Minireview
Comparative genomics of leucine-rich repeats containing G protein-coupled receptors and their ligands

https://doi.org/10.1016/j.ygcen.2007.06.022Get rights and content

Abstract

Leucine-rich repeats containing G protein-coupled receptors (LGRs) constitute a unique cluster of transmembrane proteins sharing a large leucine-rich extracellular domain for hormone binding. In mammals, LGRs steer important developmental, metabolic and reproductive processes as receptors for glycoprotein hormones and insulin/relaxin-related proteins. In insects, a receptor structurally related to human LGRs mediates the activity of the neurohormone bursicon thereby regulating wing expansion behaviour and remodelling of the newly synthesized exoskeleton. In the past decade, novel insights into the molecular evolution of LGR encoding genes accumulated rapidly due to comparative genome analyses indicating that the endocrine LGR signalling system likely emerged before the radiation of metazoan phyla and expanded throughout evolution. Here, we present a short survey on the evolution of LGRs and the hormones they interact with.

Introduction

G protein-coupled receptors (GPCRs) are transmembrane proteins that respond to a diverse array of stimuli [e.g., light, odorants, taste molecules, lipid analogous, amines, nucleotides, peptides and protein hormones] and transmit these signals intracellularly through activation of heterotrimeric G proteins (Joost and Methner, 2002). Genome analyses identified GPCRs as one of the largest functional protein “superfamilies” with about eight hundred human GPCRs (Bjarnadottir et al. 2006), one thousand in the nematode, Caenorhabditis elegans (Bargmann, 1998) and two hundred in the fruit fly, Drosophila melanogaster (Brody and Cravchik, 2000, Vanden Broeck, 2001a). The majority of GPCRs is believed to act as olfactory receptors, while a distinct subset regulates developmental processes as receptors for peptide or protein hormones.

GPCRs share several key structural features, i.e., seven α-helical membrane-spanning segments interconnected by three intra- and extracellular loops (the “serpentine region”) and an intracellular C-terminal tail. Within the large rhodopsin-like receptor family (“family A GPCRs”), leucine-rich repeats containing GPCRs (LGRs) constitute a distinct subgroup characterized by a peculiar protein architecture. A true hallmark for LGRs is the large N-terminal extracellular (ecto-)domain involved in selective hormone binding (Braun et al. 1991) that is composed of tandem arrays of leucine-rich repeat (LRR) amino acid motifs, N- and C-terminally flanked by cysteine-rich sequences (Kajava, 1998). A so-called hinge region connects this ectodomain with the serpentine region.

In mammals, LGRs mediate the activity of large glycoprotein hormones, i.e., gonadotropins [follicle stimulating hormone (FSH), luteinizing hormone (LH), choriogonadotropin (CG)] and thyroid stimulating hormone (TSH) and, in consequence, have long been known to act as key players in the endocrine regulation of reproduction (gonadotropins) and metabolism (TSH) (Grossmann et al., 1997, Themmen and Huhtaniemi, 2000). Recently, genomic approaches forced us to revise the evolution of the LGR signalling system. Novel insights into the molecular evolution of LGR encoding genes emerged and demonstrated that LGRs predate the divergence of major animal phyla and expanded throughout metazoan evolution. Here, we present an overview of recent accomplishments illustrating the molecular evolution of the LGR/hormone endocrine system in metazoans.

Section snippets

Leucine-rich repeats containing GPCRs exist in distinct animal phyla and can be classified into three subtypes

Multiple LGR encoding genes from diverse vertebrates and invertebrates have been identified in silico from genomic and/or complementary DNA resources, mainly based on the presence of the N-terminal ectodomain. Based on overall sequence similarity and the architecture of the ectodomain, i.e., the number of leucine-rich repeats and the presence or absence of a low density lipoprotein motif, LGRs can be classified into three subtypes (Fig. 1).

