Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 1.
Published in final edited form as: Curr Opin Biotechnol. 2020 Jul 10;64:210–217. doi: 10.1016/j.copbio.2020.06.004

Advances in G Protein-Coupled Receptor High-throughput Screening

Emily A Yasi 1, Nicholas S Kruyer 2, Pamela Peralta-Yahya 1,2
PMCID: PMC7483569  NIHMSID: NIHMS1608071  PMID: 32653805

Abstract

G protein-coupled receptors (GPCRs) detect compounds on the cell surface and are the starting point of a number of medically relevant signaling cascades. Indeed, over 30% of FDA approved drugs target GPCRs, making them a primary target for drug discovery. Computational and experimental high-throughput screening (HTS) approaches of clinically relevant GPCRs are a first-line drug discovery effort in biomedical research. In this opinion, we review recent advances in GPCR HTS. We focus primarily on cell-based assays, and highlight recent advances in in vitro assays using purified receptors, and computational approaches for GPCR HTS. To date, GPCR HTS has led to the identification of new and repurposing of existing drugs, and the deorphanization of GPCRs with unknown ligands. As automation equipment becomes more common, GPCR HTS will move beyond a drug discovery tool to a key technology to probe basic biological processes that will have an outsized impact on personalized medicine.

Introduction

G protein-coupled receptors (GPCRs) are the target of more than 30% of FDA approved drugs, placing them as the premier protein family for approved pharmaceuticals [1]. Only ~130 of the 360 non-sensory GPCRs are targeted by drugs [2], leaving > 200 GPCRs as potential therapeutic targets. Computational and experimental high-throughput screening (HTS) of clinically relevant GPCRs with compounds and peptides is a first-line drug discovery tool [3]. One of the earliest examples of GPCR HTS for drug discovery is the screening of > 500,000 compounds for agonists of chemokine receptor 5 using a radioligand binding assay to arrive at maravirnoc, now an antiviral HIV treatment [4].

Today, experimental GPCR HTS approaches primarily rely on cells to report on changes in the intracellular concentration of secondary messengers upon GPCR ligand binding. These changes result from the activation of one or more Gα subunit families upon GPCR stimulation. Widely used assays detect GPCR-dependent activation of Gαs, Gαi and Gαq. Activation of Gαs upregulates cAMP production, activation of Gαi inhibits cAMP production and activation of Gαq ultimately results in calcium (Ca2+) accumulation. The ability of most GPCRs to recruit β-arrestin to initiate receptor desensitization has also been leveraged to report on GPCR ligand binding. For GPCRs with a known Gα coupling subunit family, the correct cell-based assay can be chosen. However, for the ~100 non-sensory GPCRs that lack known ligands and for which the Gα subunits they couple to are unknown—i.e. orphan GPCRs [5]—multiple cell-based assays that activate different signaling pathways must be performed for ligand identification. Finally, GPCR HTS is accessible to laboratories beyond those with deep cell-based assay expertise. Radioligand binding assays as well as functional assays for Gαi, Gαs and Gαq-coupled receptors for over 175 GPCR targets can be purchased for screening or the screening can itself be outsourced (e.g. Eurofins DiscoverX, ThermoFisher, Promega).

GPCR cell-based assays require heterologous GPCR expression in an engineered cell to report on ligand binding, which has its own set of technical challenges. First, functional expression of human GPCRs outside their endogenous tissue sometimes requires addition of expression tags, which may affect ligand binding. Next, signal transduction from the GPCR to the synthetic machinery in the engineered cell may be inefficient; for example, the engineered cell may express the correct Gα subunit but the incorrect isoform. Third, most GPCR cell-based assays take place in mammalian cells that express endogenous GPCRs. For example, HEK293, a widely used cell line for GPCR HTS, expresses 75 endogenous GPCRs [6]. Given that all GPCRs couple via the same four Gα protein families, leading to concentration changes of only a handful of secondary messengers, the potential for cross talk is significant. That is, the measured reporter output may be due to activation of an endogenous GPCR rather than the desired heterologous one. Finally, as is the case with all cell-based assays, compounds may result in GPCR independent reporter output changes leading to false negatives and false positives. For example, compounds may inhibit the reporter activity or emit fluorescence at the same wavelength as the reporter [7]. Therefore, GPCR HTS should be followed by lower-throughput biochemical assays (e.g. pull down assay) to confirm hits before validation in the GPCR endogenous tissue.

