scispace - formally typeset
Open AccessPosted ContentDOI

Activation of GPR116/ADGRF5 by its tethered agonist requires key amino acids in extracellular loop 2 of the transmembrane region

Reads0
Chats0
TLDR
In this paper, the authors investigated the physiologic importance of autocatalytic cleavage upstream of the agonistic peptide sequence, an event necessary for NTF displacement and subsequent receptor activation.
Abstract
The mechanistic details of the tethered agonist mode of activation for adhesion GPCRs has not been completely deciphered. We set out to investigate the physiologic importance of autocatalytic cleavage upstream of the agonistic peptide sequence, an event necessary for NTF displacement and subsequent receptor activation. To examine this hypothesis, we characterized tethered agonist-mediated activation of GPR116 in vitro and in vivo. A knock-in mouse expressing a non-cleavable GPR116 mutant phenocopies the pulmonary phenotype of GPR116 knock-out mice, demonstrating that tethered agonist-mediated receptor activation is indispensable for function in vivo. Using site-directed mutagenesis and species swapping approaches we identified key conserved amino acids for GPR116 activation in the tethered agonist sequence and in extracellular loops 2/3 (ECL2/3). We further highlight residues in transmembrane7 (TM7) that mediate stronger signaling in mouse versus human GPR116 and recapitulate these findings in a model supporting tethered agonist:ECL2 interactions for GPR116 activation. Grant supportThis work was supported in part by HL131634 (JPB) from the National Heart, Lung and Blood Institute of the National Institutes of Health.

read more

Content maybe subject to copyright    Report

1
Activation of GPR116/ADGRF5 by its tethered agonist requires key amino acids in
extracellular loop 2 of the transmembrane region
Authors
James P. Bridges
1
, Caterina Safina
2
, Bernard Pirard
2
, Kari Brown
1
, Alyssa Filuta
1
, Rochdi
Bouhelal
2
, Sejal Patel
4
, Klaus Seuwen
2
, William E. Miller
3
, Marie-Gabrielle Ludwig
2,*
1
Department of Pediatrics, Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children’s
Hospital Medical Center, Cincinnati, OH, USA, 45229.- present address: Department of Medicine,
Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO,
USA, 80206
2
Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
3
Department of Molecular Genetics, Biochemistry and Microbioloby, University of Cincinnati
College of Medicine, Cincinnati, OH 45267
4
Novartis Institutes for Biomedical Research, Cambridge, USA
* Corresponding author; email: marie-gabrielle.ludwig@novartis.com
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted April 2, 2021. ; https://doi.org/10.1101/2021.04.01.438115doi: bioRxiv preprint

2
Grant support: This work was supported in part by HL131634 (JPB) from the National Heart,
Lung and Blood Institute of the National Institutes of Health.
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted April 2, 2021. ; https://doi.org/10.1101/2021.04.01.438115doi: bioRxiv preprint

3
Abstract
The mechanistic details of the tethered agonist mode of activation for adhesion GPCRs has not
been completely deciphered. We set out to investigate the physiologic importance of autocatalytic
cleavage upstream of the agonistic peptide sequence, an event necessary for NTF displacement
and subsequent receptor activation. To examine this hypothesis, we characterized tethered
agonist-mediated activation of GPR116 in vitro and in vivo. A knock-in mouse expressing a non-
cleavable GPR116 mutant phenocopies the pulmonary phenotype of GPR116 knock-out mice,
demonstrating that tethered agonist-mediated receptor activation is indispensable for function in
vivo. Using site-directed mutagenesis and species swapping approaches we identified key
conserved amino acids for GPR116 activation in the tethered agonist sequence and in
extracellular loops 2/3 (ECL2/3). We further highlight residues in transmembrane7 (TM7) that
mediate stronger signaling in mouse versus human GPR116 and recapitulate these findings in a
model supporting tethered agonist:ECL2 interactions for GPR116 activation.
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted April 2, 2021. ; https://doi.org/10.1101/2021.04.01.438115doi: bioRxiv preprint

