitivity of GPCRs. Adv Exp Med Biol. 2019;1115:21–52. doi: 10.1007/978-3-030-04278-3_2. [DOI] [PubMed] [Google Scholar]
- Kalipatnapu S, Chattopadhyay A. Membrane protein solubilization: recent advances and challenges in solubilization of serotonin1A receptors. IUBMB Life. 2005;57:505–512. doi: 10.1080/15216540500167237. [DOI] [PubMed] [Google Scholar]
- Katritch V, Cherezov V, Stevens RC. Structure-function of the G protein-coupled receptor superfamily. Annu Rev Pharmacol Toxicol. 2013;53:531–556. doi: 10.1146/annurev-pharmtox-032112-135923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim K, Che T, Panova O, et al. Structure of a hallucinogen-activated Gq-coupled 5-HT2A serotonin receptor. Cell. 2020;182:1574–1588. doi: 10.1016/j.cell.2020.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimura KT, Asada H, Inoue A, et al. Structures of the 5-HT2A receptor in complex with the antipsychotics risperidone and zotepine. Nat Struct Mol Biol. 2019;26:121–128. doi: 10.1038/s41594-018-0180-z. [DOI] [PubMed] [Google Scholar]
- Kirilovsky J, Schramm M. Delipidation of a β-adrenergic receptor preparation and reconstitution by specific lipids. J Biol Chem. 1983;258:6841–6849. doi: 10.1016/S0021-9258(18)32299-3. [DOI] [PubMed] [Google Scholar]
- Kovács A, Gacsályi I, Wellmann J, et al. Effects of EGIS-7625, a selective and competitive 5-HT2B receptor antagonist. Cardiovasc Drugs Ther. 2003;17:427–434. doi: 10.1023/B:CARD.0000015857.96371.43. [DOI] [PubMed] [Google Scholar]
- Kumar GA, Chattopadhyay A. Statin-induced chronic cholesterol depletion switches GPCR endocytosis and trafficking: insights from the serotonin1A receptor. ACS Chem Neurosci. 2020;11:453–465. doi: 10.1021/acschemneuro.9b00659. [DOI] [PubMed] [Google Scholar]
- Kumar S, Stecher G, Li M, Knyaz C, Tamura K. MEGA X: molecular evolutionary genetics analysis across computing platforms. Mol Biol Evol. 2018;35:1547–1549. doi: 10.1093/molbev/msy096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar GA, Sarkar P, Jafurulla M, et al. Exploring endocytosis and intracellular trafficking of the human serotonin1A receptor. Biochemistry. 2019;58:2628–2641. doi: 10.1021/acs.biochem.9b00033. [DOI] [PubMed] [Google Scholar]
- Lacapère J-J, Pebay-Peyroula E, Neumann J-M, Etchebest C. Determining membrane protein structures: still a challenge! Trends Biochem Sci. 2007;32:259–270. doi: 10.1016/j.tibs.2007.04.001. [DOI] [PubMed] [Google Scholar]
- Latorraca NR, Venkatakrishnan AJ, Dror RO. GPCR dynamics: structures in motion. Chem Rev. 2017;117:139–155. doi: 10.1021/acs.chemrev.6b00177. [DOI] [PubMed] [Google Scholar]
- Lebon G, Warne T, Tate CG. Agonist-bound structures of G protein-coupled receptors. Curr Opin Struct Biol. 2012;22:482–490. doi: 10.1016/j.sbi.2012.03.007. [DOI] [PubMed] [Google Scholar]
- Lee AG. How lipids affect the activities of integral membrane proteins. Biochim Biophys Acta. 2004;1666:62–87. doi: 10.1016/j.bbamem.2004.05.012. [DOI] [PubMed] [Google Scholar]
- Lin S-H, Lee L-T, Yang YK. Serotonin and mental disorders: a concise review on molecular neuroimaging evidence. Clin Psychopharmacol Neurosci. 2014;12:196–202. doi: 10.9758/cpn.2014.12.3.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu W, Wacker D, Gati C, et al. Serial femtosecond crystallography of G protein-coupled receptors. Science. 2013;342:1521–1524. doi: 10.1126/science.1244142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma X, Hu Y, Batebi H, et al. Analysis of β2AR-Gs and β2AR-Gi complex formation by NMR spectroscopy. Proc Natl Acad Sci U S A. 2020;117:23096–23105. doi: 10.1073/pnas.2009786117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maiti S, Shear JB, Williams RM, et al. Measuring serotonin distribution in live cells with three-photon excitation. Science. 1997;275:530–532. doi: 10.1126/science.275.5299.530. [DOI] [PubMed] [Google Scholar]
