Skip to main content

REVIEW article

Front. Endocrinol., 12 November 2018
Sec. Experimental Endocrinology
This article is part of the Research Topic Progress in Reproductive Neuroendocrinology in Vertebrates View all 7 articles

How to Contribute to the Progress of Neuroendocrinology: Discovery of GnIH and Progress of GnIH Research

  • Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo, Japan

It is essential to discover novel neuropeptides that regulate the functions of pituitary, brain and peripheral secretory glands for the progress of neuroendocrinology. Gonadotropin-releasing hormone (GnRH), a hypothalamic neuropeptide stimulating gonadotropin release was isolated and its structure was determined by Schally's and Guillemin's groups at the beginning of the 1970s. It was subsequently shown that GnRH is highly conserved among vertebrates. GnRH was assumed the sole hypothalamic neuropeptide that regulates gonadotropin release in vertebrates based on extensive studies of GnRH over the following three decades. However, in 2000, Tsutsui's group isolated and determined the structure of a novel hypothalamic neuropeptide, which inhibits gonadotropin release, in quail, an avian species, and named it gonadotropin-inhibitory hormone (GnIH). Following studies by Tsutsui's group demonstrated that GnIH is highly conserved among vertebrates, from humans to agnathans, and acts as a key neuropeptide inhibiting reproduction. Intensive research on GnIH demonstrated that GnIH inhibits gonadotropin synthesis and release by acting on gonadotropes and GnRH neurons via GPR147 in birds and mammals. Fish GnIH also regulates gonadotropin release according to its reproductive condition, indicating the conserved role of GnIH in the regulation of the hypothalamic-pituitary-gonadal (HPG) axis in vertebrates. Therefore, we can now say that GnRH is not the only hypothalamic neuropeptide controlling vertebrate reproduction. In addition, recent studies by Tsutsui's group demonstrated that GnIH acts in the brain to regulate behaviors, including reproductive behavior. The 18 years of GnIH research with leading laboratories in the world have significantly advanced our knowledge of the neuroendocrine control mechanism of reproductive physiology and behavior as well as interactions of the HPG, hypothalamic-pituitary-adrenal and hypothalamic-pituitary-thyroid axes. This review describes how GnIH was discovered and GnIH research progressed in this new research era of reproductive neuroendocrinology.

Introduction

A newresearch field in endocrinology was created by the discovery of neurosecretion, which was named neuroendocrinology. In 1928, Scharrer proposed the concept of neurosecretion that consider hypothalamic neurons terminating in the neurohypophysis produce and secrete neurohormones to regulate endocrine organs. In 1949, Bargmann established this seminal concept proposed by Scharrer. Subsequently, two important hypothalamic neuropeptides, i.e., oxytocin (1) and vasopressin (2), were identified as neurohormones secreted from the neurohypophysis in mammals.

Harris (3) hypothesized that hypothalamic neurons that terminate at the median eminence (ME) may also secrete neurohormones from the ME into the hypophysial portal system to regulate the secretion of anterior pituitary hormones, such as thyroid stimulating hormone (TSH), gonadotropins, i.e., luteinizing hormone (LH) and follicle-stimulating hormone (FSH), growth hormone (GH) and adrenocorticotropic hormone (ACTH). Subsequently, Schally's and Guillemin's groups confirmed this seminal hypothesis by the discovery of important neurohormones, including thyrotropin-releasing hormone (TRH) (4, 5), gonadotropin-releasing hormone (GnRH) (6, 7) and growth hormone-inhibiting hormone (somatostatin) (8), in the brain of mammals. Thus, Schally and Guillemin contributed significantly to the advancement of neuroendocrinology by the discoveries of these neurohormones and they were awarded a Nobel Prize in 1977.

As mentioned above, Schally's (7) and Guillemin's (6) groups discovered a hypothalamic neuropeptide stimulating the release of gonadotropins, i.e., LH and FSH, from the anterior pituitary gland of mammals in the beginning of the 1970s, and the peptide was named GnRH. Subsequent studies demonstrated that GnRH is highly conserved among vertebrates (912). Based on extensive studies on GnRH over the next three decades after its discovery, we thought that GnRH is the sole hypothalamic neuropeptide controlling gonadotropin release in vertebrates.

However, in 2000, Tsutsui's group isolated and identified the chemical structure of a novel hypothalamic neuropeptide in quail, an avian species, which inhibits gonadotropin release, and named it gonadotropin-inhibitory hormone (GnIH) (13). The discovery of GnIH opened a new research era of reproductive neuroendocrinology from a novel standpoint. Subsequent studies conducted by Tsutsui's group demonstrated that GnIH is highly conserved among vertebrates from agnathans to humans, acting as an important neurohormone that inhibits vertebrate reproduction [for reviews, see (1427)]. Tsutsui's group's recent studies have further shown important functions of GnIH beyond the control of reproduction (28, 29). It now appears that GnIH acts in the brain to regulate behavior, including reproductive behavior by regulating the biosynthesis of neurosteroids, such as neuroestrogen, in the brain (29). Therefore, the following 18 years of GnIH research in collaboration with world's leading laboratories has advanced our understanding of the neuroendocrine control mechanism of reproductive physiology and behavior [for reviews, see 1418, 2024, 26, 31)].

The discovery of GnIH has changed our understanding about reproductive neuroendocrinology in the past 18 years. Herein this review describes the discovery of GnIH and the progress of intensive research on GnIH focusing on its structure, biosynthesis, biological action, mode of action, and its functional significance in this new research era of reproductive neuroendocrinology. This review also provides a broad overview of the unity and diversity of GnIH structure and biological action and molecular evolution of GnIH in vertebrates.

Discovery of GnIH as a Key Regulator of Reproduction

History of the Discovery of GnIH

Tsutsui and colleagues discovered GnIH in the quail brain, while searching a novel hypothalamic neuropeptide that has a C-terminal Arg-Phe-NH2 motif (RFamide peptide) (13). Price and Greenberg (32) first identified an RFamide peptide that has a cardioexcitatory effect from the ganglia of the venus clam in the late 1970s. The structure of the isolated peptide was Phe-Met-Arg-Phe-NH2 (FMRFamide). Various RFamide peptides that act as neurotransmitters, neuromodulators and hormones had been isolated in other invertebrates after this initial discovery. It was found that FMRFamide-immunoreactive (-ir) neurons were terminating near the anterior pituitary gland in vertebrates (33, 34). Therefore, the existence of unknown hypothalamic RFamide peptides regulating the secretion of anterior pituitary hormones was suggested. This is why Tsutsui's group investigated the existence of an RFamide peptide in the quail brain.

In 2000, Tsutsui and colleagues successfully isolated a novel neuropeptide having a C-terminal RFamide motif, Ser-Ile-Lys-Pro-Ser-Ala-Tyr-Leu-Pro-Leu-Arg-Phe-NH2 (SIKPSAYLPLRFamide) from the extract of quail brains, using an antibody against RFamide for a competitive enzyme-linked immunosorbent assay on high-performance liquid chromatography (HPLC) fractions (13) (Figure 1A). Importantly, this isolated new RFamide peptide inhibited gonadotropin release from the anterior pituitary of quail actively in vitro, which was the first demonstration of a hypothalamic neuropeptide inhibiting gonadotropin release in any vertebrate (13). Given its action on gonadotropin release and its localization in the hypothalamic-hypophysial system, this novel neuropeptide was named GnIH (13) (Figure 2). Cell bodies and terminals for GnIH neurons are located in the paraventricular nucleus (PVN) and ME, respectively, in birds (13). The C-terminal structure of quail GnIH is identical to the chicken LPLRFamide peptide that was reported to be the first RFamide peptide isolated in vertebrates (38), although the chicken LPLRFamide peptide can be a fragment of the chicken GnIH peptide which was found to have a sequence of SIRPSAYLPLRFamide in a recent study (39).

FIGURE 1
www.frontiersin.org

Figure 1. Multiple sequence alignment and phylogenetic analysis of chordate GnIH (LPXRFa) and NPFF (PQRFa) precursor proteins as well as synteny analysis of human GnIH and NPFF genes. (A) Multiple sequence alignment of vertebrate GnIH (LPXRFa) and NPFF (PQRFa) precursor proteins highlighting the sequences of identified and predicted biologically active peptides. Precursor protein sequences were aligned by EMBL-EBI Clustal Omega Multiple Sequence Alignment software. Biochemically identified mature peptide sequences are shown in bold. Adapted from Ubuka and Tsutsui (35). (B) Phylogenetic tree of chordate GnIH (LPXRFa) and NPFF (PQRFa) precursor proteins. The evolutionary history was inferred using the Neighbor-Joining method. The percentage of replicate trees in which the associated taxa clustered together in the bootstrap test (1,000 replicates) are shown next to the branches. The tree is drawn to scale, with branch lengths in the same units as those of the evolutionary distances used to infer the phylogenetic tree. The evolutionary distances were computed using the Poisson correction method and are in the units of the number of amino acid substitutions per site. Evolutionary analyses were conducted in MEGA7 (36). Accession numbers are human (Homo sapiens) GnIH precursor (Human GnIH; NP_071433.3), Japanese quail (Coturnix japonica) GnIH precursor (Quail GnIH; XP_015709159.1), Japanese fire belly newt (Cynops pyrrhogaster) GnIH precursor (Newt LPXRFamide peptide; BAJ78290.1), West Indian Ocean coelacanth (Latimeria chalumnae) GnIH precursor (Coelacanth LPXRFamide peptide; XP_005993154.1), zebrafish (Danio rerio) GnIH precursor (Zebrafish LPXRFamide peptide, NP_001076418.1), spotted gar (Lepisosteus oculatus) GnIH precursor (Gar LPXRFamide peptide; XP_015213317.1), sea lamprey (Petromyzon marinus) GnIH precursor (Petromyzon marinus LPXRFamide peptide; BAL52329.1), Japanese amphioxus (Branchiostoma japonicum) GnIH precursor (Branchiostoma japonicum RFamide peptide; BAO77760.1), human NPFF precursor isoform 1 (Human NPFF isoform 1; NP_003708.1), human NPFF precursor isoform 2 (Human NPFF isoform 2; NP_001307225.1), Japanese quail NPFF precursor (Quail NPFF; XP_015705838.1), Western painted turtle (Chrysemys picta bellii) NPFF precursor (Turtle NPFF; XP_005307776.1), zebrafish NPFF precursor (Zebrafish NPFF; BAF34891.1), spotted gar NPFF precursor isoform X2 (Gar NPFF isoform 2; XP_015199730.1), sea lamprey NPFF precursor (Petromyzon marinus PQRFamide peptide; BAE79779.1), Florida lancelet (Branchiostoma floridae) RFamide precursor 1 (Branchiostoma floridae RFamide peptide 1; XP_002599251.1), Florida lancelet RFamide precursor 2 (Branchiostoma floridae RFamide peptide 2; XP_002609543.1). Fruit fly (Drosophila melanogaster) FMRFamide precursor (Fruit fly FMRFamide; NP_523669.2) served as outgroup (root) of the evolutionary tree. (C) Synteny analysis of human GnIH and NPFF genes. Paralogous genes are linked by dotted lines. Adapted from Osugi et al. (37).