4
Introduction
Adhesion G Protein-Coupled Receptors (aGPCRs) are involved in a variety of
pathophysiological processes, from development of the brain and the musculoskeletal system to
modulation of metabolism (Olaniru et al., 2019), immune responsiveness and angiogenesis.
Important roles in the central and peripheral nervous system have been described (reviewed in
Folts et al., 2019), exemplified by the genetic linkage of GPR56/ADGRG1 variants with bilateral
frontoparietal polymicrogyria (BFPP) pathology. The capacity of aGPCRs to modulate
inflammatory responses has been shown in several contexts (Lin et al., 2017), with leading data
on apoptotic cell sensing and phagocytosis (EMR2/ADGRE2, BAI1/ADGRB1), activation of mast
cells (EMR2 genetic link to vibratory urticaria) or NK cells (GPR56), and resistance to listeriosis
(CD97/ADGRE5). In addition, several lines of evidence suggest key roles for aGPCRs in cancer
where they may, for example, relay cellular signaling of mechanical cues (reviewed in Scholz,
2018).
A specific aspect of aGPCR biology is their mechanosensory role, an aspect that may
prove to be a general feature of these receptors (Langenhan, 2019). These properties are related
to the structure of aGPCRs and their mode of activation that was recently identified. Several
methods of activating aGPCRs have been identified, including using endogenous ligands, peptide
agonists and small molecules (reviewed in Bassilana et al., 2019). What may be seen as the
primary activation mode of aGPCRs is activation via a tethered agonistic peptide (Liebscher et
al., 2014; Stoveken et al., 2015). Intracellular autocatalytic cleavage of aGPCRs at the GPS site
upstream of the seven transmembrane domain (7TM) generates the NTF and CTF (N-terminal
and C-terminal fragments, respectively), with the amino terminal 15 to 27 amino acids of the CTF
being referred to as the tethered agonist. The NTF and CTF then re-associate non-covalently
during trafficking to the cytoplasmic membrane such that the tethered agonist is buried within the
C-terminal portion of the NTF (Araç et al., 2012). Interactions of the NTF with extracellular matrix
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted April 2, 2021. ; https://doi.org/10.1101/2021.04.01.438115doi: bioRxiv preprint

5
(ECM) components, or with neighboring cells, serves as an anchor for the receptor. Upon binding
of an additional ligand that changes the structural conformation, or generation of tension (e.g.
shear stress, stiffness of the ECM), the tethered agonist is released from the NTF thereby
activating the 7TM of the receptor. Several studies have shown constitutive basal activity of CTF
constructs and activation of full-length aGPCRs with exogenous synthetic peptides (GAP)
corresponding to their cognate tethered agonist sequence (Liebscher et al., 2014; Demberg et
al., 2015; Müller et al., 2015; Stoveken et al., 2015; Wilde et al., 2016; Brown et al., 2017; reviewed
in Bassilana et al., 2019). Some receptors such as CELSR1/ADGRC1, GPR115/ADGRF4,
GPR111/ADGRF2 do not possess a consensus GPS cleavage site and are not cleaved. Further,
the biological function of GPR114/ADGRG5 and LAT1 (LPHN1/ADGRL1) is not dependent on
the cleavage at the GPS site and such receptors may be activated by structural changes not
requiring the dissociation of the NTF and the CTF (Langenhan 2019; Scholz et al., 2017; Beliu et
al., 2021). In contrast, release of the NTF, concomitant to receptor activation, has been shown for
GPR56 upon binding to the ECM laminin 111 and transglutaminase 2 (Luo et al., 2014) and for
EMR2 bound to the ECM dermatan sulfate and subjected to mechanical cues (vibration) (Boyden
et al., 2016; Le et al., 2019; Naranjo et al., 2020).
However, the molecular details for the activation of aGPCRs by their tethered agonists
have not been well described; in part due to difficulty of generating 3D structures of GPCRs.
Multiple sequence alignments from previous reports revealed a 25-30% amino acid identity
between aGPCRs and class B1 GPCRs (Bjarnadóttir et al., 2007; Stacey et al., 2000). Available
3D structures of class B1 GPCRs (19 structures, 11 X-ray and 8 cryo-electron microscopy (cryo-
EM)) in the GPCR DB, www.gpcrdb.org as accessed on January 8
th
2020, have provided
templates for homology models which could be used to gain insights into the structure of the CTF
of aGPCRs. However, it remains unclear as to what extent this knowledge on class B receptors
will be directly applicable to the mechanism of aGPCR activation.
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted April 2, 2021. ; https://doi.org/10.1101/2021.04.01.438115doi: bioRxiv preprint

Figures
Citations
More filters
Journal ArticleDOI

Emerging roles of adhesion G protein-coupled receptors.