Here, we review advances in GPCR HTS over the last two years. We primarily focus on cell-based assays and we classify them by the intracellular proteins to which they couple (Figure 1). We also highlight key advances in in vitro GPCR HTS as well as computational approaches that reduce the number of compounds to be tested experimentally. Taken together, it is an exciting time for GPCR drug discovery and deorphanization.

Figure 1. High-throughput GPCR cell-based assays.

Figure 1.

A. GPCR activation of Gαs upregulates adenylate cyclase (AC) resulting in increased cAMP levels. cAMP activates protein kinase A, which phosphorylates the transcription factor CREB ultimately resulting in reporter gene expression. B. GPCR activation of Gαi downregulates AC resulting in a decrease of cAMP levels. C. GPCR activation of Gαq upregulates phospholipase C (PLC), which breaks down PIP2 (phosphatidylinositol biphosphate) into IP3 (inositol triphosphate) and DAG (diacylglycerol). IP3 goes on to activate Ca2+ channels in the endoplasmic reticulum increasing the Ca2+ concentration in the cytosol, which is detected via chemical dyes or protein sensors. D. In β-arrestin recruitment-based assays, GPCR activation recruits β-arrestin linked to a protease (yellow) that cleaves the transcription factor (TF) linked to the GPCR via a protease cleavable linker (yellow). Transcription factor release ultimately results in reporter gene expression. E. In Saccharomyces cerevisiae, human GPCRs can couple to yeast or human/yeast Gα chimeras and via Gβ activate the yeast mating pathway ultimately leading to expression of a reporter gene. Gold circles: agonist.

1. Cell-based GPCR high-throughput assays

1.1. Secondary messenger- based assays: cyclic adenosine monophosphate (cAMP)

Upon GPCR ligand binding, Gαs upregulates adenylate cyclase, which in turn increases the levels of cAMP. Activated by cAMP, protein kinase A phosphorylates the CREB transcription factor, leading to transcription activation of a reporter gene downstream of a minimal promoter containing CRE binding sites (Figure 1A). Using β-galactosidase as the reporter, Chen et al. applied this assay to screen the β2-adrenergic receptor (β2AR) against ~7,000 chemicals in neuroepithelial cells and confirmed that β2AR is activated by higenamine, a treatment for heart failure [8]. Importantly, depending on the ligand, some GPCRs couple to different Gα subunit families leading to biased signaling [9]. The β2AR is known to activate Gαs, Gαi or β-arrestin [10], thus a β2AR ligand identified via cAMP accumulation preferentially activates Gαs signaling. Additional assays that couple β2AR via Gαi or β-arrestin should be used to identify β2AR ligands that preferentially signal via those pathways. Alternative cAMP reporter systems include an immunoassay to directly measure cAMP levels (Eurofins) and an assay that measures the decrease in ATP levels as protein kinase A consumes ATP to phosphorylate transcription factors (Promega).

Having a single reporter output (e.g. luminescence, fluorescence) requires spatial separation of cells expressing different GPCRs during compound screening. Testing a single GPCR against a single chemical allows identification of both high and low affinity binders. Nevertheless, the screening throughput is limited to that achievable using 96- or 384-well plates in a couple of days. By linking activation of each GPCR to a different RNA barcode reporter, cells expressing different GPCRs can be pooled together and tested against a single chemical en masse, thus significantly increasing the throughput of the screen (Figure 2). This strategy was used to deorphanize G protein-coupled olfactory receptors (ORs), which couple to Gα olfactory (Gαolf) leading to cAMP accumulation. Specifically, Jones et al. screened ~39 murine ORs against 181 odorants and successfully deorphanized 15 ORs [11●●]. Only if a specific OR was activated, the assigned barcode was transcribed and quantified via RNA sequencing. Of note, the many GPCRs to one compound screening strategy effectively has all GPCRs competing for the same ligand, leading to the isolation of the highest affinity binders. Although for GPCR deorphanization this may be a desired outcome, for pharmaceutical applications low affinity binders may be a better starting scaffold in terms of pharmacological profile or more amenable for a medicinal chemistry campaign.

Figure 2. Cell-based screening approaches.

Figure 2.

A. One-to-one GPCR ligand screening. Cell lines expressing different GPCRs are spatially separated in different wells and their ligand binding ability assessed independently. This approach is used when GPCRs are linked to the same reporter, e.g. fluorescence or luminescence reporters. B. Many-to-one GPCR ligand screening. Each GPCR is linked to a different DNA barcode. Cell lines expressing different GPCRs are pooled and en masse screened against a single ligand. Activation of each GPCR leads to different transcription levels of the barcode that can be quantified using RNA sequencing.

