Review
The Potential of 19F NMR Application in GPCR Biased Drug Discovery

https://doi.org/10.1016/j.tips.2020.11.001Get rights and content

Highlights

  • Studies indicate that G protein-coupled receptor (GPCR) activation is a multistate transition process instead of a simple switch, suggesting the possibility of modulating the GPCR function through tuning of these individual states.

  • A biased ligand was proposed to specifically target one signaling pathway over others in a multiple-signaling coexistence system. However, despite the progress in structural biology, a detailed mechanism of how this is achieved remains elusive.

  • Research has indicated that different GPCR conformations interact with different downstream partners, dictating various pharmacological outputs.

  • 19F NMR has been demonstrated to be extremely promising in delineating GPCR conformations, attributable to its quantitative properties and high sensitivity toward electrostatic changes of the probe microenvironment. Thus, there is tremendous potential for biased drug discovery by establishing a rigorous correlation between individual receptor conformations and the pharmacological consequence upon ligand binding.

Although structure-based virtual drug discovery is revolutionizing the conventional high-throughput cell-based screening system, its limitation is obvious, together with other critical challenges. In particular, the resolved static snapshots fail to represent a full free-energy landscape due to homogenization in structural determination processing. The loss of conformational heterogeneity and related functional diversity emphasize the necessity of developing an approach that can fill this space. In this regard, NMR holds undeniable potential. However, outstanding questions regarding the NMR application remain. This review summarizes the limitations of current drug discovery and explores the potential of 19F NMR in establishing a conformation-guided drug screening system, advancing the cell- and structure-based discovery strategy for G protein-coupled receptor (GPCR) biased drug screening.

Section snippets

Biased Agonism in the GPCR

Biased agonism is a ligand-based signaling preference [1] observed when multiple signal pathways coexist in a signaling process. Since its introduction in the 1990s [2], the study of biased agonism has overwhelmingly centered on G protein-coupled receptors (GPCRs) (see Glossary) and, in particular, focused on two classical signal pathways: G protein and β-arrestin [3] (Figure 1). As a matter of fact, the concept of biased agonism can be applied to any signaling where the ligand-activated

Lack of Molecular Understanding of Biased Signaling

While significant progress has been made in linking distinct pharmacological phenomena to various ligand bindings and their signaling effectors, such as taking advantage of approaches like bioluminescence resonance energy transfer (BRET) [10,11], the following has not yet been fully established: (i) techniques for quantitative analyses of ligand functional selectivity [12] in order to identify the roles of individual signal pathways in a multisignaling system; (ii) techniques for predicting the

Missing Parts in Current Drug Screening Systems

With differential ligand binding, GPCRs can interact selectively with various intracellular partners, resulting in different downstream signaling and pharmacological effects. Although studies have advanced our understanding of the GPCR activation process, the functional relevance of each individual conformation and their roles in the signaling process remains elusive. The increased number of protein structures and the advancement of computational tools such as pharmacophore models [32],

Limitation of Cell- and Structure-Based Drug Discovery Systems

With the advancement of X-ray crystallography and cryo-electron microscopy (cryo-EM), structural biology has made tremendous progress. So far, over 370 structures of more than 70 GPCRs [48] have been resolved, providing unprecedented structural insights into receptor activation and allostery. Despite these, X-ray and cryo-EM are unable to elucidate dynamics of individual proteins or PPIs. Structural snapshots cannot capture a continuous conformational transition, extant structures associated

Progress of NMR Application in Receptor Conformational Delineation

It has been reported that the fluorine nucleus has a distinctly high gyromagnetic ratio and, thus, greatest sensitivity for NMR, next to tritium and 1H nuclei [67]. 19F NMR as a result exhibits a broad scope of chemical shifts over 1000 ppm [68], indicating a remarkable sensitivity to surrounding environmental changes, with potential to detect the subtle electrostatic changes associated with receptor activation. This provides a plausibility for rational design of biased drugs by delineating the