TL;DR: A review of the current knowledge around aGPCR activation, including ligand binding sites, the mechanism of autoproteolysis-inducing (GAIN) domain-mediated receptor activation and how a GPCR transmembrane domains may relate to activation can be found in this article.
References
More filters
Journal ArticleDOI

Protter: interactive protein feature visualization and integration with experimental proteomic data

TL;DR: Protter, a web-based tool that supports interactive protein data analysis and hypothesis generation by visualizing both annotated sequence features and experimental proteomic data in the context of protein topology, is presented.
Journal ArticleDOI

A novel evolutionarily conserved domain of cell-adhesion GPCRs mediates autoproteolysis.

TL;DR: The GAIN domain embodies a unique, evolutionarily ancient and widespread autoproteolytic fold whose function is likely relevant for GPCR signalling and for multiple human diseases.
Journal ArticleDOI

Family-B G-protein-coupled receptors.

TL;DR: The structures and functions of family-B GPCRs are described and a simplified nomenclature for these proteins are proposed and it is suggested that these proteins have a common evolutionary origin for all of them.
Journal ArticleDOI

Lifting the lid on GPCRs: the role of extracellular loops

TL;DR: A review of the key roles of extracellular loops in ligand binding, activation and regulation of both family A and family B GPCRs is presented in this article, with a focus on the role of ECL segments.
Journal ArticleDOI

A Tethered Agonist within the Ectodomain Activates the Adhesion G Protein-Coupled Receptors GPR126 and GPR133.

TL;DR: It is shown that a short peptide sequence within the ectodomain of two aGPCRs functions as a tethered agonist and may prompt the development of specific ligands for this currently untargeted GPCR family.
Related Papers (5)
Frequently Asked Questions (12)
Q1. What have the authors contributed in "Activation of gpr116/adgrf5 by its tethered agonist requires key amino acids in extracellular loop 2 of the transmembrane region" ?

A specific aspect of aGPCR biology is their mechanosensory role, an aspect that may be a general feature of these receptors this paper. 

In order to rule out potential artifacts resulting from oxidation, the methionine corresponding to position 999 in the mCTF was replaced by norleucine (Nle). 

While the endogenous ligand or the biological process(es) leading to displacement of the non-covalently linked N-terminal domain and subsequent GPR116 activation in vivo remains unknown, their data advance the mechanistic understanding of GPR116 activation in the context of pulmonary surfactant homeostasis and may facilitate the development of novel receptor modulators that can be used to treat clinically relevant lung diseases. 

the five key amino acids in the ECLs that form suggested contact points for the tethered ligand were used to provide constraints and to include the ECLs in the modeling. 

A knock-in mouse expressing a noncleavable GPR116 mutant phenocopies the pulmonary phenotype of GPR116 knock-out mice, demonstrating that tethered agonist-mediated receptor activation is indispensable for function in vivo. 

In their previous study, mutation of the tethered agonist amino acids to alanines using sequential steps of 3 amino acids for each mutant highlighted the role of this N-terminal sequence in receptor activation. 

In a related series of experiments the authors aimed at elucidating whether conservative aminoacid replacements within these key residues in mCTFbax would restore receptor activation of the single point alanine mutants. 

replacing L1166 with an isoleucine, as is observed in some aGPCRs, was not tolerated in GPR116, suggesting a specific role of this amino acid. 

In parallel, while analyzing the potential binding mode of a low molecular weight antagonistspecific for the murine receptor discovered in house at Novartis the authors began considering additional other amino acids deeper in the transmembrane domains of the receptor that might be responsible for the species differences in efficacy. 

The tyrosine at position 1158 is located 6 amino acids upstream of the CWL cluster and itis conserved in most aGPCRs, except in the ADGRA and ADGRG subfamily members, and ADGRV1 (Suppl Fig11). 

The H991A-V5 cDNA construct was generated by gene synthesis (Genewiz, South Plainfield, NJ), sequenced verified and cloned into pcDNA3.1+ for expression in cultured cells. 

Similar to observations with the full-length receptors, the hCTFbax is less responsive to14GAP14 than the mCTFbax receptor (Fig5B).