GPCRs can bind both peptides and small molecules. Typically, peptide libraries are more expensive to synthesize than small molecule libraries, and thus likely less often studied. Instead of relying on a large library of purified peptides, Yaginuma et al. engineered Saccharomyces cerevisiae to secrete mutants of the known glucagon-like peptide-1 receptor (hGLP1R) agonist, Ex4. The engineered yeast were encapsulated together with a mammalian reporter cell line expressing hGLP1R that secreted the β-galactosidase reporter. Droplets containing activated hGLP1R were highly fluorescent, making them easily identified and isolated via fluorescence microscopy. This assay identified one novel peptide for hGLP1R out of the 400 peptides screened [12●●]. With improved throughput, specifically in the droplet screening, ligand screening could be increased to explore the entire mutational space for a given peptide.

Opposite of Gαs, the activation of Gαi results in a decrease of cAMP. To measure this, an adenylate cyclase activator, such as forskolin, is first added to increase cAMP levels before GPCR-dependent Gαi activation, allowing an observable decline in cAMP-dependent signal (Figure 1B). Using this strategy, Myers et al. screened the apelin receptor, a drug target for heart failure, against a Bristol Myers Squibb proprietary library and identified aryl hydroxy pyrimidinones as agonists for this receptor [13].

1.2. Calcium-based assays

Activation of Gαq ultimately results in an increase in intracellular calcium levels (Figure 1C). Calcium dyes, such as Fluo-4, can be used to report on calcium fluctuation. Calcium dye fluorescence is often read using a Fluorescent Imaging Plate Reader (FLIPR®, Molecular Devices) assay. Smith et al. used a FLIPR® assay to screen a 360,000 member small molecule library to identify 25 agonists, four positive allosteric modulators and 41 antagonists of the muscarinic acetylcholine receptor M4 [14]. The main drawbacks of dye-based assays are their cost ($25/96 well plate) and additional assay time (~1 hour), which limit their HTS applications. Genetically encoded calcium biosensors (GECIs) are a less expensive alternative to calcium dyes. One of the most widely used GECIs is GCaMP, a protein fusion of calmodulin, green fluorescent protein (GFP) and the calmodulin substrate M13 peptide. Binding of Ca2+ displaces M13 resulting in GFP fluorescence [15].

In 2018, Mella et al. introduced genetically encoded sensors for Ca2+ and cAMP that can be used simultaneously, thus enabling GPCR activation of multiple Gα subunit families in the same experiment [16●]. In these sensors, a plasma membrane localized fluorescent protein is linked to a peptide (calmodulin for Ca2+ and thyroid A-kinase anchoring protein for cAMP) that undergoes a conformational change upon chemical binding resulting in increased fluorescence. Mella et al. screened the endothelin B receptor against 1,200 chemicals, and identified two new ligands that only signaled via Gαq (Ca2+ accumulation) and one ligand that signals via both Gαs and Gαq (cAMP and Ca2+ accumulation).

Before raising calcium levels, Gαq activation increases in DAG and IP3 concentrations, with IP3 ultimately metabolizing to inositol monophosphate (IP1). Although an assay that detects IP1 [17], and several fluorescent-based DAG sensors exist [18], they have not been used for GPCR HTS.

1.3. β-arrestin recruitment-based assays

Activated GPCRs eventually undergo desensitization via recruitment of β-arrestin, a process that has been exploited to report on GPCR activation. A GPCR fused to a transcription factor via a protease cleavage site recruits β-arrestin linked to a protease, resulting in cleavage of the transcription factor and expression of the reporter gene [19] (Figure 1D). In 2015, this assay was expanded to > 300 GPCRs by appending the C-terminus of the vasopressin receptor to promote efficient β-arrestin recruitment [20]. Fan et al. used this expanded assay to screen 320 GPCRs in parallel against three chemicals and identified selective agonists for the D2 dopamine receptor, a key neuropsychiatric drug target [21]. Li et al. improved the reliability of the β-arrestin recruitment assay by integrating the reporter gene and β-arrestin into the genome [22]. This assay was used to screen the D2 dopamine receptor against 1,000 natural products and identified wilfortrine as an agonist for the first time [22].