- Malmberg Å, Strange PG. Site-directed mutations in the third intracellular loop of the serotonin 5-HT1A receptor alter G protein coupling from Gi to Gs in a ligand-dependent manner. J Neurochem. 2000;75:1283–1293. doi: 10.1046/j.1471-4159.2000.751283.x. [DOI] [PubMed] [Google Scholar]
- Mandala VS, Williams JK, Hong M. Structure and dynamics of membrane proteins from solid-state NMR. Annu Rev Biophys. 2018;47:201–222. doi: 10.1146/annurev-biophys-070816-033712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manglik A, Kim TH, Masureel M, et al. Structural insights into the dynamic process of β2-adrenergic receptor signaling. Cell. 2015;161:1101–1111. doi: 10.1016/j.cell.2015.04.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Massaccesi L, Laudadio E, Mobbili G, et al. Cholesterol-mediated oligomerization pathways of serotonin G-coupled receptor 5-HT2C. Int J Biol Macromol. 2020;160:1090–1100. doi: 10.1016/j.ijbiomac.2020.05.231. [DOI] [PubMed] [Google Scholar]
- McCorvy JD, Roth BL. Structure and function of serotonin G protein-coupled receptors. Pharmacol Ther. 2015;150:129–142. doi: 10.1016/j.pharmthera.2015.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCorvy JD, Wacker D, Wang S, et al. Structural determinants of 5-HT2B receptor activation and biased agonism. Nat Struct Mol Biol. 2018;25:787–796. doi: 10.1038/s41594-018-0116-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyagi H, Asada H, Suzuki M et al (2020) The discovery of a new antibody for BRIL-fused GPCR structure determination. Sci Rep 10:11669. https//doi.org/10.1038/s41598-020-68355-x [DOI] [PMC free article] [PubMed]
- Mozumder S, Bej A, Srinivasan K, et al. Comprehensive structural modeling and preparation of human 5-HT2A G-protein coupled receptor in functionally active form. Biopolymers. 2019;111:e23329. doi: 10.1002/bip.23329. [DOI] [PubMed] [Google Scholar]
- Nichols DE, Nichols CD. Serotonin receptors. Chem Rev. 2008;108:1614–1641. doi: 10.1021/cr078224o. [DOI] [PubMed] [Google Scholar]
- Niesler B, Frank B, Kapeller J, Rappold GA. Cloning, physical mapping and expression analysis of the human 5-HT3 serotonin receptor-like genes HTR3C, HTR3D and HTR3E. Gene. 2003;310:101–111. doi: 10.1016/S0378-1119(03)00503-1. [DOI] [PubMed] [Google Scholar]
- Nothdurfter C, Tanasic S, Di Benedetto B, et al. Impact of lipid raft integrity on 5-HT3 receptor function and its modulation by antidepressants. Neuropsychopharmacol. 2010;35:1510–1519. doi: 10.1038/npp.2010.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Leary OF, Cryan JF (2010) The behavioral genetics of serotonin: relevance to anxiety and depression. In: Müller C, Jacobs B (eds) Handbook of Behavioral Neuroscience. Elsevier, pp 749–789. 10.1016/S1569-7339(10)70109-0
- Oates J, Watts A. Uncovering the intimate relationship between lipids, cholesterol and GPCR activation. Curr Opin Struct Biol. 2011;21:802–807. doi: 10.1016/j.sbi.2011.09.007. [DOI] [PubMed] [Google Scholar]
- Oates J, Faust B, Attrill H, et al. The role of cholesterol on the activity and stability of neurotensin receptor 1. Biochim Biophys Acta. 2012;1818:2228–2233. doi: 10.1016/j.bbamem.2012.04.010. [DOI] [PubMed] [Google Scholar]