FIGURE 2
www.frontiersin.org

Figure 2. GnIH actions and regulation of GnIH biosynthesis by environmental and internal factors. Cell bodies for GnIH neurons are located in the hypothalamus, paraventricular nucleus in birds and the dorsomedial hypothalamic area in mammals. GnIH neuronal terminals are located to the median eminence (ME) and GnRH1 neurons in the preoptic area in birds and mammals. GnIH receptor is expressed in gonadotropes in the pituitary and GnRH1 neurons in birds and mammals. Thus, GnIH inhibits gonadotropin synthesis and release by directly acting on gonadotropes in the pituitary and by inhibiting the activity of GnRH1 neurons via GnIH receptor in birds and mammals. GnIH neurons project not only to GnRH1 neurons but also to kisspeptin neurons in the hypothalamus in mammals. Kisspeptin neurons express GnIH receptor. GnIH and GnIH receptor are expressed in steroidogenic cells and germ cells in gonads, and GnIH acts in an autocrine/paracrine manner to suppress sex steroid production and germ cell differentiation and maturation in birds and mammals. GnIH participates not only in neuroendocrine functions but also in the control of behavior in birds and mammals. GnIH inhibits reproductive behaviors, such as sexual and aggressive behaviors, by acting within the brain. Furthermore, GnIH inhibits reproductive behaviors by stimulating the biosynthesis of neuroestrogen (E2) in the POA. GnIH neurons further project to many other neurons in the brain suggesting multiple actions of GnIH. Environmental factors, such as photoperiod, stress and social interaction, and internal factors, such as melatonin, glucocorticoid and norepinephrine, are important for the control of reproduction and reproductive behaviors. GnIH expression and release are modulated via a melatonin-dependent process. Melatonin increases GnIH expression in quail and rats, but melatonin decreases GnIH expression in hamsters and sheep. Stress increases GnIH expression by the actions of glucocorticoids in birds and mammals. Thus, GnIH is a mediator of stress-induced reproductive disruption. The social environment also changes GnIH expression and release mediated by the action of norepinephrine. Stimulatory regulations are shown by arrows, whereas inhibitory regulations are shown by blunt end lines. Lines with a question mark indicate morphological evidence without demonstration of physiological actions. ME, median eminence; POMC, proopiomelanocortin; MCH, melanin-concentrating hormone; CRH, corticotropin-releasing hormone.

After the discovery of GnIH peptide, GnIH precursor cDNA was sequenced in quail (40) and in other avian species, chicken, sparrows, zebra finches and starlings [for reviews, see (15, 17, 18, 2123)]. The GnIH precursor cDNA encodes GnIH and GnIH-related peptides GnIH-RP-1 and GnIH-RP-2, which possess a common LPXRFamide (X = L or Q) motif in the C-terminal in all avian species investigated (Figure 1A). GnIH was also isolated as an endogenous mature peptide in starlings (41) and zebra finches (42) and mature GnIH-RP-2 was also isolated in quail (40) (Figure 1A).

GnIH is considered to be a key neuropeptide that inhibits avian reproduction as GnIH inhibits gonadotropin release in avian species [for reviews, see (15, 17, 18, 2123)]. To investigate the biological action of GnIH in vivo, mature male quail were treated with GnIH chronically (43). Chronic treatment with GnIH decreases the expressions of the common α, LHβ and FSHβ subunit mRNAs and the circulating LH concentration. In addition, chronic GnIH treatment induces apoptosis of testicular cells and decreases seminiferous tubules' size in mature male birds (43). In immature birds, GnIH treatment suppresses growth of the testis (43). These findings indicate that GnIH suppresses the development and maintenance of gonads by decreasing synthesis and release of gonadotropin in birds (Figure 2).

Structure and Biological Action of GnIH in Vertebrates

To determine if findings in avian species extend to mammals including primates, Tsutsui and colleagues identified GnIH in the mammalian and primate hypothalamus (4448). All the identified and predicted mammalian and primate GnIHs possess a common C-terminal LPXRFamide (X = L or Q) motif, like avian GnIH and GnIH-RPs [for reviews, see (1518, 2123)] (Figure 1A). Therefore, GnIHs identified in birds, mammals and primates were designated as LPXRFamide peptides based on their structures. On the other hand, mammalian and primate GnIHs are also called RFamide-related peptide 1 and 3 (RFRP-1 and−3) (Figure 1A). Multiple sequence alignment of vertebrate LPXRFamide precursor proteins shows that human RFRP-1 aligns with quail GnIH-RP-1. Human RFRP-3 aligns with a GnIH-like peptide (APNLSNRSa) of quail. On the other hand, quail GnIH aligns with an RFRP-like peptide (SAGATANLPLRSa) of human (Figure 1A). Importantly, it was clearly shown that in vivo administration of avian GnIH to female Syrian hamsters centrally or peripherally inhibits LH release (44). Administration of hamster GnIHs (RFRP-1 and−3) centrally inhibits LH release in Siberian hamsters (45). It was also shown that central administration of rat GnIH (RFRP-3) to male rats inhibits LH release (49) and GnRH-elicited gonadotropin release (50, 51). In sheep (52, 53) and cows (54), mammalian GnIH (RFRP-3) administration reduces LH pulse amplitude and inhibits GnRH-elicited gonadotropin synthesis and release. The structure of human GnIH (RFRP-3) was found to be the same as that of ovine GnIH (RFRP-3) (47). Therefore, the biological action of human GnIH (RFRP-3) was examined in the ovine pituitary with Clarke's and Bentley's groups. Importantly, human/ovine GnIH (RFRP-3) clearly inhibits the release of both LH and FSH stimulated by GnRH (52). Thus, it was established that mammalian and primate GnIHs inhibit the synthesis and release of gonadotropin and GnRH-elicited gonadotropin release [for reviews, see (1418, 2123)] (Figure 2).

Tsutsui and colleagues identified GnIHs further in reptiles, amphibians and fish to place GnIH findings into a broader perspective in vertebrates. The putative and identified GnIHs in these vertebrate species also possess a C-terminal LPXRFamide (X = L or Q) motif as GnIHs of birds, mammals and primate (5562) (Figure 1A). It thus appears that GnIHs exist in vertebrates from fish to humans [see (1525, 27, 30)] for reviews). In fish, Sawada et al. (59) reported that GnIH precursor cDNA encodes three GnIHs, gfLPXRFa-1,−2 and−3 in goldfish. Subsequently, several studies in fish found that goldfish GnIHs have both stimulatory and inhibitory effects on gonadotropin synthesis and release, which may depend on reproductive conditions (6367). Zhang et al. (67) also showed that GnIH of zebrafish, zfLPXRF-3, suppresses plasma LH levels in goldfish.

Most of the studies in mammals, birds, and fish showed the inhibitory effect of GnIH in the HPG axis. However, several studies in mammals and many studies in fish have shown its stimulatory effect (68). GnIH inhibits LH release in the breeding season when their endogenous LH level is high but stimulates LH release in non-breeding season when their LH level is basal in Siberian hamsters (45). Inhibitory or stimulatory effects of GnIH on the HPG axis depends on the reproductive stages in fish (69). In experiments using higher concentration or longer duration of GnIH administration can stimulate the HPG axis (68). It is considered that the action of GnIH in the HPG axis is modulated by sex-steroid concentration, the action of neuroestrogen synthesized by the activity of aromatase in the brain, estrogen membrane receptor, heteromerization and internalization of GnIH, GnRH, and estrogen membrane receptors (68). The dual action of GnIH in the HPG axis may have a physiological role for reproductive homeostasis according to developmental and reproductive stages of the animal (68).

Although extensive studies have demonstrated that GnIHs are present in the brain of representative species of gnathostomes, the presence of GnIH had not been identified in agnathans, the most ancient lineage of vertebrates (70). Tsutsui and colleagues therefore searched for agnathan GnIH in collaboration with Sower's and Nozaki's groups (71). Because synteny analysis showed the existence of GnIH gene in sea lamprey, Osugi et al. (71) cloned lamprey GnIH precursor cDNA that encodes three GnIHs. Subsequently, these mature GnIHs were isolated from the brain of sea lamprey by immunoaffinity purification and mass spectrometry (71). The isolated lamprey GnIHs possess a common C-terminal PQRFamide motif (71), unlike GnIHs isolated in gnathostomes (Figure 1A).

GnIH neurons exist in the lamprey hypothalamus (71) with GnIH-ir fibers extending to GnRH3 neurons (71). On the other hand, few lamprey GnIH-ir fibers exist in the neurohypophysis compared with lamprey GnRH3-ir fibers (71). Based on the morphology of hypothalamic GnIH neurons, Osugi et al. (71) then analyzed the biological action of lamprey GnIHs on lamprey GnRHs and gonadotropin β subunit expressions and found that lamprey GnIH increases lamprey GnRH3 and gonadotropin β subunit expressions (71). These findings indicate that GnIH exists in the brain of lamprey, which is the oldest lineage of vertebrates and GnIH stimulates the expression of gonadotropin β in the pituitary by acting on GnRH3 neurons (71). Accordingly, GnIH peptide may have emerged as a stimulatory neuropeptide for the regulation of gonadotropin secretion in agnathans and changed its function into an inhibitory neuropeptide during evolution of vertebrates.

Molecular Evolution of GnIH in Vertebrates

As described above, most GnIHs are LPXRFamide (X = L or Q) peptides that belong to the RFamide peptide family [for reviews, see (1525, 27, 30)]. In vertebrates, four more groups, i.e., the prolactin-releasing peptide (PrRP) group, the pyroglutamylated RFamide peptide (QRFP)/26RFamide group, the kisspeptin group, and the neuropeptide FF (NPFF; PQRFamide peptide) group, have been recognized [for reviews, see (15, 16, 25, 27)]. Because the C-terminal structure of NPFF peptides have a C-terminal PQRP motif in vertebrates, which is similar to that of GnIH peptides, the NPFF peptide gene in agnathans was needed to be clarified. Tsutsui and colleagues therefore identified the cDNAs of NPFF precursor in the brain of lamprey and hagfish (72, 73). Importantly, phylogenetic analysis showed that agnathans possess both GnIH and NPFF genes (Figure 1A). Subsequently, agnathan NPFF peptides were identified in sea lamprey and hagfish. The identified agnathan NPFF peptides and GnIH peptides had the same C-ternimal PQRFamide motif (7173).

Because agnathans have both GnIH and NPFF genes and their mature peptides have the same C-terminal PQRFamide motif (7173), it was strongly suggested that the GnIH and NPFF genes were derived from a common ancestral gene in protochordates. To investigate this possibility, Tsutsui and colleagues further identified an amphioxus PQRFamide peptide precursor cDNA that encodes three putative PQRFamide peptides (37). Subsequently, the mature peptides of amphioxus PQRFamide were identified by immunoaffinity chromatography and mass spectrometry (37). The amphioxus PQRFamide peptide precursor was suggested to have occurred before the divergence of GnIH and NPFF groups in vertebrates by phylogenetic analysis (37) (Figure 1B). It was found that the amphioxus PQRFamide peptide gene, GnIH gene and NPFF gene of vertebrates have a conserved synteny region around the loci of the genes (37). Vertebrate GnIH and NPFF genes exist near the HOXA and HOXC clusters (Figure 1C), respectively, whereas the amphioxus PQRFamide peptide gene exists near the HOX cluster, suggesting that GnIH and NPFF genes have separated through whole-genome duplication event (37). Based on these findings, it is considered that the amphioxus PQRFamide peptide gene is close to the ancestor of the GnIH and NPFF genes (37, 74). Accordingly, the GnIH and NPFF genes may have separated by whole-genome duplication from an ancestral gene conserved in the protochordate during vertebrate evolution (Figures 1A–C).

Progression of GnIH Research Focusing on the Molecular Mechanism of Action of GnIH on Gonadotropin Secretion

Discovery of the Receptor for GnIH

Tsutsui and colleagues identified the receptor for GnIH in quail to clarify the mode of GnIH action on gonadotropin secretion. The identified GnIH receptor was the G-protein coupled receptor GPR147 (75), which is also named neuropeptide FF receptor 1 (NPFF1). Yin et al. (75) showed that GnIH and GnIH-RPs bind the membrane fraction of COS-7 cells transfected with GnIH receptor cDNA with high affinities. GnIH can act directly on gonadotropes to reduce gonadotropin release in birds, because GnIH receptor is expressed in gonadotropes in the anterior pituitary [for reviews, see (1418, 2123, 31)] (Figure 2). In addition, GnIH neurons project to GnRH1 neurons that also express GnIH receptor (76, 41) (Figure 2). Accordingly, it is considered that GnIH acts not only on gonadotropes but also on GnRH1 neurons to inhibit gonadotropin synthesis and release in avian species [for reviews, see (1418, 2123, 31)] (Figure 2).