A new application of GPCR HTS is the screening of human microbial metabolomes to identify microbial metabolites that activate GPCRs. Colosimo et al. screened the fractionated metabolomes of seven bacteria that make up the simplified human gut microbiome against 241 GPCRs and identified eight GPCRs that showed reproducible β-arrestin recruitment and passed their hit threshold level. A following bioassay-guided fractionation sought to determine the activating compound for hydroxycarboxylic acid, neurotransmitter and histamine receptors. Testing of the fractions of interest with the GPCR-based assay resulted in the identification of nine microbial compounds activating 15 GPCRs, including five orphan GPCRs [23●]. In a different study, Chen et al. focused on 144 bacteria isolated from 11 inflammatory bowel disease patients, and screened the monoculture supernatants against 314 human GPCRs. This screen revealed a high level of interaction between commensal bacteria and the aminergic receptors in the human gut. Specifically, follow up studies in germ-free mice showed that histamine and phenethylamine produced by Morganella morganii play important roles in colonic motility and efficacy of anti-depressants, respectively. In addition, this study also found potential hits for 17 orphan GPCRs, with further investigation identifying L-phenylalanine as an agonist for GPR56/AGRG1 and GPR97/AGRG3, indicating a potential connection between microbiota metabolites and digestion or satiety [24●●].

Reporters used in β-arrestin recruitment-based assays include luciferase, GFP, split luciferase, β-lactamase and RNA barcodes. A luciferase signal can be read rapidly in plate readers; however, it requires mammalian cell lysis, thus limiting the reporter to end-point detection [25]. In comparison, GFP has a long maturation time and requires a high-throughput cell imaging system for signal acquisition when using mammalian cells. Split luciferase enables continuous monitoring of GPCR activation [26]; however, it has not yet been used for GPCR HTS applications. In a split TEV β-arrestin recruitment-based assay, Galinski et al. used RNA barcodes to assess 19 aminergic receptors against five known ligands [27].

1.4. Non-mammalian cell-based assays

To our knowledge, the only application of GPCR HTS outside of mammalian cells has been in the yeast S. cerevisiae. Indeed, S. cerevisiae is an ideal host to screen human GPCRs. In yeast, human GPCRs can couple to the yeast mating pathway directly via an endogenous Gαs, GPA1, or a GPA1/human Gα chimera [28] leading to activation of growth, fluorescent or luminescent reporters (Figure 1E). The ability to use growth reporters in yeast enables the screening of millions of GPCR variants against one chemical en masse, which has been exploited to engineer GPCRs exclusively activated by inert drugs (DREADDs), a widely used chemogenetic tool in neuroscience [29]. Yeast show advantage over mammalian cells due to the faster growth rate (3 hrs vs 12 hrs for mammalian cells) and its robustness, as it can be stored at 4°C for a month without the need for passage. Certainly, the use of yeast for human GPCR HTS also has some drawbacks, such as the lack of proper GPCR glycosylation or phosphorylation, which may result in the non-functional expression of GPCRs. As with any GPCR HTS, the ligand must be validated in the GPCR endogenous tissue for activity. Taken together, yeast has the potential to provide a more robust and faster platform for human GPCR screening.

In 2019, Yasi et al. screened the human serotonin 4 receptor (5-HTR4), the pharmacological target for irritable bowel syndrome with constipation, against 1,206 compounds and ultimately identified three 5-HTR4 agonists leading to an increase in human colon cell motility [30●●]. The use of a luciferase reporter was pivotal as a GFP reporter lead only to a three-fold increase in signal after activation and a limit of detection in the μM range [31]. Swapping GFP for luciferase improved the assay dynamic range to more than 30-fold and lowered the limit of detection to the low nM range. Further, the use of luciferase reduced the total assay time from four to 2.5 hours, and enabled a screening throughput of one compound per second. Of note, 5-HTR4 coupled to GPA1; however, this may not be true of other GPCRs and thus yeast/human Gα chimeras may need to be used.

Olfactory receptors have also been shown to couple to the yeast mating pathway [32, 33]. Yasi et al. screened seven ORs against 57 chemicals resulting in the deorphanization of two receptors [34]. Sarria et al. used the olfactory receptor OR1G1 to detect medium-chain fatty acids produced by Escherichia coli directly in the culture broth, opening the doors to use GPCR-based assays for the high-throughput screening of microbially produced compounds [35]. OR-based assays in yeast are currently linked to GFP expression that requires a flow cytometer for read out, limiting the throughput to one compound per minute.