- Olivier B. Serotonin: a never-ending story. Eur J Pharmacol. 2015;753:2–18. doi: 10.1016/j.ejphar.2014.10.031. [DOI] [PubMed] [Google Scholar]
- Opella SJ. Structure determination of membrane proteins in their native phospholipid bilayer environment by rotationally aligned solid-state NMR spectroscopy. Acc Chem Res. 2013;46:2145–2153. doi: 10.1021/ar400067z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paila YD, Chattopadhyay A. Membrane cholesterol in the function and organization of G-protein coupled receptors. Subcell Biochem. 2010;51:439–466. doi: 10.1007/978-90-481-8622-8_16. [DOI] [PubMed] [Google Scholar]
- Pal S, Chattopadhyay A. Extramembranous regions in G protein-coupled receptors: Cinderella in receptor biology? J Membr Biol. 2019;252:483–497. doi: 10.1007/s00232-019-00092-3. [DOI] [PubMed] [Google Scholar]
- Pal S, Aute R, Sarkar P, et al. Constrained dynamics of the sole tryptophan in the third intracellular loop of the serotonin1A receptor. Biophys Chem. 2018;240:34–41. doi: 10.1016/j.bpc.2018.05.008. [DOI] [PubMed] [Google Scholar]
- Park SH, Das BB, Casagrande F, et al. Structure of the chemokine receptor CXCR1 in phospholipid bilayers. Nature. 2012;491:779–783. doi: 10.1038/nature11580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng Y, McCorvy JD, Harpsøe K, et al. 5-HT2C receptor structures reveal the structural basis of GPCR polypharmacology. Cell. 2018;172:719–730.e14. doi: 10.1016/j.cell.2018.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peroutka SJ, Howell TA. The molecular evolution of G protein-coupled receptors: focus on 5-hydroxytryptamine receptors. Neuropharmacology. 1994;33:319–324. doi: 10.1016/0028-3908(94)90060-4. [DOI] [PubMed] [Google Scholar]
- Polovinkin L, Hassaine G, Perot J, et al. Conformational transitions of the serotonin 5-HT3 receptor. Nature. 2018;563:275–279. doi: 10.1038/s41586-018-0672-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prossnitz ER, Quehenberger O, Cochrane CG, Ye RD. The role of the third intracellular loop of the neutrophil N-formyl peptide receptor in G protein coupling. Biochem J. 1993;294:581–587. doi: 10.1042/bj2940581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pucadyil TJ, Chattopadhyay A. Role of cholesterol in the function and organization of G-protein coupled receptors. Prog Lipid Res. 2006;45:295–333. doi: 10.1016/j.plipres.2006.02.002. [DOI] [PubMed] [Google Scholar]
- Pydi SP, Singh N, Upadhyaya J, et al. The third intracellular loop plays a critical role in bitter taste receptor activation. Biochim Biophys Acta. 2014;1838:231–236. doi: 10.1016/j.bbamem.2013.08.009. [DOI] [PubMed] [Google Scholar]
- Rapport MM. Serum vasoconstrictor (serotonin). V The presence of creatinine in the complex A proposed structure of the vasoconstrictor principle. J Biol Chem. 1949;180:961–969. doi: 10.1016/S0021-9258(19)51208-X. [DOI] [PubMed] [Google Scholar]
- Rapport MM, Green AA, Page IH. Partial purification of the vasoconstrictor in beef serum. J Biol Chem. 1948;174:735–741. doi: 10.1016/S0021-9258(18)57355-5. [DOI] [PubMed] [Google Scholar]
- Rapport MM, Green AA, Page IH. Crystalline serotonin. Science. 1948;108:329–330. doi: 10.1126/science.108.2804.329. [DOI] [PubMed] [Google Scholar]