In mammals, Hinuma et al. (77) identified a specific receptor for mammalian GnIH, which is identical to GPR147 and named it OT7T022. Bonini et al. (78) found two GPCRs for NPFF, NPFF1 (identical to GPR147) and NPFF2 (identical to GPR74). GPR147 and GPR74 are paralogous (79). Binding experiments of GnIH and NPFF for GPR147 and GPR74 showed that the affinity of GnIH is higher for GPR147, whereas the agonistic activity of NPFF is potent for GPR74 (78, 80, 81). These findings indicate that the primary receptor for GnIH is GPR147 (NPFF1, OT7T022).

Mode of GnIH Action on Target Cells

Tsutsui and colleagues investigated the effect of GnIH in a mouse gonadotrope cell line, LβT2 to further clarify the cell signaling cascade in gonadotropes triggered by GnIH. Son et al. (82) found that LβT2 cells express GnIH receptor mRNA by RT-PCR. Son et al. (82) further demonstrated that GnIH inhibits GnRH-induced signaling pathways as follows: mouse GnIHs reduced cAMP production and phosphorylation of extracellular signal-regulated kinase (ERK) induced by GnRH (82). Importantly, mouse GnIHs reduced GnRH-induced LHβ expression and LH release (82). Inhibitors of adenylate cyclase (AC) and protein kinase A (PKA) suppressed the stimulatory effect of GnRH on gonadotropin expression, but inhibitor of protein kinase C (PKC) did not (82). These findings indicate that mouse GnIH reduces GnRH-stimulated gonadotropin secretion by interfering with GnRH actions via an AC/cAMP/PKA-dependent ERK pathway (82).

Following the discovery of GnIH and its inhibitory action on the hypothalamic-pituitary-gonadal (HPG) axis, kisspeptin was discovered in mammals. In contrast to GnIH, kisspeptin stimulates GnRH neurons and up-regulates the HPG axis in mammals (8386). Because GnIH neurons project not only to GnRH1 neurons in the preoptic area (POA) but also to kisspeptin neurons in the hypothalamus, GnIH neurons may regulate the activities of both GnRH1 neurons and kisspeptin neurons [for reviews, see (1418, 2123, 87)] (Figure 2). Interestingly, GnIH neurons also project to GnRH2 neurons and many other neurons in the brain, which suggest multiple actions of GnIH [for reviews, see (1418, 2123)] (Figure 2).

Progression of GnIH Research Focusing on Multiple Actions of GnIH

Gonadal GnIH Action on Reproduction

Based on extensive GnIH studies, it now appears that GnIH is a key neurohormone for the regulation of reproduction, which reduces gonadotropin synthesis and release by suppressing pituitary gonadotropes and GnRH1 neurons in vertebrates (Figure 2). In addition to the central actions of GnIH, there are reports showing that gonadal GnIH is directly involved in the regulation of gonadal activity locally [for reviews, see (1418, 2123, 31)] (Figure 2). Steroidogenic and germ cells in the gonads express both GnIH and GnIH receptor in birds and mammals (8894). Furthermore, several reports show that GnIH acts in the gonads to suppress sex steroid production and germ cell differentiation and maturation in an autocrine and/or paracrine manner (8894) (Figure 2).

Central GnIH Action on Feeding Behavior

Importantly, central GnIH participates not only in neuroendocrine control of reproduction but also in behavioral control. Animals use photoperiod to phase breeding with anticipated times of maximal food availability in the environment where energy availability fluctuates (95). When food becomes scarce during the breeding season, reproduction is temporarily inhibited (96, 97). Metabolic challenges such as food deprivation inhibit reproductive axis functioning and sexual motivation (98102). Tsutsui and colleagues therefore investigated whether GnIH relays metabolic information to the HPG axis and regulates neural feeding circuits [for reviews, see (14, 30)] by the following avian and mammalian studies.

In birds, intracerebroventricular (ICV) injection of GnIH to chicks stimulates food intake (103). ICV injection of anti-GnIH antiserum suppresses fasting induced appetite in support of the stimulatory role of GnIH in feeding (103). Fraley et al. (104) also reported that ICV injection of GnIH decreases plasma LH concentration and increases feeding in adult Pekin ducks. Tachibana et al. (105) further investigated if the orexigenic effect of GnIH involves the opioid and nitric oxide (NO) systems to establish the central mechanism underlying the GnIH action on feeding. The orexigenic effect of ICV injected GnIH is decreased by co-injection of an opioid μ-receptor antagonist β-funaltrexamine, but not by an opioid δ-receptor antagonist ICI-174,864 and an opioid β-receptor antagonist nor-binaltorphimine in chicks (105). In addition, feeding behavior induced by GnIH was not affected by co-injection of a non-selective NO synthase inhibitor (105). McConn et al. (39) also examined the central orexigenic mechanism induced by GnIH in chicks. In the hypothalamus, neuropeptide Y (NPY) mRNA increases, and pro-opiomelanocortin (POMC) mRNA decreases following ICV administration of chicken GnIH (73, Figure 2), McConn et al. (39) further investigated the lateral hypothalamic area (LHA) because ICV injection of GnIH increases c-fos-ir cells in this brain area. They found that melanin-concentrating hormone (MCH) mRNA increases by administration of GnIH (Figure 2). Accordingly, these avian findings indicate that opioid μ-receptor, NPY, POMC and MCH-positive neurons play important roles in the orexigenic response of GnIH (Figure 2).

In mammals, ICV administration of GnIH also increases food intake in rats (49) and sheep (106). Qi et al. (107) showed that GnIH neurons project to NPY, POMC, orexin, and MCH neurons that regulate feeding behavior (Figure 2). Fu and van den Pol (108) showed that chicken and human GnIHs both inhibit POMC neurons and suppress excitation of kisspeptin cells by a mechanism based on opening potassium channels in mouse brain slices (Figure 2). Jacobi et al. (109) found that GnIH inhibits the firing rate in POMC neurons and has an inhibitory effect on action potential activity in NPY neurons in mice (Figure 2). In addition, Jacobi et al. (109) found that NPY neurons have close contacts from GnIH fibers (Figure 2). Thus, these mammalian findings indicate that GnIH participates not only in the regulation of reproduction but also in the regulation of feeding behavior in mammals as in birds.

Central GnIH Action on Reproductive Behaviors

It now appears that central GnIH regulates reproductive behaviors, such as sexual and aggressive behaviors by acting in the brain (29, 110, 111) (Figure 2). In birds, Bentley et al. (110) found that central administration of GnIH inhibits copulation solicitation of female white-crowned sparrows stimulated by the song of males. GnRH2 stimulates copulation solicitation in female white-crowned sparrows stimulated by the song of males (112). As GnIH neurons extend to GnRH2 neurons and GnIH receptor is expressed in GnRH2 neurons in songbirds (41), GnIH may suppress copulation solicitation by suppressing the activity of GnRH2 neurons in songbirds (110) (Figure 2). Ubuka et al. (111) therefore investigated this hypothesis by examining the behavior of male and female white-crowned sparrows modified by RNA interference (RNAi) to the GnIH gene with Wingfield' group. It was found that GnIH RNAi reduces resting time and spontaneous production of complex vocalizations, but stimulates agonistic vocalizations. Furthermore, GnIH RNAi increases song production in male birds challenged by novel male song playbacks (111). These findings indicate that GnIH RNAi induces arousal. Ubuka et al. (111) further found that GnIH mRNA expression in the PVN is negatively correlated with the activity of male and female birds. Importantly, GnIH RNAi decreases the density of GnIH neuronal fibers in the ventral tegmental area in female birds, and the number of GnRH1 and GnRH2 neurons with close GnIH neuronal fiber appositions is negatively correlated with the activity of male birds (111) (Figure 2). Ubuka et al. (29) further demonstrated that GnIH suppresses aggressive behavior in male quail. Accordingly, GnIH may suppress both sexual and aggressive behaviors in birds [see (14, 22, 30, 31) for reviews] (Figure 2).

In mammals, Johnson et al. (49) also found that ICV administration of GnIH inhibits male sexual behavior in rats. Piekarski et al. (113) reported that ICV administration of GnIH reduces sexual motivation and vaginal scent marking but does not suppress lordosis behavior in female hamsters. GnIH administration alters fos expression in the medial POA, medial amygdala and bed nucleus of the stria terminalis, key neural loci implicated in female sexual behavior (113). These mammalian findings indicate that GnIH is an important modulator of female proceptive sexual behavior and motivation (Figure 2). GnIH neurons also project to dopamine neurons in the rhesus macaque brain [(46), Figure 2). Accordingly, GnIH not only acts to regulate the HPG axis but also act to drive the neural circuitry underlying socially-motivated behavior in mammals, as in birds.

Central GnIH Action on Neurosteroid Biosynthesis

There are several reports indicating that neuropeptides and neurosteroids interaction plays an important role in the regulation of brain functions [for a review, see (114)]. Recently, Ubuka et al. (29) found that GnIH increases neuroestrogen synthesis by stimulating the activity of cytochrome P450 aromatase (P450arom) in the quail brain (29) (Figure 2). Importantly, the action of GnIH on the stimulation of neuroestrogen synthesis changes the expression of aggressive behavior in this bird (29) (Figure 2). These results provide a new concept of GnIH that regulates aggressive behavior by modifying the neurosteroid milieu in the brain.

It is established that sexually mature male quail actively fight with intense aggressiveness, unlike female quails (115, 116). Aggressive behavior of male quail is testicular androgen dependent (115117). However, generally no correlation is observed between aggressiveness and circulating testosterone (T) concentration (117). It is also established that aromatizable androgens, such as T and androstenedione (AD) facilitate aggression in males, but non-aromatizable androgens, such as dihydrotestosterone (DHT) do not, and that T-induced aggression is blocked by administration of P450arom inhibitors (118, 117). Based on these findings, testicular androgen action on aggressive behavior requires aromatization into estrogen (neuroestrogen) in the brain (119, 120, 121).

GnIH neurons project to brain areas, such as the POA (41, 122, 123) and the periaqueductal central gray [PAG; (41)] in birds. GnIH receptor is expressed in the POA (41, 75) and PAG (41). It is known that these brain areas regulate aggressive behavior (124, 125). The POA is also known to be the most critical site of aromatization of testicular androgen by P450arom, and neuroestrogen directly regulates aggressive behavior in male quail (126, 127). Because GnIH decreases aggressive behavior in male birds as described above (29, 111), Ubuka et al. (29) hypothesized that GnIH may decrease aggressive behavior by regulating P450arom activity and neuroestrogen synthesis in the brain (Figure 2). Therefore, Ubuka et al. (29) examined whether GnIH-ir neuronal fibers innervate P450arom cells and P450arom-ir cells express GnIH receptor in the POA in male quail. Ubuka et al. (29) found that abundant GnIH-ir neuronal fibers exist in the vicinity of P450arom-ir cells in the POA and GnIH receptor is expressed in P450arom-ir cells in the POA. They (29) further found that GnIH stimulates the activity of P450arom and increases neuroestrogen concentration in the POA through GnIH receptor (29) (Figure 2). This is the first evidence that GnIH, a hypothalamic neuropeptide, decreases aggressive behavior by stimulating neuroestrogen synthesis in the brain. Subsequently, the effect of centrally administered various doses of estradiol-17β (E2) was tested on the aggressive behavior of male quail. They (29) found that centrally administered higher doses of E2 decreases aggressive behavior (29). This finding indicates that production of neuroestrogen in the brain is essential for the expression of aggressive behavior, but high concentrations of neuroestrogen in the brain decrease aggressive behavior. Accordingly, it is considered that GnIH suppresses aggressive behavior by increasing neuroestrogen synthesis beyond its optimum concentration for the expression of aggressive behavior through activation of P450arom in the brain of male birds (29) (Figure 2).