2. In vitro GPCR high-throughput assays

The low stability of purified GPCRs, makes their screening outside cells or membrane systems challenging, and GPCRs engineered for stability [36] are used in in vitro GPCR assays. Among the most widely used in vitro GPCR HTS assays are fluorescence polarization and affinity mass spectrometry (MS). Fluorescence polarization can identify GPCRs bound to fluorescently labelled ligands based on their molecular tumbling speed (Figure 3A). Heine et al. screened neurotensin receptor type 1 (NTS1) against 1,272 chemicals by displacing a fluorescently labeled NTS1 peptide ligand [37]. Eight of the 15 hits were confirmed via surface plasmon resonance. As expected, the availability of fluorescently labelled substrates is a key limitation of this approach. In affinity mass spectrometry, a GPCR is incubated with hundreds of compounds simultaneously. While unbound compounds are washed off, the GPCR ligands are eluted using protein denaturation and analyzed via mass spectrometry [38] (Figure 3B). Lu et al. expanded the number of compounds that can be screened via affinity MS from hundreds to tens of thousands by introducing an enrichment step of eluting the GPCR ligands and incubating them again with a purified receptor [39●]. A thermostable adenosine receptor was screened either as a purified receptor or embedded in a membrane against ~20,000 chemicals, and the assay identified 70% and 50% of ligands, respectively, previously identified using non-enrichment affinity MS. Although some compounds are not identified via the enrichment assay, the throughput is highly increased. Of note, thermostabilized GPCRs could affect ligand binding.

Figure 3. High-throughput in vitro GPCR screening.

Figure 3.

A. In fluorescence polarization, the fluorescently labelled ligand has a fast tumbling rate leading to a decrease in fluorescence. Upon ligand binding, the tumbling speed of the ligand slows down increasing its fluorescence. B. In affinity mass spectrometry, a GPCR is incubated with hundreds of ligands simultaneously. Ligands bound to the GPCR are eluted and analyzed via liquid chromatography/mass spectrometry. The number of the ligands can be narrowed down by re-incubating eluted ligands with fresh GPCR.

3. In silico GPCR-based high-throughput assays

Computational methods can reduce the number of potential GPCR ligands (biased and non-biased) and allosteric regulators needed to test experimentally. Foster et al. used computational approaches rooted in GPCR-peptide ligand coevolution to predict 120 potential GPCR peptide ligands, and 21 GPCRs likely to respond to these peptides [40●]. As GPCRs can couple to multiple signaling pathways and their response may appear different depending on the pathway they activate, the GPCR ligand interactions were assessed using three different assays: mass spectrometry distribution, receptor internalization and β-arrestin recruitment. Ultimately, this work led to the deorphanization of five GPCRs.

Another in silico approach, computational docking, takes advantage of ultra-large chemical libraries, such as ZINC and ChEMBL, expanding the chemical diversity of screened potential GPCR ligands from thousands to hundreds of millions [41]. Virtual docking has been successfully applied to over a dozen classes of GPCRs [41]. Lyu et al. virtually screened 138 million chemicals against the D4 dopamine receptor, the pharmacological target for schizophrenia [42]. The top 549 chemicals predicted by virtual docking were synthesized with a radiolabel, and, using a radioactivity binding assay, 22% of the hits were validated. In another example, Stein et al. virtually screened over 150 million chemicals against melatonin receptor 1 (MT1) [43●●]. After manual inspection the top 40 chemicals were synthesized, and 15 of them showed activity for either melatonin receptor 1 or 2 (61% similar) using a cAMP-dependent or β-arrestin-dependent assay. As computational docking significantly reduces the number of compounds to be tested, lower-throughput approaches are relevant. Notably, virtual docking is most effective when a crystal structure for the desired GPCR is available [44]. This currently limits the virtual docking approach, as 85% of non-olfactory GPCRs do not have a structure.

4. Future Outlook

The complexity of GPCR signaling, activating different pathways sometimes simultaneously, leads to the lack of a universal GPCR cell-based assay. Therefore, before embarking on GPCR HTS, there needs to be a deep understanding of the GPCR signaling pathway to be probed in order to choose the appropriate cell-based assay and understand its limitations. Such an understanding is not essential for in vitro assays, as GPCRs are evaluated in purified form or as part of membranes. Independent of the method used for GPCR ligand identification, the ligand needs to be confirmed using biochemical binding assays and ultimately validated in the GPCR endogenous tissue.

As automation equipment becomes more widely available in academic laboratories, GPCR HTS will be used to probe basic biological processes. For example, different GPCR isoforms tend to be expressed in different cell types. By applying GPCR HTS, we can begin to understand the different cell types that are activated by different ligands. Further, different patient populations tend to have different GPCR alleles, and GPCR HTS could help elucidate the extent to which different GPCR alleles are activated. With the potential of having each patient’s genome sequenced in the near future, we can envision GPCR HTS as a common occurrence to generate lifesaving therapies.