Concluding Remarks and Future Perspectives

The current drug discovery strategy is predominantly based on the measurement of dose-dependent downstream signaling such as the signaling levels of cAMP or Ca2+. Therefore, receptor activation is typically described as an on/off two-state switch. As a result of this oversimplification, developed drugs based on the current system tend to overactivate or oversuppress downstream signaling, resulting in concomitant side effects. A detailed mechanistic understanding of correlations between ligand,

Acknowledgments

The article was supported by Nexus Initiative (L.Y.) from University of South Florida (USF) as well as startup funding (L.Y) from the department of Cell Biology, Microbiology and Molecular Biology at USF.

Disclaimer Statement

L.Y. is an independent principal investigator at University of South Florida as well as being affiliated with H. Lee Moffitt Cancer Center and Research Institute. None of the authors have conflicts of interest with their affiliations.

Glossary

Balanced agonist
a drug that increases activities of multiple signaling pathways.
Biased ligand
a drug that increases the activity of a specific signaling pathway.
Cryo-electron microscopy (cryo-EM)
a procedure that deep-freezes a sample and uses electrons to make an image of protein structure.
G protein-coupled receptor (GPCR)
a class of seven-transmembrane proteins that transmit extracellular to intracellular signals and are triggered by a wide range of factors, including light, compounds, peptides,

References (87)

  • M. Congreve

    Impact of GPCR structures on drug discovery

    Cell

    (2020)
  • Q. Wan

    Mini G protein probes for active G protein–coupled receptors (GPCRs) in live cells

    J. Biol. Chem.

    (2018)
  • L. Ye

    Utilizing tagged paramagnetic shift reagents to monitor protein dynamics by NMR

    Biochim. Biophys. Acta, Proteins Proteomics

    (2017)
  • J. Broecker

    Crystallogenesis of membrane proteins mediated by polymer-bounded lipid nanodiscs

    Structure

    (2017)
  • R. Puthenveetil et al.

    Solution NMR: a powerful tool for structural and functional studies of membrane proteins in reconstituted environments

    J. Biol. Chem.

    (2019)
  • H. Winkler

    Tomographic subvolume alignment and subvolume classification applied to myosin V and SIV envelope spikes

    J. Struct. Biol.

    (2009)
  • F. Förster et al.

    Structure determination in situ by averaging of tomograms

    Methods Cell Biol.

    (2007)
  • P. Zhang

    Advances in cryo-electron tomography and subtomogram averaging and classification

    Curr. Opin. Struct. Biol.

    (2019)
  • J.S. Smith

    Biased signalling: from simple switches to allosteric microprocessors

    Nat. Rev. Drug Discov.

    (2018)
  • M.G. Eason

    Contribution of ligand structure to activation of alpha 2-adrenergic receptor subtype coupling to Gs

    Mol. Pharmacol.

    (1994)
  • H. Wei

    Independent-arrestin 2 and G protein-mediated pathways for angiotensin II activation of extracellular signal-regulated kinases 1 and 2

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • M.C. Michel

    Dynamic bias and its implications for GPCR drug discovery

    Nat. Rev. Drug Discov.

    (2014)
  • S. Rajagopal

    Quantifying ligand bias at seven-transmembrane receptors

    Mol. Pharmacol.

    (2011)
  • T.A. Johnson

    Identification of the first marine-derived opioid receptor “balanced” agonist with a signaling profile that resembles the endorphins

    ACS Chem. Neurosci.

    (2016)
  • Y. Namkung

    Functional selectivity profiling of the angiotensin II type 1 receptor using pathway-wide BRET signaling sensors

    Sci. Signal.

    (2018)
  • H. Kobayashi

    Bioluminescence resonance energy transfer-based imaging of protein-protein interactions in living cells

    Nat. Protoc.

    (2019)
  • N.C. Dale

    NanoBRET: the bright future of proximity-based assays

    Front. Bioeng. Biotechnol.

    (2019)
  • M.C. Michel et al.