Ubuka et al. (29) further investigated how GnIH stimulates P450arom activity. P450arom activity is not only controlled by P450arom gene Cyp19 transcription by steroids in the long term, but also by phosphorylation of P450arom stimulated by neurotransmitters, such as glutamate in the short term (128). Balthazart's group reported that P450arom activity is rapidly down-regulated by phosphorylation in the hypothalamus of male quail (128132). It is therefore possible that GnIH activates P450arom by dephosphorylation of phosphorylated P450arom. Importantly, Ubuka et al. (29) showed that ICV administration of GnIH decreases phosphorylated P450arom in the POA in the short term, and that the action of GnIH on neuroestrogen synthesis in the POA is abolished by concomitant administration of RF9, a potent antagonist of GnIH receptor (133, 134) or fadrozole, an inhibitor of P450arom (135, 136). These findings indicate that GnIH stimulates neuroestrogen synthesis in the POA by activating P450arom through dephosphorylation after binding GnIH receptor in P450arom cells.

Progression of GnIH Research Focusing on the Regulation of GnIH Biosynthesis by Environmental and Internal Factors

Melatonin Regulation of GnIH Biosynthesis Under Photoperiodic Condition

Clarification of the mechanisms regulating GnIH expression in the brain is important for the understanding of the physiological role of GnIH in reproduction. Photoperiodic mammals regulate their reproductive activities depending on the annual cycle of changes in the duration of nocturnal secretion of melatonin (95). There is also evidence that melatonin contributes to the regulation of seasonal changes in gonadotropin secretion and gonadal activity in birds (137140), despite there is an accepted dogma that reproductive activity of birds is not regulated by seasonal changes in melatonin secretion (141, 142).

Based on this background, Tsutsui and colleagues clarified the action of melatonin on the regulation of GnIH expression in quail, a highly photoperiodic avian species. Ubuka et al. (143) found that removal of melatonin by pinealectomy (Px) and orbital enucleation, minimizes GnIH mRNA and GnIH peptide expressions in the brain of quail (143). By contrast, melatonin administration increases the expressions of GnIH mRNA and GnIH peptide in the quail brain (143). Importantly, a melatonin receptor subtype Mel1c is expressed in GnIH neurons in quail (143), indicating that melatonin induces GnIH expression by acting directly on GnIH neurons (Figure 2). Chowdhury et al. (144) further found that melatonin increases not only GnIH expression but also its release in quail (Figure 2). Interestingly, GnIH release is negatively correlated with plasma LH concentration with diurnal changes (144). In quail, GnIH release is increased in short day (SD) photoperiods, when the duration of nocturnal secretion of melatonin increases (144). Accordingly, it appears that melatonin derived from the pineal gland and eyes acts on GnIH neurons directly via Mel1c to induce GnIH expression and release in birds (22, 23, 143, 144) (Figure 2).

In contrast to quail, melatonin decreases GnIH expression in Syrian and Siberian hamsters, both photoperiodic mammals (45, 145, 146) (Figure 2). GnIH expression is reduced in sexually quiescent hamsters exposed to SD photoperiods, compared with sexually active animals under long day (LD) photoperiods. Importantly, these photoperiodic changes in GnIH expression are abolished in Px hamsters and melatonin injections to LD hamsters decrease GnIH expression to SD level (45, 146). Similar seasonal patterns of GnIH expression have been also observed in European (147) and Turkish (148) hamsters, and the semi-desert rodent, Jerboa (149). In addition, there are reports showing that GnIH expression is regulated by melatonin in sheep (150, 151) and rats (152). These results indicate that GnIH expression is regulated photoperiodically by a melatonin-dependent process both in mammals and birds, although species differences exist in the regulation of GnIH expression by melatonin [see (1418, 22, 23) for reviews] (Figure 2). Furthermore, there is evidence that gonadal GnIH also responds to melatonin directly in a seasonal manner in songbirds (153, 154).

Glucocorticoid Regulation of GnIH Biosynthesis Under Stress Condition

It is known well that reproduction can be reduced by stress in vertebrates (155). Kirby et al. (156) found that both chronic and acute immobilization stress increases expression of GnIH in the dorsomedial hypothalamic area (DMH) associated with the inhibition of the HPG axis in rats (Figure 2). Adrenalectomy abolishes stress-induced increase in GnIH expression (156). Glucocorticoid receptor (GR) is expressed in GnIH neurons, indicating that adrenal glucocorticoids act directly on GnIH neurons, which may contribute to the increase in GnIH expression (156) (Figure 2). Taken together, these findings imply that GnIH may be an important mediator of stress-induced suppression of reproduction in mammals (156).

Son et al. (157) found that GR is expressed in GnIH neurons in the PVN and corticosterone [CORT; the major glucocorticoid in birds and rodents, (158, 159)] treatment increases GnIH expression, indicating that glucocorticoids can directly regulate GnIH expression in quail (Figure 2).

Furthermore, Son et al. (157) clarified the mechanism of activation of GnIH expression by CORT in rHypoE-23, a GnIH-expressing neuronal cell line, derived from rat hypothalamus. Importantly, GR is expressed in rHypoE-23 cells and CORT treatment increases GnIH expression in these cells (157). It thus appears that stress reduces gonadotropin secretion, at least in part, through the increase in GnIH expression in mammals and birds. Furthermore, there is evidence that gonadal GnIH responds to metabolic challenge as well as stress in a seasonal manner in songbirds (160).

There are also reports suggesting that GnIH stimulates the hypothalamic-pituitary-adrenal (HPA) axis (107, 161, 162). Qi et al. (107) showed that GnIH neurons project to corticotropin-releasing hormone (CRH) and oxytocin neurons in the PVN in sheep (Figure 2). Kaewwongse et al. (161) showed that ICV injection of RFRPs increases plasma concentration of ACTH and oxytocin in rats. On the other hand, Ullah et al. (162) showed that intravenously injected RFRP-1 increases cortisol level in monkeys. These results suggest that GnIH stimulates the HPA axis, although its mechanism of action is not understood well (Figures 2, 3).

FIGURE 3
www.frontiersin.org

Figure 3. The crosstalk of GnIH with internal factors of different endocrine axes. The interaction between the HPA axis and the axis is mediated by GC and GnIH. Stress suppresses gonadotropin secretion through the increase in GnIH expression in mammals and birds. In addition, the interaction between the axis and the axis was demonstrated. TH-mediated HPG regulation is initiated by inhibiting the expression of GnIH, which acts at the most upstream level of the HPG axis by inhibiting the activity of GnRH neurons to reduce circulating levels of gonadotropins (LH and FSH) and gonadal sex steroids. High concentrations of TH decrease GnIH expression, whereas a lower level of TH increases GnIH expression. The increased GnIH expression induced by hypothyroidism delays pubertal onset. Stimulatory regulations are shown by arrows, whereas inhibitory regulations are shown by blunt end lines. Lines with a question mark indicate morphological evidence without demonstration of physiological actions. HPA, hypothalamic-pituitary-adrenal; HPG, hypothalamic-pituitary-gonadal; HPT, hypothalamic-pituitary-thyroid; GC, glucocorticoid; TH, thyroid hormone.

Norepinephrine Regulation of GnIH Biosynthesis Under Social Environment

In addition to photoperiod and stress, social environment may influence GnIH expression (Figure 2). Calisi et al. (163) manipulated nesting opportunities for pairs of European starlings and GnRH, GnIH, and GnIH mRNA contents in the brain were examined. Fewer numbers of GnIH cells were observed in birds with nest boxes than those without nest boxes, but GnRH did not vary with or without nest box. These findings suggest that GnIH is involved in reproductive function in response to social environment (163).

There are reports showing that plasma T concentrations in male quail is rapidly decreased by the presence of a female bird (164, 165). Based on these observations, Tsutsui and colleagues looked into the neurochemical mechanism of social interaction that alters reproductive physiology (Figure 2). Tobari et al. (28) first found that norepinephrine (NE) release rapidly increases in the PVN of male quail when viewing a conspecific female (Figure 2). GnIH expression increases in the PVN of male quail as well, with associated decreases in plasma LH concentrations, when males view a female (28) (Figure 2). Subsequently, Tobari et al. (28) showed that NE application to male quail increases GnIH release. GnIH neurons express α2A-adrenergic receptor and GnIH neurons are innervated by noradrenergic fibers (28). These findings indicate that female presence stimulates GnIH release by the increase in NE release in the PVN, resulting in decreases in circulating LH and T levels in male quail (28) (Figure 2).

Role of GnIH in Hypothyroidism-Induced Delayed Puberty

Thyroid disorder is known to be associated with abnormal pubertal development. However, the mechanism of thyroid hormone (TH) action on pubertal onset remains unclear, although interactions between the hypothalamic-pituitary-thyroid (HPT) and HPG axes have been suggested (166168) (Figure 3). Recently, Tsutsui and colleagues challenged this possibility by testing TH-mediated regulation of the HPG axis will be initiated by the change in the expression of GnIH, which acts at the most upstream level of the HPG axis by inhibiting the activity of GnRH neurons to reduce gonadotropin secretion from the pituitary gonadotropes [for reviews, see (1518, 2123, 44)] (Figure 3). To investigate the possible role of GnIH as a novel mediator between the HPT and HPG axes, Kiyohara et al. (169) investigated if abnormal thyroid status has an effect on pubertal onset and the HPG axis in female mice. Long-term treatment of the female mice with propylthiouracil (PTU) induced hypothyroidism and significant delay in pubertal onset (169). Importantly, hypothalamic GnIH mRNA expression is increased in hypothyroid female mice (169). Furthermore, circulating LH and estradiol-17β (E2) levels decreased in hypothyroid animals (169). It is therefore considered that pubertal onset may be delayed by hypothyroidism through the increase in GnIH expression and the decrease in LH and E2 levels in female mice (169) (Figure 3).

Kiyohara et al. (169) induced hypothyroidism in GnIH-knockout (KO) female mice, in order to demonstrate the involvement of GnIH in hypothyroidism induced pubertal disorder (169). Administration of PTU to GnIH-KO mice induces hypothyroidism with the reduced level of triiodothyronine, T3 (169). However, delayed puberty induced by hypothyroidism observed in wild type (WT) female mice was not observed in PTU-administered GnIH-KO female mice (169). Accordingly, it is considered that GnIH mediates hypothyroid induced delayed pubertal onset (Figure 3).

Molecular studies were further conducted to clarify the regulatory mechanism of GnIH on hypothyroidism-induced delayed puberty, as follows. Firstly, it was found that GnIH neurons in the hypothalamus express TH receptors (TRα and TRβ) (169) so that TH signals can be directly conveyed to GnIH neurons by TRs. Secondly, several putative TH-response elements (TREs) exist within 3 kb upstream from the mouse GnIH ORF. However, both TRs do not directly bind these TREs by chromatin immunoprecipitation (ChIP) assays (169), suggesting that TH (T3) may act via non-genomic action by membrane TRs. Importantly, H3 acetylation (H3Ac) correlated with gene activation is increased in hypothyroid female mice compared to control mice (169). Accordingly, thyroid status may modify chromatin structure of the GnIH promoter region resulting in the change in GnIH gene expression.

Some papers show that the elevated TRH level in hypothyroidism induces hyperprolactinemia and changes GnRH pulsatile secretion, leading to delayed LH response and delayed puberty (167). Other papers report that the increased TSH level activates gonadal function by the stimulation of receptor in gonads that is responsible for precocious puberty, as the structure of and receptors is similar (166, 168). However, the mechanism of how TH acts on the axis was not elucidated, although these papers suggest the presence of a mediator in the interaction of and HPG axes (166168). Kiyohara et al. (169) found that thyroid dysfunction increases GnIH expression in the hypothalamus through chromatin modification of the promoter region of the GnIH gene in female mice. These results propose a novel function of GnIH serving as a mediator between the HPT and HPG axes (Figure 3). Female mice with hypothyroidism show pubertal delay with the increase in GnIH expression. Kiyohara et al. (169) further found that GnIH mediates the effect of hypothyroidism on pubertal delay, because delayed puberty was not observed in hypothyroidism induced GnIH-KO female mice. Accordingly, it is considered that GnIH is a critical factor that mediates abnormal thyroid status effect on pubertal onset [for a review, see (26)] (Figure 3).