Bioinformatics and virtual docking rapidly scale down the number of ligands to be evaluated experimentally and can rely on lower-throughput biochemical binding assays. However, the necessity for the specific GPCR crystal structure for docking currently limits this approach to well-studied GPCRs. Given that there are only 64 unique crystal structures known (https://gpcrdb.org/structure/statistics), virtual docking approaches are not currently suitable to identify ligands for the majority of GPCRs. In the future, machine learning may be able to address this problem by combining bioinformatics, virtual docking on GPCR models and limited experimental GPCR HTS data to generate ligand predictions.

Acknowledgements

This work was funded by an NIH MIRA Award (R35GM124871) to P.P-Y.

Footnotes

The authors declare no competing financial interests.

5. References

  • 1.Hauser AS, Attwood MM, Rask-Andersen M, Schioth HB, Gloriam DE: Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov 2017, 16:829–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sriram K, Insel PA: GPCRs as targets for approved drugs: how many targets and how many drugs? Mol Pharmacol 2018, 93:251–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Macarron R, Banks MN, Bojanic D, Burns DJ, Cirovic DA, Garyantes T, Green DVS, Hertzberg RP, Janzen WP, Paslay JW et al. : Impact of high-throughput screening in biomedical research. Nat Rev Drug Discov 2011, 10:188–195. [DOI] [PubMed] [Google Scholar]
  • 4.Dorr P, Westby M, Dobbs S, Griffin P, Irvine B, Macartney M, Mori J, Rickett G, Smith-Burchnell C, Napier C et al. : Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity. Antimicrob Agents Chemother 2005, 49:4721–4732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Laschet C, Dupuis N, Hanson J: The G protein-coupled receptors deorphanization landscape. Biochem Pharmacol 2018, 153:62–74. [DOI] [PubMed] [Google Scholar]
  • 6.Atwood BK, Lopez J, Wager-Miller J, Mackie K, Straiker A: Expression of G protein-coupled receptors and related proteins in HEK293, AtT20, BV2, and N18 cell lines as revealed by microarray analysis. BMC Genom 2011, 12:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Aldrich C, Bertozzi C, Georg GI, Kiessling L, Lindsley C, Liotta D, Merz KM Jr., Schepartz A, Wang S: The ecstasy and agony of assay interference compounds. ACS Cent Sci 2017, 3:143–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chen Y, Guo B, Zhang H, Hu L, Wang J: Higenamine, a dual agonist for beta 1- and beta 2-adrenergic receptors identified by screening a traditional chinese medicine library. Planta Med 2019, 85:738–744. [DOI] [PubMed] [Google Scholar]
  • 9.Smith JS, Lefkowitz RJ, Rajagopal S: Biased signalling: from simple switches to allosteric microprocessors. Nat Rev Drug Discov 2018, 17:243–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Jean-Charles PY, Kaur S, Shenoy SK: GPCRs signaling shrough beta-arrestin-dependent mechanisms. J Cardiovasc Pharmacol 2017, 70:142–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Jones EM, Jajoo R, Cancilla D, Lubock NB, Wang J, Satyadi M, Cheung R, de March C, Bloom JS, Matsunami H, Kosuri S: A scalable, multiplexed assay for decoding GPCR-ligand interactions with RNA sequencing. Cell Syst 2019, 8:254–260.●● A GPCR assay that allows for pooling of multiple cells expressing different GPCRs against a single chemical by linking each GPCR to a different RNA barcode reporter.
  • 12.Yaginuma K, Aoki W, Miura N, Ohtani Y, Aburaya S, Kogawa M, Nishikawa Y, Hosokawa M, Takeyama H, Ueda M: High-throughput identification of peptide agonists against GPCRs by co-culture of mammalian reporter cells and peptide-secreting yeast cells using droplet microfluidics. Sci Rep 2019, 9:10920.●● Using peptide secreting yeast, screened a 400 member peptide library against the human glucagon-like peptide-1 receptor. Combination yeast and mammalian assay shows promise for reducing the cost of peptide library screening.
  • 13.Myers MC, Bilder DM, Cavallaro CL, Chao HL, Su S, Burford NT, Nayeem A, Wang T, Yan M, Langish RA et al. : Discovery and SAR of aryl hydroxy pyrimidinones as potent small molecule agonists of the GPCR APJ. Bioorg Med Chem Lett 2020, 30:126955. [DOI] [PubMed] [Google Scholar]