Subtype A contains receptors for glycoprotein hormones

A first LGR subclass comprises vertebrate receptors for cystine knot-forming gonadotropins (FSH, LH, CG) and TSH, all ∼30 kDa dimeric proteins composed of a common α-subunit (glycoprotein hormone subunit alpha 1, GPA1) that non-covalently associates with a hormone-specific β-subunit (FSHβ, LHβ, CGβ or TSHβ). Apart from these “classic” glycoprotein hormone subunits, two additional cystine knot-containing proteins, related to the common α- and hormone-specific β-subunit, respectively, are encoded

Subtype B contains human orphan receptors with essential roles in development and receptors for the arthropod bursicon hormone

LGRs of subtype B are characterized by the presence of 13–18 LRRs. Here, three paraloguous mammalian receptors for unknown ligands (LGRs 4–6) form a cluster (McDonald et al., 1998, Hsu et al., 1998, Hsu et al., 2000) (Fig. 1). Taking advantage of gene trapping approaches and immunohistochemical analyses, a detailed atlas of mouse LGR4 expression was realized, indicating expression in a broad range of tissues including kidney, bone/cartilage, heart, stomach and nervous cells (Mazerbourg et al.,

Subtype C contains receptors for human relaxin-related proteins

In addition to 7–9 LRRs in the ectodomain, all subtype C LGRs contain a N-terminal cysteine-rich low density lipoprotein (LDL) motif not found in other LGRs. The first member of this subtype was cloned from the central nervous system of the mollusc, Lymnea stagnalis (snail) and is predominantly expressed in a small subset of neurons within the central nervous system and to a lesser extent in the heart (Tensen et al. 1994). At present, this mollusc receptor still has the status of an orphan

General discussion

Leucine-rich repeats containing GPCRs set an interesting example of how a receptor subfamily “expanded” through comparative genomics. Genome analysis and molecular cloning efforts revealed the exceptional preservation of LGRs in metazoa, both structurally and at the amino acid sequence level. The early origin of LGRs is illustrated by the existence of a receptor related to human glycoprotein hormone receptors in cnidarians [the sea anemone (A. elegantissima)], one of the most primitive animal

Conclusion

Comparative genomics has proven a valuable tool for deciphering co-evolution of GPCRs and their (putative) ligands leading to novel insights in the evolutionary conservation of signalling systems between vertebrates and invertebrates (Bargmann, 1998, Vanden Broeck, 2001a, Vanden Broeck, 2001b, Hewes and Taghert, 2001, Claeys et al., 2005) and which has been described here for leucine-rich repeats containing GPCRs. Where initially it was assumed that LGRs existed exclusively in vertebrates, new

Acknowledgments

We gratefully acknowledge Julie Puttemans for figure drawing. Our research is financially supported by the “Belgian program on Interuniversity Poles of Attraction (IUAP/PAI PG/14)’’ and the National Research Foundation-Flanders (FWO-Flanders). T. Van Loy, M.B. Van Hiel, H. Verlinden and L. Badisco obtained a Ph.D. fellowship from the “Instituut voor de aanmoediging van Innovatie door Wetenschap en Technologie in Vlaanderen’’ (IWT). J. Poels is a postdoctoral fellow from FWO-Flanders.

References (59)

  • J.D. Baker et al.

    Mutations in the Drosophila glycoprotein hormone receptor, rickets, eliminate neuropeptide-induced tanning and selectively block a stereotyped behavioral program

    J. Exp. Biol.

    (2002)
  • C.I. Bargmann

    Neurobiology of the Caenorhabditis elegans genome

    Science

    (1998)
  • R. Baumeister et al.

    Endocrine signaling in Caenorhabditis elegans controls stress response and longevity

    J. Endocrinol.

    (2006)
  • T.K. Bjarnadottir et al.

    Comprehensive repertoire and phylogenetic analysis of the G protein-coupled receptors in human and mouse

    Genomics

    (2006)
  • T. Braun et al.

    Amino-terminal leucine-rich repeats in gonadotropin receptors determine hormone selectivity

    EMBO J.

    (1991)
  • T. Brody et al.

    Drosophila melanogaster G protein-coupled receptors

    J. Cell Biol.

    (2000)
  • R.D. Burke et al.

    A genomic view of the sea urchin nervous system

    Dev. Biol.

    (2006)
  • I. Claeys et al.

    Insect neuropeptide and peptide hormone receptors: current knowledge and future directions

    Vitam. Horm.

    (2005)
  • I. Claeys et al.

    Insulin-related peptides and their conserved signal transduction pathway

    Peptides

    (2002)
  • S. Costagliola et al.

    Specificity and promiscuity of gonadotropin receptors

    Reproduction

    (2005)
  • E.M. Dewey et al.