    Biased agonism in drug discovery – is it too soon to choose a path?

    Mol. Pharmacol.

    (2018)
  • C.M. Costa-Neto

    A pluridimensional view of biased agonism

    Mol. Pharmacol.

    (2016)
  • T. Kenakin et al.

    Seven transmembrane receptors as shapeshifting proteins: the impact of allosteric modulation and functional selectivity on drug discovery

    Pharmacol. Rev.

    (2010)
  • D.P. Staus

    Allosteric nanobodies reveal the dynamic range and diverse mechanisms of G-protein-coupled receptor activation

    Nature

    (2016)
  • L. Ye

    Activation of the A2A adenosine G-protein-coupled receptor by conformational selection

    Nature

    (2016)
  • L. Ye

    Mechanistic insights into allosteric regulation of the A2A adenosine G-protein-coupled receptor by physiological cations

    Nat. Commun.

    (2018)
  • M. Casiraghi

    Functional modulation of a G protein-coupled receptor conformational landscape in a lipid bilayer

    J. Am. Chem. Soc.

    (2016)
  • T.H. Kim

    The role of ligands on the equilibria between functional states of a G protein-coupled receptor

    J. Am. Chem. Soc.

    (2013)
  • J.J. Liu

    Biased signaling pathways in β2-adrenergic receptor characterized by 19F-NMR

    Science

    (2012)
  • R. Horst

    β2-adrenergic receptor activation by agonists studied with 19F NMR spectroscopy

    Angew. Chem. Int. Ed.

    (2013)
  • L. Sušac

    A2A adenosine receptor functional states characterized by 19F-NMR

    Proc. Natl. Acad. Sci. U. S. A.

    (2018)
  • D. Hilger

    Structure and dynamics of GPCR signaling complexes

    Nat. Struct. Mol. Biol.

    (2018)
  • S.G.B. Furness

    Ligand-dependent modulation of G protein conformation alters drug efficacy

    Cell

    (2016)
  • T. Kenakin et al.

    Signalling bias in new drug discovery: detection, quantification and therapeutic impact

    Nat. Rev. Drug Discov.

    (2012)
  • D. Wacker

    Structural features for functional selectivity at serotonin receptors

    Science

    (2013)
  • R. Pascual

    A new pharmacophore model for the design of sigma-1 ligands validated on a large experimental dataset

    Front. Pharmacol.

    (2019)
  • Cited by (15)

    • <sup>19</sup>F NMR: A promising tool for dynamic conformational studies of G protein-coupled receptors

      2022, Structure
      Citation Excerpt :

      This difference altered the electrostatic environments that surrounded the probes BTFMA and TET, causing the different observed chemical shifts. Biomolecular dynamics play a major role in the context of ligand binding, receptor activation, allosteric modulation, and biased signaling, and are highly relevant to a specific physiological or pharmacological output (Latorraca et al., 2017; Wang et al., 2021b). So far, GPCR dynamics have been primarily studied computationally using molecular dynamics (MD) simulations (Mafi et al., 2022) due to the challenges of analyzing them experimentally.

    • Conformational dynamics in GPCR signaling by NMR

      2022, Magnetic Resonance Letters
    • Small, but powerful and attractive: <sup>19</sup>F in biomolecular NMR

      2022, Structure
      Citation Excerpt :

      NMR studies on membrane proteins, in particular GPCRs, have exploited 19F labels to investigate conformational changes upon agonist/antagonist binding in this important class of receptors, with the goal to correlate specific receptor conformations to the pharmacological outcome of ligand binding. Indeed, studies of this kind possess an appealing potential for drug screening and drug discovery (see recent reviews by Wang and others [Picard and Prosser, 2021; Wang et al., 2021b]). In most cases, 19F labels are conjugated to naturally present cysteines near the cytoplasmic face of the receptor (Frei et al., 2020) or on strategically introduced Cys residues at positions sensitive to signal transduction.

    View all citing articles on Scopus
    View full text