- Rasmussen SGF, DeVree BT, Zou Y, et al. Crystal structure of the β2 adrenergic receptor-Gs protein complex. Nature. 2011;477:549–555. doi: 10.1038/nature10361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raymond JR, Mukhin YV, Gettys TW, Garnovskaya MN. The recombinant 5-HT1A receptor: G protein coupling and signalling pathways. Br J Pharmacol. 1999;127:1751–1764. doi: 10.1038/sj.bjp.0702723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosendorff A, Ebersole BJ, Sealfon SC. Conserved helix 7 tyrosine functions as an activation relay in the serotonin 5HT2C receptor. Brain Res Mol Brain Res. 2000;84:90–96. doi: 10.1016/s0169-328x(00)00227-8. [DOI] [PubMed] [Google Scholar]
- Roth BL, Sheffler DJ, Kroeze WK. Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophrenia. Nat Rev Drug Discov. 2004;3:353–359. doi: 10.1038/nrd1346. [DOI] [PubMed] [Google Scholar]
- Safdari HA, Pandey S, Shukla AK, Dutta S. Illuminating GPCR signaling by cryo-EM. Trends Cell Biol. 2018;28:591–594. doi: 10.1016/j.tcb.2018.06.002. [DOI] [PubMed] [Google Scholar]
- Sarkar P, Chattopadhyay A. Cholesterol interaction motifs in G protein-coupled receptors: slippery hot spots? Wiley Interdiscip Rev Syst Biol Med. 2020;12:e1481. doi: 10.1002/wsbm.1481. [DOI] [PubMed] [Google Scholar]
- Sarkar P, Kumar GA, Pal S, Chattopadhyay A (2018) Biophysics of serotonin and the serotonin1A receptor: fluorescence and dynamics. In: Pilowsky P (ed) serotonin: the mediator that spans evolution. Elsevier, pp 3–22. 10.1016/B978-0-12-800050-2.00001-2
- Sengupta D, Chattopadhyay A. Identification of cholesterol binding sites in the serotonin1A receptor. J Phys Chem B. 2012;116:12991–12996. doi: 10.1021/jp309888u. [DOI] [PubMed] [Google Scholar]
- Sengupta D, Chattopadhyay A. Molecular dynamics simulations of GPCR-cholesterol interaction: an emerging paradigm. Biochim Biophys Acta. 2015;1848:1775–1782. doi: 10.1016/j.bbamem.2015.03.018. [DOI] [PubMed] [Google Scholar]
- Sengupta D, Kumar GA, Chattopadhyay A. Interaction of membrane cholesterol with GPCRs: implications in receptor oligomerization. In: Giovanni G, Herrick-Davis K, Milligan G, editors. G protein-coupled receptor dimers. Heidelberg: Springer; 2017. pp. 415–429. [Google Scholar]
- Sengupta D, Prasanna X, Mohole M, Chattopadhyay A. Exploring GPCR-lipid interactions by molecular dynamics simulations: excitements, challenges and the way forward. J Phys Chem B. 2018;122:5727–5737. doi: 10.1021/acs.jpcb.8b01657. [DOI] [PubMed] [Google Scholar]
- Shan J, Khelashvili G, Mondal S, Mehler EL, Weinstein H. Ligand-dependent conformations and dynamics of the serotonin 5-HT2A receptor determine its activation and membrane-driven oligomerization properties. PLoS Comput Biol. 2012;8:e1002473. doi: 10.1371/journal.pcbi.1002473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shapiro DA, Kristiansen K, Weiner DM, Kroeze WK, Roth BL. Evidence for a model of agonist-induced activation of 5-hydroxytryptamine2A serotonin receptors that involves the disruption of a strong ionic interaction between helices 3 and 6. J Biol Chem. 2002;277:11441–11449. doi: 10.1074/jbc.M111675200. [DOI] [PubMed] [Google Scholar]
- Sjögren B, Svenningsson P. Caveolin-1 affects serotonin binding and cell surface levels of human 5-HT7(a) receptors. FEBS Lett. 2007;581:5115–5121. doi: 10.1016/j.febslet.2007.09.059. [DOI] [PubMed] [Google Scholar]
- Sjögren B, Svenningsson P. Depletion of the lipid raft constituents, sphingomyelin and ganglioside, decreases serotonin binding at human 5-HT7(a) receptors in HeLa cells. Acta Physiol. 2007;190:47–53. doi: 10.1111/j.1365-201X.2007.0168.x. [DOI] [PubMed] [Google Scholar]
