Elsevier

Peptides

Volume 29, Issue 5, May 2008, Pages 700-710
Peptides

Solution structure of urotensin-II receptor extracellular loop III and characterization of its interaction with urotensin-II

https://doi.org/10.1016/j.peptides.2008.02.024Get rights and content

Abstract

Urotensin-II (U-II) is a vasoactive hormone that acts through a G-protein-coupled receptor named UT. Recently, we have shown, using the surface plasmon resonance technology that human U-II (hU-II) interacts with the hUT(281–300) fragment, a segment containing the extracellular loop III (EC-III) and short extensions of the transmembrane domains VI and VII (TM-VI and TM-VII). To further investigate the interaction of UT receptor with U-II, we have determined the solution structure of hUT(281–300) by high-resolution NMR and molecular modeling and we have examined, also using NMR, the binding with hU-II at residue level. In the presence of dodecylphosphocholine micelles, hUT(281–300) exhibited a type III β-turn (Q285–L288), followed by an α-helical structure (A289–L299), the latter including a stretch of transmembrane helix VII. Upon addition of hU-II, significant chemical shift perturbations were observed for residues located just on the N-terminal side of the β-turn (end of TM-VI/beginning of EC-III) and on one face of the α-helix (end of EC-III/beginning of TM-VII). These data, in conjunction with intermolecular NOEs, suggest that the initiation site of EC-III, as well as the upstream portion of helix VII, would be involved in agonist binding and allow to propose points of interaction in the ligand–receptor complex.

Introduction

Urotensin-II (U-II) is a disulfide-bridged peptide that was originally isolated from the goby Gillichthys mirabilis urophysis [5]. U-II isoforms were identified in many species of fish [6], [14], as well as in amphibians [13] and mammals [15], [16] including human [15]. Human U-II (hU-II) is characterized, as other isoforms, by a conserved cysteine-linked macrocycle CFWKYC but is the shortest isoform with 11 amino acids (ETPD [CFWKYC]V-OH) [15]. Several reports have revealed the potent vasoconstricting effects of U-II, and they described it as the most potent mammalian vasoconstrictor identified to date. However, the physiological role of U-II in the mammalian cardiovascular hemodynamics and vascular smooth muscle tone control is still not fully understood. For instance, diverse effects have been reported, from vasoconstricting to vasodilating responses [19]. Nevertheless, despite the heterogeneity of its vascular activities, U-II exerts a large array of physiological effects suggesting that it might be involved in pathological processes such as cardiovascular and renal diseases [55].

It has been shown that U-II action is mediated through a specific cell-surface receptor belonging to the superfamily of G-protein-coupled receptors (GPCRs). The U-II human receptor has been identified in 1999 [3], [34] and is predominantly expressed in cardiovascular tissues and the motor neurons of spinal cord [3]. Previously known as an orphan G-protein-coupled receptor (GPR14), it is now designated as UT receptor [18]. This receptor belongs to class A, the rhodopsin-like family, and it contains 389 residues. It shares the GPCR common structure made of seven basic transmembrane domains, connected by extracellular and intracellular loops (Fig. 1).

Several data from structure–activity relationship (SAR) studies described the pharmacophoric requirements of U-II for receptor activation [17], [23], [29]. Hence, it has been shown that the N-terminal segment of the U-II molecule is not essential for the affinity and the activity, but that its conserved C-terminal cyclic core [CFWKYC] is necessary for high-affinity binding to UT receptor. Moreover, these studies demonstrated the importance of the Trp, Lys, and Tyr residues that are required for receptor recognition and activation [17], [26]. NMR spectroscopy analyses of U-II in DMSO [22], water [17] and SDS [10] provided information about the precise spatial orientation of residues that are crucial for the interaction with the receptor. Thus, U-II structure shows that the N-terminal domain is in random coil, in contrast to the core region of the peptide that adopts a highly ordered compact conformation, induced by the disulfide bridge, with the presence on one side of the molecule of a hydrophobic cluster formed by the Phe, Trp and Tyr residues [22], [26]. These data allow to postulate that this cluster may interact with a hydrophobic pocket located in the UT receptor or at its surface.