  • 14.Smith E, Chase P, Niswender CM, Utley TJ, Sheffler DJ, Noetzel MJ, Lamsal A, Wood MR, Conn PJ, Lindsley CW et al. : Application of parallel multiparametric cell-based FLIPR detection assays for the identification of modulators of the muscarinic acetylcholine receptor 4 (M4). J Biomol Screen 2015, 20:858–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wu N, Nishioka WK, Derecki NC, Maher MP: High-throughput-compatible assays using a genetically-encoded calcium indicator. Sci Rep 2019, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mella RM, Kortazar D, Roura-Ferrer M, Salado C, Valcarcel M, Castilla A, Villace P: Nomad biosensors: A new multiplexed technology for the screening of GPCR ligands. SLAS Technol 2018, 23:207–216.● Simultanoues use of a calcium and cAMP sensor to detect both Gαs and Gαq activation and elucidate GPCR biased signaling in a single assay.
  • 17.O’Halloran F, Bruen C, McGrath B, Schellekens H, Murray B, Cryan JF, Kelly AL, McSweeney PLH, Giblin L: A casein hydrolysate increases GLP-1 secretion and reduces food intake. Food Chem 2018, 252:303–310. [DOI] [PubMed] [Google Scholar]
  • 18.Tewson PH, Martinka S, Shaner NC, Hughes T, Quinn AM: New DAG and cAMP sensors optimized for live-cell assays in automated laboratories. J Biomol Screen 2016, 21:298–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Barnea G, Strapps W, Herrada G, Berman Y, Ong J, Kloss B, Axel R, Lee KJ: The genetic design of signaling cascades to record receptor activation. Proc Natl Acad Sci USA 2008, 105:64–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kroeze WK, Sassano MF, Huang X-P, Lansu K, McCorvy JD, Giguere PM, Sciaky N, Roth BL: PRESTO-Tango as an open-source resource for interrogation of the druggable human GPCRome. Nat Struct Mol Biol 2015, 22:362–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fan L, Tan L, Chen Z, Qi J, Nie F, Luo Z, Cheng J, Wang S: Haloperidol bound D2 dopamine receptor structure inspired the discovery of subtype selective ligands. Nat Commun 2020, 11:1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Li F, Jiang X, Luo L-L, Xu Y-M, Huang X-X, Huang G, Zhang Y: A piggyBac-based TANGO GFP assay for high throughput screening of GPCR ligands in live cells. Cell Commun Signal 2019, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Colosimo DA, Kohn JA, Luo PM, Piscotta FJ, Han SM, Pickard AJ, Rao A, Cross JR, Cohen LJ, Brady SF: Mapping interactions of microbial metabolites with human G-protein-coupled receptors. Cell Host Microbe 2019, 26:273–282.● Used bioassay guided fractionation to identify GPCR-activating compounds from microbiome culture. Opens the door for new experimental deorphanization strategy.
  • 24.Chen H, New P-K, Yang Y, Rosen CE, Bielecka AA, Kuchroo M, Cline GW, Kruse AC, Ring AM, Crawford JM, Palm NW: A forward chemical genetic screen reveals gut microbiota metabolites that modulate host physiology. Cell 2019, 177:1217–1231.●● Used β-arrestin recruitment assay to probe activation of GPCRs by human gut microbiota. This work identified an important member of the microbiome with links to both digestion and neural health.
  • 25.Laroche G, Giguère PM: Measurement of β-arrestin recruitment at GPCRs using the tango assay. Methods Mol Biol 2019, 1947:257–267. [DOI] [PubMed] [Google Scholar]
  • 26.Littmann T, Buschauer A, Bernhardt G: Split luciferase-based assay for simultaneous analyses of the ligand concentration- and time-dependent recruitment of beta-arrestin2. Anal Biochem 2019, 573:8–16. [DOI] [PubMed] [Google Scholar]
  • 27.Galinski S, Wichert SP, Rossner MJ, Wehr MC: Multiplexed profiling of GPCR activities by combining split TEV assays and EXT-based barcoded readouts. Sci Rep 2018, 8:8137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Dong S, Rogan SC, Roth BL: Directed molecular evolution of DREADDs: a generic approach to creating next-generation RASSLs. Nat Protoc 2010, 5:561–573. [DOI] [PubMed] [Google Scholar]