Conclusions

GnIH is a novel hypothalamic neuropeptide that inhibits gonadotropin synthesis and release. Studies on GnIH in the past 17 years have demonstrated that GnIH is highly conserved among vertebrates from agnathans to humans, which acts as a key neuropeptide inhibiting reproduction across vertebrates. GnIH inhibits gonadotropin synthesis and release through actions on gonadotropes and GnRH neurons via GPR147, the GnIH receptor. Thus, the discovery of GnIH has markedly advanced the progress of reproductive neuroendocrinology. Recent studies have further indicated that GnIH is involved in pubertal disorder induced by thyroid dysfunction. This is a novel function of GnIH mediating the interaction of the HPT-HPG axes in puberty.

Kisspeptin was discovered in mammals following the discovery of GnIH. GnIH and kisspeptin are new members of the RFamide peptide family, which act on the HPG axis to down-regulate and up-regulate the reproductive system, respectively. Therefore, we can now say that GnRH is not the sole hypothalamic neuropeptide that regulates reproduction. In the hypothalamus, GnIH neurons also projects to kisspeptin neurons. Importantly, GnRH and kisspeptin neurons both express GnIH receptor. Further studies may reveal unknown interactions among GnIH, GnRH and kisspeptin.

Furthermore, GnIH acts on the pituitary as well as the brain to regulate not only reproduction but also reproductive behaviors in vertebrates. GnIH activates P450arom and increases synthesis of neuroestrogen in the brain. GnIH may also change other neurosteroids' formation by activating or inactivating various steroidogenic enzymes. Steroidogenic enzymes are not only expressed in the brain but also expressed in the pineal gland, which is an endocrine organ located closely to the brain, and actively produces various neurosteroids de novo from cholesterol (170174). As GnIH receptor is expressed in the pineal gland (Sato, M., Narihiro, M., Ubuka, T., Haraguchi., S., Tsutsui., K., unpublished observation), GnIH may regulate neurosteroidogenesis in the pineal gland as in the brain. Future studies are required to develop the emerging concept that the hypothalamic neuropeptide GnIH may modify neurosteroid synthesis in the brain and pineal gland to regulate brain functions.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This review is dedicated to Tsutsui's beloved wife, Rieko Tsutsui. The works described in this review were supported at least partially by Grants-in-Aid for Scientific Research from the Ministry of Education, Science and Culture, Japan (15207007, 16086206, 18107002, 22132004, and 22227002 to KT). The authors are grateful to the following collaborators, J. C. Wingfield, G. E. Bentley, L. J. Kriegsfeld, P. J. Sharp, I. J. Clarke, R. P. Millar, S. A. Sower, Y. Tobari, Y. L. Son, M. Kiyohara, H. Yin, Y. Muneoka, K. Ukena, T. Osugi, V. S. Chowdhury, E. Saigoh, O. Koizumi, M. Ueno, H. Minakata, H. Satake, E. Iwakoshi, D. Daukss, K. Gazda, T. Kosugi, G. Bedecarrats, M. Hisada, T. Kawada, N. L. McGuire, R. Calisi, N. Perfito, S. O'Brien, I. T. Moore, J. P. Jensen, G. J. Kaur, D. W. Wacker, N. A. Ciccone, I. C. Dunn, T. Boswell, S. Kim, Y. C. Huang, J. Reid, J. Jiang, P. Deviche, T. W. Small, M. A. Ottinger, T. Tachibana, M. Furuse, M. A. Cline, D. F. Mei, A. Mason, E. M. Gibson, S. A. Humber, S. Jain, W. P. Williams III, S. Zhao, I. P. Sari, Y. Qi, J. T. Smith, H. C. Parkington, J. Iqbal, Q. Li, A. Tilbrook, K. Morgan, A. J. Pawson, M. Murakami, T. Matsuzaki, T. Iwasa, T. Yasui, M. Irahara, M. A. Johnson, G. S. Fraley, H. Oishi, C. Klausen, C. B. Gilks, T. Yano, P. C. K. Leung, M. Binns, P. A. Cadigan, H. Lai, M. Kitani, A. Suzuuchi, V. Pham, S. Kikuyama, K. Yamamoto, A. Koda, I. Hasumuma, F. Toyoda, K. Sawada, K. Tsunekawa, P. Singh, S. Anjum, A. Krishna, R. Sridaran, S. Sethi, C. M. Chaturvedi, M. R. Jafarzadeh Shirazi, M. J. Zamiri, A. Tamadon, M. Amano, S. Moriyama, M. Iigo, K. Uchida, M. Nozaki, H. Kawauchi, M. Shahjahan, T. Ikegami, H. Doi, A. Hattori, H. Ando, S. Honda, N. Harada, S. Haraguchi, Y. Fukuda, M. Hatori, T. Hirota, M. Iitsuka, N. Kurabayashi, K. Kokame, R. Sato, A. Nakai, T. Miyata, K. Inoue, H. Teranishi, T. Mizuno, M. Narihiro, K. Ishikawa, S. Ishii, M. Sato, K. Tashiro, T. Tokita, J. Y. Seong, J. L. Do Rego, J. Leprince, G. Pelletier, and H. Vaudry.

References

1. Livermore AH, Du Vigneaud V. Preparation of high potency oxytocic material by the use of counter-current distribution. J Biol Chem. (1949) 180:365–73.

PubMed Abstract | Google Scholar

2. Turner RA, Pierce JG, Du Vigneaud V. The purification and the amino acid content of vasopressin preparations. J Biol Chem. (1951) 191:21–8.

PubMed Abstract | Google Scholar

3. Harris GW. Neuronal control of the pituitary gland. Physiol Rev. (1948) 28:139–79.

Google Scholar

4. Boler J, Enzmann F, Folkers K, Bowers CY, Schally AV. The identity of chemical and hormonal properties of the thyrotropin releasing hormone and pyroglutamyl-histidyl-proline amide. Biochem Biophys Res Commun. (1969) 37:705–10. doi: 10.1016/0006-291X(69)90868-7

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Burgus R, Dunn TF, Desiderio D, Guillemin R. Molecular structure of the hypothalamic hypophysiotropic TRF factor of ovine origin: mass spectrometry demonstration of the PCA-His-Pro-NH2 sequence. C R Acad Sci (Paris) (1969) 269:1870–3.

PubMed Abstract | Google Scholar

6. Burgus R, Butcher M, Amoss M, Ling N, Monahan M, Rivier J, et al. Primary structure of the ovine hypothalamic luteinizing hormone-releasing factor (LRF) (LH-hypothalamus-LRF-gas chromatography-mass spectrometry-decapeptide-Edman degradation). Proc Natl Acad Sci USA (1972) 69:278–82. doi: 10.1073/pnas.69.1.278

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Matsuo H, Baba Y, Nair RM, Arimura A, Schally AV. Structure of the porcineLH- and FSH-releasing hormone. I The proposed amino acid sequence. Biochem Biophys Res Commun. (1971) 43:1334–9.

PubMed Abstract | Google Scholar

8. Brazeau P, Vale W, Burgus R, Ling N, Butcher M, Rivier J, et al. Hypothalamic polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science (1973) 179:77–9. doi: 10.1126/science.179.4068.77

PubMed Abstract | CrossRef Full Text | Google Scholar

9. King JA, Millar RP. Structure of chicken hypothalamic luteinizing hormone-releasing hormone. I Structural determination on partially purified material. J Biol Chem. (1982) 257:10722–8.

PubMed Abstract | Google Scholar

10. Miyamoto K, Hasegawa Y, Minegishi T, Nomura M, Takahashi Y, Igarashi M, et al. Isolation and characterization of chicken hypothalamic luteinizing hormone-releasing hormone. Biochem Biophys Res Commun. (1982) 107:820–7. doi: 10.1016/0006-291X(82)90596-4

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Miyamoto K, Hasegawa Y, Nomura M, Igarashi M, Kangawa K, Matsuo H. Identification of the second gonadotropin-releasing hormone in chicken hypothalamus: evidence that gonadotropin secretion is probably controlled by two distinct gonadotropin-releasing hormones in avian species. Proc Natl Acad Sci USA (1984) 81:3874–8. doi: 10.1073/pnas.81.12.3874

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Sherwood NM, Sower SA, Marshak DR, Fraser BA, Brownstein MJ. Primary structure of gonadotropin-releasing hormone in lamprey brain. J Biol Chem. (1986) 15:4812–9.

Google Scholar

13. Tsutsui K, Saigoh E, Ukena K, Teranishi H, Fujisawa Y, Kikuchi M, et al. A novel avian hypothalamic peptide inhibiting gonadotropin release. Biochem Biophys Res Commun. (2000) 275:661–7. doi: 10.1006/bbrc.2000.3350

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Kriegsfeld LJ, Ubuka T, Bentley GE, Tsutsui K. Seasonal control of gonadotropin-inhibitory hormone (GnIH) in birds and mammals. Front. Neuroendocrinol. (2015) 37, 65–75. doi: 10.1016/j.yfrne.2014.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Tsutsui K. Review: a new key neurohormone controlling reproduction, gonadotropin-inhibitory hormone (GnIH): Biosynthesis, mode of action and functional significance. Prog Neurobiol. (2009) 88:76–88. doi: 10.1016/j.pneurobio.2009.02.003

CrossRef Full Text | Google Scholar

16. Tsutsui K. Review: how to contribute to the progress of neuroendocrinology: New insights from discovering novel neuropeptides and neurosteroids regulating pituitary and brain functions. Gen Comp Endocrinol (2016) 227:3–15. doi: 10.1016/j.ygcen.2015.05.019

CrossRef Full Text | Google Scholar

17. Tsutsui K, Bentley GE, Bedecarrats GT, Osugi T, Ubuka T, Kriegsfeld LJ. Review: Gonadotropin-inhibitory hormone (GnIH) and its control of central and peripheral reproductive function. Front Neuroendocrinol. (2010) 31:284–95. doi: 10.1016/j.yfrne.2010.03.001

CrossRef Full Text | Google Scholar

18. Tsutsui K, Bentley GE, Kriegsfeld LJ, Osugi T, Seong JY, Vaudry H. Review: Discovery and evolutionary history of gonadotrophin-inhibitory hormone and kisspeptin: new key neuropeptides controlling reproduction. J Neuroendocrinol. (2010) 22:716–27. doi: 10.1111/j.1365-2826.2010.02018.x

CrossRef Full Text | Google Scholar

19. Tsutsui K, Bentley GE, Ubuka T, Saigoh E, Yin H, Osugi T, et al. Review: The general and comparative biology of gonadotropin-inhibitory hormone (GnIH). Gen Comp Endocrinol. (2007) 153:365–70. doi: 10.1016/j.ygcen.2006.10.005

CrossRef Full Text | Google Scholar

20. Tsutsui K, Ubuka T. Gonadotropin-inhibitory hormone. In: Kastin AJ, Vaudry H, editors. Handbook of Biologically Active Peptides. London: Academic Press (2012). p. 802–11.