In contrast with U-II, because of its numerous SAR and conformational studies, our knowledge about the conformation and structural determinants of the UT receptor remains limited. Indirect methods such as bioinformatic techniques were used to explore UT conformation and ligand/UT complexes. The first theoretical model of gU-II/rUT was proposed by Kinney et al. [26]. They built a molecular complex in which the lysine residue of U-II was aligned towards the Asp130 side chain of transmembrane domain III. This structural domain arrangement allowed tight contacts between the receptor and the Trp, Lys and Tyr side chains of U-II. More recently, Lavecchia et al. have developed a model of peptidic or non-peptidic U-II agonist–hUT complexes, which gave similar information to those of Kinney et al. on the putative ligand-binding pocket [31]. Although both models offer a qualitative assessment of potential interactions of ligand/UT, they can only serve to generate hypotheses that must be validated by experimental data. Boucard et al. were the first to provide further data on the nature of the interaction between hU-II/rUT [8]. Using directed mutagenesis in combination with photolabeling techniques, they showed a close proximity of the core residues of U-II with the fourth transmembrane domain of rUT.

In the case of membrane proteins such as GPCRs, the traditional methods of NMR or X-ray crystallography are generally not applicable for obtaining high-resolution 3D structures. Alternative approaches are therefore required to get information on the receptor architecture and the structural characteristics that are involved in the recognition process. This is extremely important considering theoretical models, generated by homology modeling, provide little information on the structure of the extra- and intracellular regions of the receptor. Synthetic peptides representing domains of the molecule were used to gain structural information relevant to describe the intact membrane receptor [12], [35], [39], [46], [48], [54]. Although this method might not be absolutely sure, its usefulness was demonstrated for bacteriorhodopsin and rhodopsin [1], [2], [25]. For both proteins, it was shown using NMR spectroscopy that synthetic fragments exhibited the same secondary structures as those found in the crystal structure of the whole receptor [1], [2], [25].

Hence, in a previous study, we used this strategy to probe some of the UT structural features. We first synthesized three fragments corresponding to hUT(110–127), hUT(182–212) and hUT(281–300). Each peptide included an extracellular domain in which, at both extremities, a few residues of the putative transmembrane helices were added (Fig. 1). Using the surface plasmon resonance technology, we showed that hU-II interacted with hUT(281–300) [7], indicating for the first time that the hUT third extracellular loop III (EC-III) might be involved in the hU-II recognition process. Thus, in the present paper, we describe a study aiming at determining hUT extracellular loop III solution structure and characterizing its interaction with hU-II. We first solved by high-resolution NMR the three-dimensional structure of hUT(281–300) in DPC micelles, a membrane-mimetic environment. Then, we examined the binding with hU-II at residue level using the chemical shift perturbation approach.

Section snippets

Materials

Phospholipids dodecylphosphocholine-d38 (99.1%) were purchased from C/D/N isotopes (Pointe-Claire, Canada), D2O and sodium 2,2-dimethyl-2-silapentane-5-sulfonate (DSS) were from Euriso-top (Gif-sur-Yvette, France).

hU-II and the hUT(281–300) fragment were synthesized and purified in our laboratory following the previously published procedure [7].

NMR spectroscopy

All NMR experiments were performed on a Bruker Avance DMX 600 NMR spectrometer (Wissembourg, France), equipped with a triple resonance cryoprobe

NMR study

hUT(281–300) proton resonance assignment was carried out at 313 K by following the strategy proposed by Wüthrich [53]. Spin systems were identified using a combination of 2D TOCSY and COSY spectra and neighboring residues were connected through 2D NOESY experiments. The two proline residues present in the hUT(281–300) sequence (P287 and P290) both displayed the typical NOE pattern of strong Hiα/Hi+1(Pro)δ accounting for a trans conformation of the Xxx-Pro amide bonds. Nevertheless, compared to

Discussion

A body of evidence shows that U-II plays a key role in renal and cardiovascular physiology [55]. Thus, the U-II/UT biological system exhibits a remarkable potential for the development of novel therapeutic strategies, especially those related to the treatment of cardiovascular diseases. Such developments require a precise knowledge of the interactions between U-II and its receptor. Understanding these interactions is the first step towards the rational design of peptidic and non-peptidic U-II

Acknowledgements

The authors thank the Centre de Ressources Informatiques de Haute-Normandie (France) for NMR and molecular modeling software facilities. We also thank Marie Chabbert for helpful discussions. Financial supports were obtained from the Canadian Institutes for Health Research (CIHR), the Ministère de l’Éducation, des Loisirs et du Sport du Québec and the Région Haute-Normandie.

