Original article
Homology modeling of MT1 and MT2 receptors

https://doi.org/10.1016/j.ejmech.2007.12.001Get rights and content

Abstract

Melatonin is a neurohormone synthesized and secreted mainly during the dark period of the circadian cycle by the pineal gland. It has already been proved to be involved in a number of chronobiological processes, most of them being mediated by its membranar receptors MT1 and MT2. Both are members of the GPCR class and, despite the interest they elicit, their 3D structure is still to be described. Models for both human MT1 and MT2 receptors have been constructed by homology modeling, using the X-ray structure of bovine rhodopsin as template. These models have been evaluated in terms of hydrophobic properties of the helices and refined to take into account the rearrangement of GPCRs necessary for their activation, thus leading to a putative activated model for each subtype.

Graphical abstract

We report here the construction of a 3D structure for both human MT1 and MT2 receptors and their careful modification in an attempt to propose a valid melatonin binding site.

  1. Download : Download full-size image

Introduction

Melatonin, N-acetyl-5-methoxytryptamine (Fig. 1) was isolated and its structure described in 1959 [1]. It is principally synthesized by the pineal gland and secreted during the dark period of the circadian cycle [2], due to the suppressive effect of light on the stimulatory retinohypothalamic pathway [3], [4]. This light-regulated cycle gives to melatonin a major role in seasonal and chronobiological processes. It is already employed for veterinary use, such as stimulation of fur growth for the mink or regulation of the ovine reproduction [5], [6]. Among the possibilities under investigation for its use in human clinics, its chronobiotic properties are widely involved. Numerous works have been devoted to the resynchronization of disturbed biological rhythms such as sleep shift caused by jet lag or night work, insomnia and seasonal affective disorders [7], [8], [9], [10], [11]. Overall, two kinds of melatonin binding sites have been identified so far. They have been localized and characterized initially on the basis of their affinity for 2-[125I]-iodomelatonin, giving rise to high affinity ML1 sites and low affinity ML2 sites [12]. Melatonin exerts the major part of its aforementioned effects by the activation of two high affinity binding sites denoted as MT1 [13] and MT2 [14], which belong to the G-protein coupled receptor superfamily, and a low affinity binding site, referred to as MT3, according to the IUPHAR nomenclature [15], which was recently identified as quinone reductase 2 (QR2 EC 1.6.99.2) [16], [17], an enzyme closely related to the detoxifying enzyme quinone reductase 1. Moreover, there is also another high affinity binding site, the subtype Mel1c that was first cloned from Xenopus laevis but not yet identified in mammals [18]. As the IUPHAR nomenclature concerns mammals only, Mel1c has kept its original name. The MT1 subtype is found in the retina, the kidneys and the brain, more particularly in the suprachiasmatic nuclei and thus should be implicated in the transmission of the circadian effects of melatonin and reproduction. This receptor could also be involved in peripheral vasoconstriction. The MT2 subtype is found in the retina and the brain too, but contrary to MT1, it does not appear in the suprachiasmatic nuclei. It could play a role in the physiology of the retina and in body temperature regulation [19], [20]. Structurally, MT1 is a 350 amino acid protein, presenting a sequence homology of 60% with the 362 amino acid MT2 [14]. The GPCR class is an important target for drugs due to the great diversity of their ligands, ranging from photons to peptides or proteins. Some studies suggest that 30–50% of known drugs actually target a GPCR to produce their effects [21], [22], [23]. These receptors display a common characteristic fold composed of seven transmembrane alpha-helices. Moreover, they present highly conserved residues, such as the DRY sequence of the intracellular end of transmembrane domain 3 (TM3) implied in the G-protein activation. This sequence is replaced by an NRY motif for melatonin receptors, which plays exactly the same role by coupling the receptor and a Gi or Gq protein, leading to a decrease in cAMP [24], [25]. However, GPCRs are extremely difficult to isolate and crystallize as their inclusion in the cell membrane contributes to their structural stability and renders them poorly purifiable [26]. This leads to the impossibility to determine directly their structure by standard means. The only way to cope with this problem is the construction of theoretical models based on the only GPCR crystallographic structure available, bovine rhodopsin, using a homology modeling approach [27], [28]. However, it is assumed that the high-resolution X-ray structure of bovine rhodopsin is in an inactive state and cannot stand for agonist ligand activated GPCRs. This GPCR activation process can be simulated by rotation of some transmembrane domains as suggested by experimental data [29], [30], [31]. We report here the construction of a 3D structure for both human MT1 and MT2 receptors and their careful modification toward an activated state using mutational and ligand SAR data in an attempt to propose a valid melatonin binding site. Such models may be very helpful to facilitate the design and development of new selective ligands.