Google Scholar

21. Tsutsui K, Ubuka T, Bentley GE, Kriegsfeld LJ. Review: gonadotropin-inhibitory hormone (GnIH): discovery, progress and prospect. Gen Comp Endocrinol. (2012) 177:305–14. doi: 10.1016/j.ygcen.2012.02.013

CrossRef Full Text | Google Scholar

22. Tsutsui K, Ubuka T, Bentley GE, Kriegsfeld LJ. Review: regulatory mechanisms of gonadotropin-inhibitory hormone (GnIH) synthesis and release in photoperiodic animals. Front Neurosci. (2013) 7:60. doi: 10.3389/fnins.2013.00060

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Tsutsui K, Ubuka T, Son YL, Bentley GE, Kriegsfeld LJ. Review: contribution of GnIH research to the progress of reproductive neuroendocrinology. Front. Endocrinol. (2015) 6:179. doi: 10.3389/fendo.2015.00179

CrossRef Full Text | Google Scholar

24. Tsutsui K, Ubuka T, Yin H, Osugi T, Ukena K, Bentley GE, et al. Review: mode of action and functional significance of avian gonadotropin-inhibitory hormone (GnIH). J Exp Zool. (2006) 305A:801–6. doi: 10.1002/jez.a.305

CrossRef Full Text | Google Scholar

25. Tsutsui K, Ukena K. Review: hypothalamic LPXRF-amide peptides in vertebrates: identification, localization and hypophysiotropic activity. Peptides (2006) 27:1121–9. doi: 10.1016/j.peptides.2005.06.036

CrossRef Full Text | Google Scholar

26. Tsutsui K, Son YL, Kiyohara M, Miyata I. Review: discovery of GnIH and its role in hypothyroidism-induced delayed puberty. Endocrinology (2018) 159:62–8. doi: 10.1210/en.2017-00300

CrossRef Full Text | Google Scholar

27. Ukena K, Tsutsui. K. (2005). A new member of the hypothalamic RF-amide peptide family, LPXRF-amide peptides: structure, localization, and function. Mass Spectrom Rev. 24, 469–486. doi: 10.1002/mas.20031

CrossRef Full Text | Google Scholar

28. Tobari Y, Son YL, Ubuka T, Hasegawa Y, Tsutsui K. A new pathway mediating social effects on the endocrine system: female presence acting via norepinephrine release stimulates gonadotropin-inhibitory hormone in the paraventricular nucleus and suppresses luteinizing hormone in quail. J. Neurosci. (2014) 34:9803–11. doi: 10.1523/JNEUROSCI.3706-13.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Ubuka T, Haraguchi S, Tobari Y, Narihiro M, Ishikawa K, Hayashi T, et al. Hypothalamic inhibition of socio-sexual behaviors by increasing neuroestrogen synthesis. Nat Commun. (2014) 5:3061. doi: 10.1038/ncomms4061

CrossRef Full Text | Google Scholar

30. Tsutsui K, Ubuka T. Review: GnIH control of feeding and reproductive behaviors. Front Endocrinol. (2016) 7:170. doi: 10.3389/fendo.2016.00170

CrossRef Full Text | Google Scholar

31. Ubuka T, Son YL, Bentley GE, Millar RP, Tsutsui K. Gonadotropin-inhibitory hormone (GnIH), GnIH receptor and cell signaling. Gen Comp Endocrinol. (2013) 190:10–7. doi: 10.1016/j.ygcen.2013.02.030

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Price DA, Greenberg MJ. Structure of a molluscan cardioexcitatory neuropeptide. Science (1977) 197:670–1. doi: 10.1126/science.877582

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Raffa RB. The action of FMRFamide (Phe-Met-Arg-Phe-NH2) and related peptides on mammals. Peptides (1988) 9:915–22. doi: 10.1016/0196-9781(88)90141-6

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Rastogi RK, D'Aniello B, Pinelli C, Fiorentino M, Di Fiore MM, Di Meglio M, et al. FMRFamide in the amphibian brain: a comprehensive survey. Microsc Res Technol. (2001) 54:158–72. doi: 10.1002/jemt.1130

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Ubuka T, Tsutsui K. Comparative and evolutionary aspects of gonadotropin-inhibitory hormone and FMRFamide-like peptide systems. Front. Neurosci. (2018) 12:747. doi: 10.3389/fnins.2018.00747

CrossRef Full Text | Google Scholar

36. Kumar S, Stecher G, Tamura K. MEGA7: Molecular evolutionary genetics analysis version 7.0 for bigger datasets. Mol Biol Evol. (2016) 33:1870–4. doi: 10.1093/molbev/msw054

CrossRef Full Text | Google Scholar

37. Osugi T, Okamura T, Son YL, Ohkubo M, Ubuka T, Henmi Y, et al. Evolutionary origin of GnIH and NPFF in chordates: insights from novel amphioxus RFamide peptides. PLoS ONE (2014) 9:e100962. doi: 10.1371/journal.pone.0100962

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Dockray GJ, Reeve Jr. JR, Shively J, Gayton RJ, Barnard CS. (1983). A novel active pentapeptide from chicken brain identified by antibodies to FMRFamide. Nature 305, 328-330. doi: 10.1038/305328a0

CrossRef Full Text | Google Scholar

39. McConn B, Wang G, Yi J, Gilbert ER, Osugi T, Ubuka T, et al. (2014). Gonadotropin-inhibitory hormone-stimulation of food intake is mediated by hypothalamic effects in chicks. Neuropeptides 48, 327–34. doi: 10.1016/j.npep.2014.09.001

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Satake H, Hisada M, Kawada T, Minakata H, Ukena K, Tsutsui K. Characterization of a cDNA encoding a novel avian hypothalamic neuropeptide exerting an inhibitory effect on gonadotropin release. Biochem J. (2001) 354:379–85. doi: 10.1042/bj3540379

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Ubuka T, Kim S, Huang YC, Reid J, Jiang J, Osugi T, et al. Gonadotropin-inhibitory hormone neurons interact directly with gonadotropin-releasing hormone-I and -II neurons in European starling brain. Endocrinology (2008) 149:268–78. doi: 10.1210/en.2007-0983

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Tobari Y, Iijima N, Tsunekawa K, Osugi T, Okanoya K, Tsutsui K, et al. Identification of gonadotropin-inhibitory hormone in the zebra finch (Taeniopygia guttata): Peptide isolation, cDNA cloning and brain distribution. Peptides (2010) 31:816–26. doi: 10.1016/j.peptides.2010.01.015

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Ubuka T, Ukena K, Sharp PJ, Bentley GE, Tsutsui K. Gonadotropin-inhibitory hormone inhibits gonadal development and maintenance by decreasing gonadotropin synthesis and release in male quail. Endocrinology (2006) 147:1187–94. doi: 10.1210/en.2005-1178

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Kriegsfeld LJ, Mei DF, Bentley GE, Ubuka T, Mason AO, Inoue K, et al. Identification and characterization of a gonadotropin-inhibitory system in the brains of mammals. Proc Natl Acad Sci USA (2006) 103:2410–5. doi: 10.1073/pnas.0511003103

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Ubuka T, Inoue K. Fukuda Y, Mizuno T, Ukena K, Kriegsfeld LJ, Tsutsui K. (2012). Identification, expression, and physiological functions of Siberian hamster gonadotropin-inhibitory hormone. Endocrinology 153, 373–85. doi: 10.1210/en.2011-1110

CrossRef Full Text | Google Scholar

46. Ubuka T, Lai H, Kitani M, Suzuuchi A, Pham V, Cadigan PA, et al. Gonadotropin-inhibitory hormone identification, cDNA cloning, and distribution in rhesus macaque brain. J Comp Neurol. (2009) 517:841–55. doi: 10.1002/cne.22191

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Ubuka T, Morgan K, Pawson AJ, Osugi T, Chowdhury VS, Minakata H, et al. Identification of human GnIH homologs, RFRP-1 and RFRP-3, and the cognate receptor, GPR147 in the human hypothalamic pituitary axis. PLoS ONE (2009) 4:e8400. doi: 10.1371/journal.pone.0008400

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Ukena K, Iwakoshi E, Minakata H, Tsutsui K. A novel rat hypothalamic RFamide-related peptide identified by immunoaffinity chromatography and mass spectrometry. FEBS Lett. (2002) 512:255–8. doi: 10.1016/S0014-5793(02)02275-5

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Johnson MA, Tsutsui K, Fraley GS. Rat RFamide-related peptide-3 stimulates GH secretion, inhibits LH secretion, and has variable effects on sex behavior in the adult male rat. Horm Behav. (2007) 51:171–80. doi: 10.1016/j.yhbeh.2006.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Murakami M, Matsuzaki T, Iwasa T, Yasui T, Irahara M, Osugi T, et al. Hypophysiotropic role of RFamide-related peptide-3 in the inhibition of LH secretion in female rats. J Endocrinol. (2008) 199:105–12. doi: 10.1677/JOE-08-0197

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Rizwan MZ, Porteous R, Herbison AE, Anderson GM. Cells expressing RFamide-related peptide-1/3, the mammalian gonadotropin-inhibitory hormone orthologs, are not hypophysiotropic neuroendocrine neurons in the rat. Encocrinology (2009) 150:1413–20. doi: 10.1210/en.2008-1287

CrossRef Full Text | Google Scholar

52. Clarke IJ, Sari IP Qi Y, Smith JT, Parkington HC, Ubuka T, Iqbal J, et al. Potent action of RFamide-related peptide-3 on pituitary gonadotropes indicative of a hypophysiotropic role in the negative regulation of gonadotropin secretion. Endocrinology (2008) 149:5811–21. doi: 10.1210/en.2008-0575

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Sari IP, Rao A, Smith JT, Tilbrook AJ, Clarke IJ. Effect of RF-amide-related peptide-3 on luteinizing hormone and follicle-stimulating hormone synthesis and secretion in ovine pituitary gonadotropes. Endocrinology (2009) 150:5549–56. doi: 10.1210/en.2009-0775

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Kadokawa H, Shibata M, Tanaka Y, Kojima T, Matsumoto K, Oshima K, et al. Bovine C-terminal octapeptide of RFamide-related peptide-3 suppresses luteinizing hormone (LH) secretion from the pituitary as well as pulsatile LH secretion in bovines. Domest Anim Endocrinol. (2009) 36:219–24. doi: 10.1016/j.domaniend.2009.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Chartrel N, Dujardin C, Leprince J, Desrues L, Tonon MC, Cellier E, et al. Isolation, characterization, and distribution of a novel neuropeptide, Rana RFamide (R-RFa), in the brain of the European green frog Rana esculenta. J Comp Neurol. (2002) 448:111–27. doi: 10.1002/cne.10253

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Chowdhury VS, Ubuka T, Osugi T, Shimura T, Tsutsui K. Identification, localization and expression of LPXRFamide peptides, and melatonin-dependent induction of their precursor mRNA in the newt brain. J Endocrinol. (2011) 209:211–20. doi: 10.1530/JOE-10-0494

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Koda A, Ukena K, Teranishi H, Ohta S, Yamamoto K, Kikuyama S, et al. A novel amphibian hypothalamic neuropeptide: isolation, localization, and biological activity. Endocrinology (2002) 143:411–9. doi: 10.1210/endo.143.2.8630

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Sawada K, Ukena K, Kikuyama S, Tsutsui K. Identification of a cDNA encoding a novel amphibian growth hormone-releasing peptide and localization of its transcript. J Endocrinol. (2002) 174:395–402. doi: 10.1677/joe.0.1740395

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Sawada K, Ukena K, Satake H, Iwakoshi E, Minakata H, Tsutsui K. Novel fish hypothalamic neuropeptide. Eur J Biochem. (2002) 269:6000–8. doi: 10.1046/j.1432-1033.2002.03351.x

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Shahjahan M, Ikegami T, Osugi T, Ukena K, Doi H, Hattori A, et al. Synchronised expressions of LPXRFamide peptide and its receptor genes: seasonal, diurnal and circadian changes during spawning period in grass puffer. J Neuroendocrinol. (2011) 23:39–51. doi: 10.1111/j.1365-2826.2010.02081.x

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Ukena K, Iwakoshi-Ukena E, Osugi T, Tsutsui K. Identification and localization of gonadotropin-inhibitory hormone (GnIH) orthologs in the hypothalamus of the red-eared slider turtle, Trachemys scripta elegans. Gen Comp Endocrinol. (2015) 227:69–76. doi: 10.1016/j.ygcen.2015.06.009

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Ukena K, Koda A, Yamamoto K, Kobayashi T, Iwakoshi-Ukena E, Minakata H, et al. Novel neuropeptides related to frog growth hormone-releasing peptide: isolation, sequence, and functional analysis. Endocrinology (2003) 144:3879–84. doi: 10.1210/en.2003-0359