References (55)

  • J. Li et al.

    Structure of bovine rhodopsin in a trigonal crystal form

    J Mol Biol

    (2004)
  • Q. Liu et al.

    Identification of urotensin II as the endogenous ligand for the orphan G-protein-coupled receptor GPR14

    Biochem Biophys Res Commun

    (1999)
  • D. Marion et al.

    Rapid recording of 2D NMR spectra without phase cycling. Application to the study of hydrogen exchange in proteins

    J Magn Reson

    (1989)
  • R.T. McKay et al.

    Interaction of the second binding region of troponin I with the regulatory domain of skeletal muscle Troponin C as determined by NMR spectroscopy

    J Biol Chem

    (1997)
  • M. Nilges

    Calculation of protein structures with ambiguous distance restraints. Automated assignment of ambiguous NOE crosspeaks and disulphide connectivities

    J Mol Biol

    (1995)
  • D. Onan et al.

    Urotensin II: the old kid in town

    Trends Endocrinol Metab

    (2004)
  • S.P. So et al.

    Identification of residues important for ligand binding of thromboxane A2 receptor in the second extracellular loop using the NMR experiment-guided mutagenesis approach

    J Biol Chem

    (2003)
  • R.S. Ames et al.

    Human urotensin-II is a potent vasoconstrictor and agonist for the orphan receptor GPR14

    Nature

    (1999)
  • O.M. Becker et al.

    G protein-coupled receptors: in silico drug discovery in 3D

    Proc Natl Acad Sci USA

    (2004)
  • H.A. Bern et al.

    A reference preparation for the study of active substances in the caudal neurosecretory system of teleosts

    J Endocrinol

    (1969)
  • S. Boivin et al.

    Characterization of urotensin-II receptor structural domains involved in the recognition of U-II, URP, and urantide

    Biochemistry

    (2006)
  • A.A. Boucard et al.

    Photolabelling the rat urotensin II/GPR14 receptor identifies a ligand-binding site in the fourth transmembrane domain

    Biochem J

    (2003)
  • B.R. Brooks et al.

    CHARMM: a program for macromolecular energy minimization and dynamics calculations

    J Comput Chem

    (1983)
  • A. Carotenuto et al.

    Unraveling the active conformation of urotensin II

    J Med Chem

    (2004)
  • D.A. Chung et al.

    NMR structure of the second intracellular loop of the alpha 2A adrenergic receptor: evidence for a novel cytoplasmic helix

    Biochemistry

    (2002)
  • J.M. Conlon et al.

    Distribution and molecular forms of urotensin II and its role in cardiovascular regulation in vertebrates

    J Exp Zool

    (1996)
  • Y. Coulouarn et al.

    Cloning of the cDNA encoding the urotensin II precursor in frog and human reveals intense expression of the urotensin II gene in motoneurons of the spinal cord

    Proc Natl Acad Sci USA

    (1998)
  • Cited by (13)

    • Extracellular loop 3 substitutions K589N and A590S in FSH receptor increase FSH-induced receptor internalization and along with S588T substitution exhibit impaired ERK1/2 phosphorylation

      2018, Archives of Biochemistry and Biophysics
      Citation Excerpt :

      It might also host allosteric binding sites. EL3 seems to be essential for appropriate folding and signaling of the receptor [10], might be a binding site itself [11,12], and might also contribute to allosteric binding sites [13]. The molecular interaction of follicle stimulating hormone (FSH), with its specific receptor, the FSHR is not well understood.

    • Importance of the extracellular loops in G protein-coupled receptors for ligand recognition and receptor activation

      2011, Trends in Pharmacological Sciences
      Citation Excerpt :

      Such interactions must be receptor-specific because the presence of cysteine residues and glycosylation patterns vary considerably [52]. For the neurotensin 1 receptor, the urotensin receptor and the CC chemokine 2 receptor (CCR2), EL3 has been suggested to be part of the ligand binding site that is supposed to form a tunnel between EL3, TM6 and TM7 [18,20,54,55]. The crystal structure of the chemokine receptor CXCR4 shows that EL3, together with the N-terminus, shapes the entrance to the ligand binding pocket [12].

    View all citing articles on Scopus
    View full text