    Identification of the gene encoding bursicon, an insect neuropeptide responsible for cuticle sclerotization and wing spreading

    Curr. Biol.

    (2004)
  • K.K. Eriksen et al.

    Molecular cloning, genomic organization, developmental regulation, and a knock-out mutant of a novel leu-rich repeats-containing G protein-coupled receptor (DLGR-2) from Drosophila melanogaster

    Genome Res.

    (2000)
  • G. Fraenkel et al.

    Hormonal and nervous control of tanning in the fly

    Science

    (1962)
  • G. Fraenkel et al.

    Properties of bursicon: an insect protein hormone that controls cuticular tanning

    Science

    (1966)
  • A.R. Frand et al.

    Functional genomic analysis of C. elegans molting

    PLoS. Biol.

    (2005)
  • M. Grossmann et al.

    Novel insights into the molecular mechanisms of human thyrotropin action: structural, physiological, and therapeutic implications for the glycoprotein hormone family

    Endocr. Rev.

    (1997)
  • F. Hauser et al.

    A review of neurohormone GPCRs present in the fruitfly Drosophila melanogaster and the honey bee Apis mellifera

    Prog. Neurobiol.

    (2006)
  • F. Hauser et al.

    Molecular cloning, genomic organization, and developmental regulation of a novel receptor from Drosophila melanogaster structurally related to members of the thyroid-stimulating hormone, follicle-stimulating hormone, luteinizing hormone/choriogonadotropin receptor family from mammals

    J. Biol. Chem.

    (1997)
  • A. Herpin et al.

    Molecular characterization of a new leucine-rich repeat-containing G protein-coupled receptor from a bivalve mollusc: evolutionary implications

    Biochem. Biophys. Acta

    (2004)
  • R.S. Hewes et al.

    Neuropeptides and neuropeptide receptors in the Drosophila melanogaster genome

    Genome Res.

    (2001)
  • C.A. Hill et al.

    G protein-coupled receptors in Anopheles gambiae

    Science

    (2002)
  • T. Hiro’oka et al.

    Disulfide bond mutations in follicle-stimulating hormone result in uncoupling of biological activity from intracellular behavior

    Endocrinology

    (2000)
  • S.Y. Hsu et al.

    The three subfamilies of leucine-rich repeat-containing G protein-coupled receptors (LGR): identification of LGR6 and LGR7 and the signaling mechanism for LGR7

    Mol. Endocrinol.

    (2000)
  • S.Y. Hsu et al.

    Characterization of two LGR genes homologous to gonadotropin and thyrotropin receptors with extracellular leucine-rich repeats and a G protein-coupled, seven-transmembrane region

    Mol. Endocrinol.

    (1998)
  • S.Y. Hsu et al.

    Evolution of glycoprotein hormone subunit genes in bilateral metazoa: identification of two novel human glycoprotein hormone subunit family genes, GPA2 and GPB5

    Mol. Endocrinol.

    (2002)
  • S.Y. Hsu et al.

    Activation of orphan receptors by the hormone relaxin

    Science

    (2002)
  • A.B. Hummon et al.

    From the genome to the proteome: uncovering peptides in the Apis brain

    Science

    (2006)
  • Joost, P., Methner, A., 2002. Phylogenetic analysis of 277 human G-protein-coupled receptors as a tool for the...
  • A.V. Kajava

    Structural diversity of leucine-rich repeat proteins

    J. Mol. Biol.

    (1998)
  • Cited by (61)

    • Transcriptome analysis uncover differential regulation in cell cycle, immunity, and metabolism in Anopheles albimanus during immune priming with Plasmodium berghei

      2021, Developmental and Comparative Immunology
      Citation Excerpt :

      Direct participation of LGR5 maintains WNT signaling on stem cells (Barker et al., 2013). These proteins have key structural elements as several leucine-rich repeats (LRR) amino acid motifs distributed in tandem (Van Loy et al., 2008). In Drosophila and Anopheles gambiae some sequences have been identified as paralogue sequences of LGRs, including LGR5 (Hill et al., 2002).

    • Potential GnRH and steroidogenesis pathways in the scallop Patinopecten yessoensis

      2020, Journal of Steroid Biochemistry and Molecular Biology
    View all citing articles on Scopus
    View full text