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Amano M, Moriyama S, Iigo M, Kitamura S, Amiya N, Yamamori K, et al. Novel fish hypothalamic neuropeptides stimulate the release of gonadotrophins and growth hormone from the pituitary of sockeye salmon. J Endocrinol. (2006) 188:417–23. doi: 10.1677/joe.1.06494

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Moussavi M, Wlasichuk M, Chang JP, Habibi HR. Seasonal effect of GnIH on gonadotrope functions in the pituitary of goldfish. Mol Cell Endocrinol. (2012) 350:53–60. doi: 10.1016/j.mce.2011.11.020

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Moussavi M, Wlasichuk M, Chang JP, Habibi HR. Seasonal effect of gonadotrophin inhibitory hormone on gonadotrophin-releasing hormone-induced gonadotroph functions in the goldfish pituitary. J Neuroendocrinol. (2013) 25:506–16. doi: 10.1111/jne.12024

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Qi X, Zhou W, Li S, Lu D, Yi S, Xie R, et al. Evidences for the regulation of GnRH and GTH expression by GnIH in the goldfish, Carassius auratus. Mol Cell Endocrinol. (2013) 366:9–20. doi: 10.1016/j.mce.2012.11.001

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Zhang Y, Li S, Liu Y, Lu D, Chen H, Huang X, et al. Structural diversity of the GnIH/GnIH receptor system in teleost: its involvement in early development and the negative control of LH release. Peptides (2010) 31:1034–43. doi: 10.1016/j.peptides.2010.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Ubuka T, Parhar I. Dual actions of mammalian and piscine gonadotropin-inhibitory hormones, RFamide-related peptides and LPXRFamide peptides, in the hypothalamic–pituitary–gonadal axis. Front Endocrinol. (2018) 8:377. doi: 10.3389/fendo.2017.00377

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Muñoz-Cueto JA, Paullada-Salmerón JA, Aliaga-Guerrero M, Cowan ME, Parhar IS, Ubuka T. A journey through the gonadotropin-inhibitory hormone system of fish. Front Endocrinol. (2017) 8:285. doi: 10.3389/fendo.2017.00285

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Janvier P. Modern look for ancient lamprey. Nature (2006) 443:921–4. doi: 10.1038/443921a

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Osugi T, Daukss D, Gazda K, Ubuka T, Kosugi T, Nozaki M, et al. Evolutionary origin of the structure and function of gonadotropin-inhibitory hormone: insights from lampreys. Endocrinology (2012) 153:2362–74. doi: 10.1210/en.2011-2046

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Osugi T, Uchida K, Nozaki M, Tsutsui K. Characterization of novel RFamide peptides in the central nervous system of the brown hagfish: isolation, localization, and functional analysis. Endocrinology (2011) 152:4252–64. doi: 10.1210/en.2011-1375

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Osugi T, Ukena K, Sower SA, Kawauchi H, Tsutsui K. Evolutionary origin and divergence of PQRFamide peptides and LPXRFamide peptides in the RFamide peptide family. Insights from novel lamprey RFamide peptides. FEBS J. (2006) 273:1731–43. doi: 10.1111/j.1742-4658.2006.05187.x

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Osugi T, Ubuka T, Tsutsui K. An evolutionary scenario for GnIH in chordates. J Neuroendocrinol. (2015) 27:556–66. doi: 10.1111/jne.12246

CrossRef Full Text | Google Scholar

75. Yin H, Ukena K, Ubuka T, Tsutsui K. A novel G protein-coupled receptor for gonadotropin-inhibitory hormone in the Japanese quail (Coturnix japonica): identification, expression and binding activity. J Endocrinol. (2005) 184:257–66. doi: 10.1677/joe.1.05926

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Bentley GE, Perfito N, Ukena K, Tsutsui K, Wingfield JC. Gonadotropin-inhibitory peptide in song sparrows (Melospiza melodia) in different reproductive conditions, and in house sparrows (Passer domesticus) relative to chicken-gonadotropin-releasing hormone. J Neuroendocrinol. (2003) 15:794–802. doi: 10.1046/j.1365-2826.2003.01062.x

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Hinuma S, Shintani Y, Fukusumi S, Iijima N, Matsumoto Y, Hosoya M, et al. New neuropeptides containing carboxy-terminal RFamide and their receptor in mammals. Nat Cell Biol. (2000) 2:703–8. doi: 10.1038/35036326

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Bonini JA, Jones KA, Adham N, Forray C, Artymyshyn R, Durkin MM, et al. Identification and characterization of two G protein-coupled receptors for neuropeptide FF. J Biol Chem. (2000) 275:39324–31. doi: 10.1074/jbc.M004385200

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Fredriksson R, Lagerström MC, Lundin LG, Schiöth HB. The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol. (2003) 63:1256–72.

PubMed Abstract | Google Scholar

80. Liu Q, Guan XM, Martin WJ, McDonald TP, Clements MK, Jiang Q, et al. Identification and characterization of novel mammalian neuropeptide FF-like peptides that attenuate morphine-induced antinociception. J Biol Chem. (2001) 276:36961–9. doi: 10.1074/jbc.M105308200

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Yoshida H, Habata Y, Hosoya M, Kawamata Y, Kitada C, Hinuma S. Molecular properties of endogenous RFamide-related peptide-3 and its interaction with receptors. Biochim Biophys Acta. (2003) 1593:151–7. doi: 10.1016/S0167-4889(02)00389-0

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Son YL, Ubuka T, Millar RP, Kanasaki H, Tsutsui K. Gonadotropin-inhibitory hormone inhibits GnRH-induced gonadotropin subunit gene transcriptions by inhibiting AC/cAMP/PKA-dependent ERK pathway in LβT2 cells. Endocrinology (2012) 153:2332–43. doi: 10.1210/en.2011-1904

PubMed Abstract | CrossRef Full Text | Google Scholar

83. de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci USA (2003) 100:10972–6. doi: 10.1073/pnas.1834399100

CrossRef Full Text | Google Scholar

84. Kauffman AS, Clifton DK, Steiner RA. Emerging ideas about kisspeptin-GPR54 signaling in the neuroendocrine regulation of reproduction. Trends Neurosci. (2007) 30:504–11. doi: 10.1016/j.tins.2007.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Pinilla L. Aguilar, E. Dieguez, C. Millar, R.P. Tena-Sempere M. (2012). Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol Rev. 92, 1235–316. doi: 10.1152/physrev.00037.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS Jr, Shagoury JK, et al. (2003). The GPR54 gene as a regulator of puberty. N. Engl. J. Med. 349, 1614–27. doi: 10.1056/NEJMoa035322

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Poling MC, Quennell JH, Anderson GM, Kauffman AS. Kisspeptin neurons do not directly signal to RFRP-3 neurones but RFRP-3 may directly modulate a subset of hypothalamic kisspeptin cells in mice. J Neuroendocrinol. (2013) 25:876–86. doi: 10.1111/jne.12084

CrossRef Full Text | Google Scholar

88. Anjum S, Krishna A, Tsutsui K. Inhibitory roles of the mammalian GnIH ortholog RFRP3 in testicular activities in adult mice. J Endocrinol. (2014) 223:79–91. doi: 10.1530/JOE-14-0333

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Bentley GE, Ubuka T, McGuire NL, Chowdhury VS, Morita Y, Yano T, et al. Gonadotropin-inhibitory hormone and its receptor in the avian reproductive system. Gen Comp Endocrinol. (2008) 156:34–43. doi: 10.1016/j.ygcen.2007.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Oishi H, Klausen C, Bentley GE, Osugi T, Tsutsui K, Gilks CB, et al. The human gonadotropin-inhibitory hormone ortholog RFamide-related peptide-3 suppresses gonadotropin-induced progesterone production in human granulose cells. Endocrinology (2012) 153:3435–45. doi: 10.1210/en.2012-1066

CrossRef Full Text | Google Scholar

91. Singh P, Krishna A, Sridaran R, Tsutsui K. Changes in GnRH I, bradykinin and their receptors and GnIH in the ovary of Calotes versicolor during reproductive cycle. Gen Comp Endocrinol. (2008) 159:158–69. doi: 10.1016/j.ygcen.2008.08.016

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Singh P, Krishna A, Sridaran R, Tsutsui K. Immunohistochemical localization of GnRH and RFamide-related peptide-3 in the ovaries of mice during the estrous cycle. J Mol Histol. (2011) 42:371–81. doi: 10.1007/s10735-011-9340-8

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Singh P, Krishna A, Tsutsui K. Effects of gonadotropin-inhibitory hormone on folliculogenesis and steroidogenesis of cyclicmice. Fertil Steril. (2011) 95:1397–404. doi: 10.1016/j.fertnstert.2010.03.052

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Zhao S, Zhu E, Yang C, Bentley GE, Tsutsui K, Kriegsfeld LJ. RFamide- related peptide and messenger ribonucleic acid expression in mammalian testis: association with the spermatogenic cycle. Endocrinology (2010) 151:617–27. doi: 10.1210/en.2009-0978

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Bronson FH. Mammalian Reproductive Biology. Chicago, IL: University of Chicago Press (1989).

Google Scholar

96. Bronson FH. Food-restricted, prepubertal, female rats: rapid recovery of luteinizing hormone pulsing with excess food, and full recovery of pubertal development with gonadotropin-releasing hormone. Endocrinology (1986) 118:2483–7. doi: 10.1210/endo-118-6-2483

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Schneider JE, Wise JD, Benton NA, Brozek JM, Keen-Rhinehart E. When do we eat? Ingestive behavior, survival, and reproductive success. Horm Behav. (2013) 64:702–28. doi: 10.1016/j.yhbeh.2013.07.005

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Bronson FH. Effect of food manipulation on the GnRH-LH-estradiol axis of young female rats. Amer J Physiol. (1988) 254:R616–621.

PubMed Abstract | Google Scholar

99. Bronson FH, Marsteller FA. Effect of short-term food deprivation on reproduction in female mice. Biol Reprod. (1985) 33:660–7.

PubMed Abstract | Google Scholar

100. Schneider JE, Wade GN. Availability of metabolic fuels controls estrous cyclicity of Syrian hamsters. Science (1989) 244:1326–8. doi: 10.1126/science.2734610

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Shahab M, Zaman W, Bashir K, Arslan M. Fasting-induced suppression of hypothalamic-pituitary-gonadal axis in the adult rhesus monkey: evidence for involvement of excitatory amino acid neurotransmitters. Life Sci. (1997) 61:1293–300. doi: 10.1016/S0024-3205(97)00674-7

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Sisk CL, Bronson FH. Effects of food restriction and restoration on gonadotropin and growth hormone secretion in immature male rats. Biol Reprod. (1986) 35:554–61.