  • 29.Roth BL: DREADDs for Neuroscientists. Neuron 2016, 89:683–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yasi EA, Allen AA, Sugianto W, Peralta-Yahya P: Identification of three antimicrobials activating serotonin receptor 4 in colon cells. ACS Synth Biol 2019, 8:2710–2717.●● GPCR screen in S. cerevisiae to find agonists for the serotonin receptor 4, the pharmaceutical target for irritable bowel syndrome with constipation. Identified antibiotics as GPCR agonists. One of the most successful applications of a yeast-based GPCR high-throughput screening.
  • 31.Ehrenworth AM, Claiborne T, Peralta-Yahya P: Medium-throughput screen of microbially produced serotonin via a G-protein-coupled receptor-based sensor. Biochemistry 2017, 56:5471–5475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mukherjee K, Bhattacharyya S, Peralta-Yahya P: GPCR-based chemical biosensors for medium-chain fatty acids. ACS Synth Biol 2015, 4:1261–1269. [DOI] [PubMed] [Google Scholar]
  • 33.Minic J, Persuy M-A, Godel E, Aioun J, Connerton I, Salesse R, Pajot-Augy E: Functional expression of olfactory receptors in yeast and development of a bioassay for odorant screening. FEBS J 2005, 272:524–537. [DOI] [PubMed] [Google Scholar]
  • 34.Yasi EA, Eisen SL, Wang H, Sugianto W, Minniefield AR, Hoover KA, Branham PJ, Peralta-Yahya P: Rapid deorphanization of human olfactory receptors in yeast. Biochemistry 2019, 58:2160–2166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sarria S, Bartholow TG, Verga A, Burkart MD, Peralta-Yahya P: Matching protein interfaces for improved medium-chain fatty acid production. ACS Synth Biol 2018, 7:1179–1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Klenk C, Ehrenmann J, Schutz M, Pluckthun A: A generic selection system for improved expression and thermostability of G protein-coupled receptors by directed evolution. Sci Rep 2016, 6:21294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Heine P, Witt G, Gilardi A, Gribbon P, Kummer L, Plucktun A: High-throughput fluorescence polarization assay to identify ligands using purified G protein-coupled receptor. SLAS Discov 2019, 24:915–927. [DOI] [PubMed] [Google Scholar]
  • 38.Qin S, Meng M, Yang D, Bai W, Lu Y, Peng Y, Song G, Wu Y, Zhou Q, Zhao S et al. : High-throughput identification of G protein-coupled receptor modulators through affinity mass spectrometry screening. Chem Sci 2018, 9:3192–3199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lu Y, Qin S, Zhang B, Dai A, Cai X, Ma M, Gao Z-G, Yang D, Stevens RC, Jacobson KA et al. : Accelerating the throughput of affinity mass spectrometry-based ligand screening toward a G protein-coupled receptor. Anal Chem 2019, 91:8162–8169.● Added a re-incubation step to the affinitiy mass-spectrometry workflow for GPCR screening. The improved workflow increased throughput by two orders of magnitude.
  • 40.Foster SR, Hauser AS, Vedel L, Strachan RT, Huang X-P, Gavin AC, Shah SD, Nayak AP, Haugaard-Kedstrom LM, Penn RB et al. : Discovery of human signaling systems: pairing peptides to G protein-coupled receptors. Cell 2019, 179:895–908.● Used in silico screen to map co-evolution of peptide ligands and their GPCR targets. One of a limited number of in silico screens for peptide-GPCR binding.
  • 41.Wang J, Bhattarai A, Ahmad WI, Farnan TS, John KP, Miao Y: Computer-aided GPCR drug discovery In GPCRs: Structure, Function, and Drug Discovery. Edited by Jastrzebska B, Park PS-H. Academic Press; 2020:283–293. [Google Scholar]
  • 42.Lyu J, Wang S, Balius TE, Singh I, Levit A, Moroz YS, O’Meara MJ, Che T, Algaa E, Tolmachova K et al. : Ultra-large library docking for discovering new chemotypes. Nature 2019, 566:224–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Stein RM, Kang HJ, McCorvy JD, Glatfelter GC, Jones AJ, Che T, Slocum S, Huang X-P, Savych O, Moroz YS et al. : Virtual discovery of melatonin receptor ligands to modulate circadian rhythms. Nature 2020, 579:609–614.●● Used molecular docking to screen >150 million compounds, identifying and verifying 15 hits for melatonin receptors 1 and 2. Shows how in silico approaches can greatly improve the diversity of ligands that can be screened.
  • 44.Loo JSE, Emtage AL, Ng KW, Yong ASJ, Doughty SW: Assessing GPCR homology models constructed from templates of various transmembrane sequence identities: binding mode prediction and docking enrichment. J Mol Graph Model 2018, 80:38–47. [DOI] [PubMed] [Google Scholar]

RESOURCES