- Sjögren B, Hamblin MW, Svenningsson P. Cholesterol depletion reduces serotonin binding and signaling via human 5-HT7(a) receptors. Eur J Pharmacol. 2006;552:1–10. doi: 10.1016/j.ejphar.2006.08.069. [DOI] [PubMed] [Google Scholar]
- Sjöstrand D, Diamanti R, Lundgren CAK, et al. A rapid expression and purification condition screening protocol for membrane protein structural biology. Protein Sci. 2017;26:1653–1666. doi: 10.1002/pro.3196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skretas G, Georgiou G. Engineering G protein-coupled receptor expression in bacteria. Proc Natl Acad Sci USA. 2008;105:14747–14748. doi: 10.1073/pnas.0807741105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith SM. Strategies for the purification of membrane proteins. Methods Mol Biol. 2017;1485:389–400. doi: 10.1007/978-1-4939-6412-3. [DOI] [PubMed] [Google Scholar]
- Sodhi MSK, Sanders-Bush E. Serotonin and brain development. Int Rev Neurobiol. 2004;59:111–174. doi: 10.1016/S0074-7742(04)59006-2. [DOI] [PubMed] [Google Scholar]
- Stiedl O, Pappa E, Konradsson-Geuken Å, Ögren SO. The role of the serotonin receptor subtypes 5-HT1A and 5-HT7 and its interaction in emotional learning and memory. Front Pharmacol. 2015;6:162. doi: 10.3389/fphar.2015.00162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tatarczyńska E, Kłodzińska A, Stachowicz K, Chojnacka-Wójcik E. Effects of a selective 5-HT1B receptor agonist and antagonists in animal models of anxiety and depression. Behav Pharmacol. 2004;15:523–534. doi: 10.1097/00008877-200412000-00001. [DOI] [PubMed] [Google Scholar]
- Tate CG. A crystal clear solution for determining G-protein-coupled receptor structures. Trends Biochem Sci. 2012;37:343–352. doi: 10.1016/j.tibs.2012.06.003. [DOI] [PubMed] [Google Scholar]
- The UniProt Consortium (2019) UniProt: a worldwide hub of protein knowledge. Nucleic Acids Res 47:D506–D515. 10.1093/nar/gky1049 [DOI] [PMC free article] [PubMed]
- Theriot J, Wermuth HR, Ashurst JV (2020) Antiemetic serotonin-5-HT3 receptor blockers. In: StatPearls. StatPearls Publishing. [PubMed]
- Torrens-Fontanals M, Stepniewski TM, Aranda-García D, Morales-Pastor A, Medel-Lacruz B, Selent J. How do molecular dynamics data complement static structural data of GPCRs. Int J Mol Sci. 2020;21:5933. doi: 10.3390/ijms21165933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Twarog BM, Page IH. Serotonin content of some mammalian tissues and urine and a method for its determination. Am J Phys. 1953;175:157–161. doi: 10.1152/ajplegacy.1953.175.1.157. [DOI] [PubMed] [Google Scholar]
- Udenfriend S, Bogdanski DF, Weissbach H. Fluorescence characteristics of 5-hydroxytryptamine (serotonin) Science. 1955;122:972–973. doi: 10.1126/science.122.3177.972. [DOI] [PubMed] [Google Scholar]
- Unal H, Jagannathan R, Bhat MB, Karnik SS. Ligand-specific conformation of extracellular loop-2 in the angiotensin II type 1 receptor. J Biol Chem. 2010;285:16341–16350. doi: 10.1074/jbc.M109.094870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Underwood MD, Kassir SA, Bakalian MJ, et al. Serotonin receptors and suicide, major depression, alcohol use disorder and reported early life adversity. Transl Psychiatry. 2018;8:279. doi: 10.1038/s41398-018-0309-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Westhuizen ET, Valant C, Sexton PM, Christopoulos A. Endogenous allosteric modulators of G protein-coupled receptors. J Pharmacol Exp Ther. 2015;353:246–260. doi: 10.1124/jpet.114.221606. [DOI] [PubMed] [Google Scholar]