Section snippets

Sequence alignment

The most important point in any homology modeling study, besides the choice of the reference, is the alignment of the sequences. The greatest attention was thus paid to the careful construction of a robust alignment. The transmembrane domains form the core of GPCRs, so it appeared safer to produce an alignment conserving the integrity of these domains (Fig. 2, Fig. 3). The helices of melatonin receptors were believed to be superposable with the experimentally known helices of bovine rhodopsin.

Conclusion

We report the construction of homology models of both subtypes of human melatonin receptors from the crystallographic structure of bovine rhodopsin. The resulting models should represent an inactive form of the receptors. As our interest was focused on agonists, we had to modify the arrangement of the helix bundle in order to create a putative active conformation of the melatoninergic receptors. Taking into account the data gathered by site directed mutagenesis studies, we have rotated two

Sequence alignment

Models were constructed using full length sequence alignment of the human MT1 [13] and MT2 [14] receptors (PIR entries I38848 and I38990, respectively) with bovine rhodopsin [53] (PIR entry OOBO) (http://www-nbrf.georgetown.edu/pirwww/dbinfo/pirpsd.html) [54]. In order to favour pertinent superimposition of the residues conserved throughout the GPCR family, the alignment of the sequences was broken down in two steps, allowing for a more flexible inspection of the results and easier corrections.

References (75)

  • A. Brzezinski et al.

    Sleep Med. Rev.

    (2005)
  • F.W. Turek et al.

    Sleep Med.

    (2004)
  • O. Vakkuri et al.

    Anal. Biochem.

    (1984)
  • S.M. Reppert et al.

    Neuron

    (1994)
  • O. Nosjean et al.

    J. Biol. Chem.

    (2000)
  • O. Nosjean et al.

    Biochem. Pharmacol.

    (2001)
  • P.A. Witt-Enderby et al.

    Life Sci.

    (2003)
  • L. Oliveira et al.

    Trends Pharmacol. Sci.

    (1994)
  • T. Kokkola et al.

    Biochem. Biophys. Res. Commun.

    (1998)
  • S.H. White et al.

    J. Biol. Chem.

    (2001)
  • E. Archer et al.

    Trends Pharmacol. Sci.

    (2003)
  • E.C. Hulme et al.

    Eur. J. Pharmacol.

    (1999)
  • F. Mseeh et al.

    Eur. J. Pharmacol.

    (2002)
  • M.J. Gerdin et al.

    Biochem. Pharmacol.

    (2003)
  • P. Mazna et al.

    Biochem. Biophys. Res. Commun.

    (2005)
  • C. Navajas et al.

    Eur. J. Pharmacol.

    (1996)
  • C.J. Grol et al.

    Bioorg. Med. Chem.

    (1996)
  • R.J. Mattson et al.

    Bioorg. Med. Chem. Lett.

    (2003)
  • J. Epperson et al.

    Bioorg. Med. Chem. Lett.

    (2004)
  • J. Nathans et al.

    Cell

    (1983)
  • P. Rice et al.

    Trends Genet.

    (2000)
  • S. Conway et al.

    Biochem. Biophys. Res. Commun.

    (1997)
  • T. Kokkola et al.

    Biochem. Pharmacol.

    (2003)
  • S. Conway et al.

    Biochem. Biophys. Res. Commun.

    (2001)
  • S. Conway et al.

    J. Biol. Chem.

    (2000)
  • G. Jones et al.

    J. Mol. Biol.

    (1997)
  • A.B. Lerner et al.

    J. Am. Chem. Soc.

    (1959)
  • W.B. Quay

    Proc. Soc. Exp. Biol. Med.

    (1964)
  • E.L. Bittman et al.

    Biol. Reprod.

    (1989)
  • D.N. Krause et al.

    Annu. Rev. Pharmacol.

    (1991)
  • J. Rose et al.

    J. Anim. Sci.

    (1984)
  • E.L. Bittman et al.

    Endocrinology

    (1983)
  • D. Dawson et al.

    J. Pineal Res.

    (1993)
  • S. Folkard et al.

    Chronobiol. Int.

    (1993)
  • N.E. Rosenthal et al.

    Arch. Gen. Psychiatry

    (1984)
  • S.M. Reppert et al.

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

    (1995)
  • Cited by (0)

    View full text