- Varrault A, Le Nguyen D, McClue S, et al. 5-Hydroxytryptamine1A receptor synthetic peptides. Mechanisms of adenylyl cyclase inhibition. J Biol Chem. 1994;269:16720–16725. doi: 10.1016/S0021-9258(19)89450-4. [DOI] [PubMed] [Google Scholar]
- Wacker D, Wang C, Katritch V, et al. Structural features for functional selectivity at serotonin receptors. Science. 2013;340:615–619. doi: 10.1126/science.1232808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wacker D, Wang S, McCorvy JD, et al. Crystal structure of an LSD-bound human serotonin receptor. Cell. 2017;168:377–389.e12. doi: 10.1016/j.cell.2016.12.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wacker D, Stevens RC, Roth BL. How ligands illuminate GPCR molecular pharmacology. Cell. 2017;170:414–427. doi: 10.1016/j.cell.2017.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, Jiang Y, Ma J, et al. Structural basis for molecular recognition at serotonin receptors. Science. 2013;340:610–614. doi: 10.1126/science.1232807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wheatley M, Wootten D, Conner MT, et al. Lifting the lid on GPCRs: the role of extracellular loops. Br J Pharmacol. 2012;165:1688–1703. doi: 10.1111/j.1476-5381.2011.01629.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitaker-Azmitia PM. The discovery of serotonin and its role in neuroscience. Neuropsychopharmacol. 1999;21:2–8. doi: 10.1016/S0893-133X(99)00031-7. [DOI] [PubMed] [Google Scholar]
- Wingler LM, Lefkowitz RJ. Conformational basis of G protein-coupled receptor signaling versatility. Trends Cell Biol. 2020;30:736–747. doi: 10.1016/j.tcb.2020.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiseman DN, Otchere A, Patel JH, et al. Expression and purification of recombinant G protein-coupled receptors: a review. Protein Expr Purif. 2020;167:105524. doi: 10.1016/j.pep.2019.105524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wootten D, Christopoulos A, Sexton PM. Emerging paradigms in GPCR allostery: implications for drug discovery. Nat Rev Drug Discov. 2013;12:630–644. doi: 10.1038/nrd4052. [DOI] [PubMed] [Google Scholar]
- Xia X, Ding M, Xuan J-F, et al. Polymorphisms in the human serotonin receptor 1B (HTR1B) gene are associated with schizophrenia: a case control study. BMC Psychiatry. 2018;18:303. doi: 10.1186/s12888-018-1849-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin W, Zhou XE, Yang D, et al. Crystal structure of the human 5-HT1B serotonin receptor bound to an inverse agonist. Cell Discov. 2018;4:12. doi: 10.1038/s41421-018-0009-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yohn CN, Gergues MM, Samuels BA. The role of 5-HT receptors in depression. Mol Brain. 2017;10:28. doi: 10.1186/s13041-017-0306-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan S, Filipek S, Palczewski K, Vogel H. Activation of G-protein-coupled receptors correlates with the formation of a continuous internal water pathway. Nat Commun. 2014;5:4733. doi: 10.1038/ncomms5733. [DOI] [PubMed] [Google Scholar]
- Zarkadas E, Zhang H, Cai W et al (2020) The binding of palonosetron and other antiemetic drugs to the serotonin 5-HT3 receptor. Structure. 10.1016/j.str.2020.07.004 [DOI] [PubMed]
- Zhang X, Stevens RC, Xu F. The importance of ligands for G protein-coupled receptor stability. Trends Biochem Sci. 2015;40:79–87. doi: 10.1016/j.tibs.2014.12.005. [DOI] [PubMed] [Google Scholar]
- Zhang D, Zhao Q, Wu B. Structural studies of G protein-coupled receptors. Mol Cells. 2015;38:836–842. doi: 10.14348/molcells.2015.0263. [DOI] [PMC free article] [PubMed] [Google Scholar]