PubMed Abstract | Google Scholar

103. Tachibana T, Sato M, Takahashi H, Ukena K, Tsutsui K, Furuse M. Gonadotropin-inhibiting hormone stimulates feeding behavior in chicks. Brain Res. (2005) 1050:94–100. doi: 10.1016/j.brainres.2005.05.035

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Fraley G.S, Coombs E, Gerometta E, Colton S, Sharp PJ, Li Q, Clarke IJ. (2013). Distribution and sequence of gonadotropin-inhibitory hormone and its potential role as a molecular link between feeding and reproductive systems in the Pekin duck (Anas platyrhynchos domestica). Gen. Comp. Endocrinol. 184, 103-110. doi: 10.1016/j.ygcen.2012.11.026

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Tachibana T, Masuda N, Tsutsui K, Ukena K, Ueda H. The orexigenic effect of GnIH is mediated by central opioid receptors in chicks. Comp Biochem Physiol Part A Mol Integr Physiol. (2008) 150:21–5. doi: 10.1016/j.cbpa.2008.02.018

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Clarke IJ, Smith JT, Henry BA, Oldfield BJ, Stefanidis A, Millar RP, et al. (2012). Gonadotropin-inhibitory hormone is a hypothalamic peptide that provides a molecular switch between reproduction and feeding. Neuroendocrinology 95, 305–316. Erratum in: Neuroendocrinology (2013) 97:202. doi: 10.1159/000332822

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Qi Y, Oldfield BJ, Clarke IJ. Projections of RFamide-related peptide-3 neurons in the ovine hypothalamus, with special reference to rigions regulating energy balance and reproduction. J Neuroendocrinol. (2009) 21:690–7. doi: 10.1111/j.1365-2826.2009.01886.x

CrossRef Full Text | Google Scholar

108. Fu LY, van den Pol AN. (2010). Kisspeptin directly excites anorexigenic proopiomelanocortin neurons but inhibits orexigenic neuropeptide Y cells by an indirect synaptic mechanism. J. Neurosci. 30, 10205-10219. doi: 10.1523/JNEUROSCI.2098-10.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Jacobi JS, Coleman HA, Enriori PJ, Parkington HC Li Q, Pereira A, Cowley MA, et al. Paradoxical effect of gonadotrophin-inhibiting hormone to negatively regulate neuropeptide Y neurones in mouse arcuate nucleus. J Neuroendocrinol. (2013) 25:1308–17. doi: 10.1111/jne.12114

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Bentley GE, Jensen JP, Kaur GJ, Wacker DW, Tsutsui K, Wingfield JC. Rapid inhibition of female sexual behavior by gonadotropin-inhibitory hormone (GnIH). Horm Behav. (2006) 49:550–5. doi: 10.1016/j.yhbeh.2005.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Ubuka T, Mukai M, Wolfe J, Beverly R, Clegg S, Wang A, et al. RNA interference of gonadotropin-inhibitory hormone gene induces arousal in songbirds. PLoS ONE (2012) 7:e30202. doi: 10.1371/journal.pone.0030202

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Maney DL, Richardson RD, Wingfield JC. Central administration of chicken gonadotropin-releasing hormone-II enhances courtship behavior in a female sparrow. Horm Behav. (1997) 32:11–8. doi: 10.1006/hbeh.1997.1399

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Piekarski DJ, Zhao S, Jennings KJ, Iwasa T, Legan SJ, Mikkelsen JD, et al. Gonadotropin-inhibitory hormone reduces sexual motivation but not lordosis behavior in female Syrian hamsters (Mesocricetus auratus). Horm Behav. (2013) 64:501–10. doi: 10.1016/j.yhbeh.2013.06.006

CrossRef Full Text | Google Scholar

114. Do-Rego JL, Seong JY, Burel D, Leprince J, Luu-The V, Tsutsui K, et al. Review: Neurosteroid biosynthesis: enzymatic pathways and neuroendocrine regulation by neurotransmitters and neuropeptides. Front Neuroendocrinol. (2009) 30:259–301. doi: 10.1016/j.yfrne.2009.05.006

CrossRef Full Text | Google Scholar

115. Mills AD, Crawford LL, Domjan M, Faure JM. The behavior of the Japanese or domestic quail Coturnix japonica. Neurosci Biobehav Rev. (1997) 21:261–81. doi: 10.1016/S0149-7634(96)00028-0

CrossRef Full Text | Google Scholar

116. Selinger HE, Bermant G. Hormonal control of aggressive behavior in Japanese quail (Coturnix coturnix japonica). Behaviour (1967) 28:255–68. doi: 10.1163/156853967X00037

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Tsutsui K, Ishii S. Effects of sex steroids on aggressive behavior of adult male Japanese quail. Gen Comp Endocrinol. (1981) 44:480–6. doi: 10.1016/0016-6480(81)90336-1

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Schlinger BA, Callard GV. Aromatization mediates aggressive behavior in quail. Gen Comp Endocrinol. (1990) 79:39–53.

PubMed Abstract | Google Scholar

119. Balthazart J, Charlier TD, Cornil CA, Dickens MJ, Harada N, Konkle AT, et al. Sex differences in brain aromatase activity: genomic and non-genomic controls. Front Endocrinol. (2011) 2:34. doi: 10.3389/fendo.2011.00034

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Balthazart J, Cornil CA, Charlier TD, Taziaux M, Ball GF. Estradiol, a key endocrine signal in the sexual differentiation and activation of reproductive behavior in quail. J Exp Zool A Ecol Genet Physiol. (2009) 311:323–45. doi: 10.1002/jez.464

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Yahr P. Data and hypotheses in tales of dihydrotestosterone. Horm Behav. (1979) 13:92–6. doi: 10.1016/0018-506X(79)90037-0

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Ubuka T, Ueno M, Ukena K, Tsutsui K. Developmental changes in gonadotropin-inhibitory hormone in the Japanese quail (Coturnix japonica) hypothalamo-hypophysial system. J Endocrinol. (2003) 178:311–8. doi: 10.1677/joe.0.1780311

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Ukena K, Ubuka T, Tsutsui K. Distribution of a novel avian gonadotropin-inhibitory hormone in the quail brain. Cell Tissue Res. (2003) 312:73–9. doi: 10.1007/s00441-003-0700-x

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Absil P, Riters LV, Balthazart J. Preoptic aromatase cells project to the mesencephalic central gray in the male Japanese quail (Coturnix japonica). Horm Behav. (2001) 40:369–83. doi: 10.1006/hbeh.2001.1702

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Cornil CA, Ball GF, Balthazart J. Rapid control of male typical behaviors by brain-derived estrogens. Front Neuroendocrinol. (2012) 33:425–46. doi: 10.1016/j.yfrne.2012.08.003

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Balthazart J, Surlemont C. Androgen and estrogen action in the preoptic area and activation of copulatory behavior in quail. Physiol Behav. (1990) 48:599–609. doi: 10.1016/0031-9384(90)90198-D

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Panzica GC, Viglietti-Panzica C, Balthazart J. The sexually dimorphic medial preoptic nucleus of quail: a key brain area mediating steroid action on male sexual behavior. Front Neuroendocrinol. (1996) 17:51–125. doi: 10.1006/frne.1996.0002

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Balthazart J, Baillien M, Ball GF. Rapid control of brain aromatase activity by glutamatergic inputs. Endocrinology (2006) 147:359–66. doi: 10.1210/en.2005-0845

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Balthazart J, Baillien M, Ball GF. Rapid and reversible inhibition of brain aromatase activity. J Neuroendocrinol. (2001) 13:63–73. doi: 10.1046/j.1365-2826.2001.00598.x

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Balthazart J, Baillien M. Ball GF. (2001). Phosphorylation processes mediate rapid changes of brain aromatase activity. J. Steroid. Biochem. Mol. Biol. 79, 261–77. doi: 10.1016/S0960-0760(01)00143-1

CrossRef Full Text | Google Scholar

131. Balthazart J, Baillien M, Charlier TD, Ball GF. Calcium-dependent phosphorylation processes control brain aromatase in quail. Eur J Neurosci. (2003) 17:1591–606. doi: 10.1046/j.1460-9568.2003.02598.x

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Charlier TD, Harada N, Balthazart J, Cornil CA. Human and quail aromatase activity is rapidly and reversibly inhibited by phosphorylating conditions. Endocrinology (2011) 152:4199–210. doi: 10.1210/en.2011-0119

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Pineda R, Garcia-Galiano D, Sanchez-Garrido MA, Romero M, Ruiz-Pino F, Aguilar E, et al. Characterization of the inhibitory roles of RFRP3, the mammalian ortholog of GnIH, in the control of gonadotropin secretion in the rat: in vivo and in vitro studies. Am J Physiol Endocrinol Metab. (2010) 299:E39–E46. doi: 10.1152/ajpendo.00108.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Simonin F, Schmitt M, Laulin JP, Laboureyras E, Jhamandas JH, MacTavish D, et al. RF9, a potent and selective neuropeptide FF receptor antagonist, prevents opioid-induced tolerance associated with hyperalgesia. Proc Natl Acad Sci USA (2006) 103:466–71. doi: 10.1073/pnas.0502090103

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Steele RE, Mellor LB, Sawyer W.K., Wasvary JM, Browne LJ. In vitro and in vivo studies demonstrating potent and selective estrogen inhibition with the nonsteroidal aromatase inhibitor CGS 16949A. Steroids (1987) 50:147–61.

PubMed Abstract | Google Scholar

136. Wade J, Schlinger BA, Hodges L, Arnold AP. Fadrozole: a potent and specific inhibitor of aromatase in the zebra finch brain. Gen Comp Endocrinol. (1994) 94:53–61. doi: 10.1006/gcen.1994.1059

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Greives TJ, Kingma SA, Beltrami G, Hau M. Melatonin delays clutch initiation in a wild songbird. Biol Lett. (2012) 8:330–2. doi: 10.1098/rsbl.2011.1100

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Guyomarc'h C, Lumineau S, Vivien-Roels B, Richard J, Deregnaucourt S. (2001). Effect of melatonin supplementation on the sexual development in European quail (Coturnix coturnix). Behav. Processes 53, 121-130. doi: 10.1016/S0376-6357(01)00133-4

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Ohta M, Kadota C, Konishi H. A role of melatonin in the initial stage of photoperiodism in the Japanese quail. Biol Reprod. (1989) 40:935–41. doi: 10.1095/biolreprod40.5.935

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Rozenboim I, Aharony T, Yahav S. The effect of melatonin administration on circulating plasma luteinizing hormone concentration in castrated White Leghorn roosters. Poult Sci. (2002) 81:1354–9. doi: 10.1093/ps/81.9.1354

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Juss TS, Meddle SL, Servant RS, King VM. Melatonin and photoperiodic time measurement in Japanese quail (Coturnix coturnix japonica). Proc Biol Sci. (1993) 254:21–8. doi: 10.1098/rspb.1993.0121

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Wilson FE. Neither retinal nor pineal photoreceptors mediate photoperiodic control of seasonal reproduction in American tree sparrows (Spizella arborea) J. Exp Zool. (1991) 259:117–27.

Google Scholar

143. Ubuka T, Bentley GE, Ukena K, Wingfield JC, Tsutsui K. Melatonin induces the expression of gonadotropin-inhibitory hormone in the avian brain. Proc Natl Acad Sci USA (2005) 102:3052–7. doi: 10.1073/pnas.0403840102

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Chowdhury VS, Yamamoto K, Ubuka T, Bentley GE, Hattori A, Tsutsui K. Melatonin stimulates the release of gonadotropin-inhibitory hormone by the avian hypothalamus. Endocrinology (2010) 151:271–80. doi: 10.1210/en.2009-0908

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Mason AO, Duffy S, Zhao S, Ubuka T, Bentley GE, Tsutsui K, et al. Photoperiod and reproductive condition are associated with changes in RFamide-related peptide (RFRP) expression in Syrian hamsters (Mesocricetus auratus). J Biol Rhythms. (2010) 25:176–85. doi: 10.1177/0748730410368821

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Revel FG, Saboureau M, Pévet P, Simonneaux V, Mikkelsen JD. RFamide-related peptide gene is a melatonin-driven photoperiodic gene. Endocrinology (2008) 149:902–12.

PubMed Abstract | Google Scholar

147. Simonneaux V, Ancel C, Poirel VJ, Gauer F. Kisspeptins and RFRP-3 act in concert to synchronize rodent reproduction with seasons. Front Neurosci. (2013) 7:22. doi: 10.3389/fnins.2013.00022

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Piekarski DJ, Jarjisian SG, Perez L, Ahmad H, Dhawan N, Zucker I, et al. Effects of pinealectomy and short day lengths on reproduction and neuronal RFRP-3, kisspeptin, and GnRH in female Turkish hamsters. J Biol Rhythms (2014) 29:181–91. doi: 10.1177/0748730414532423

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Janati A, Talbi R, Klosen P, Mikkelsen JD, Magoul R, Simonneaux V, El Ouezzani S. (2013). Distribution and seasonal variation in hypothalamic RF-amide peptides in a semi-desert rodent, the jerboa. J. Neuroendocrinol. 25, 402–11. doi: 10.1111/jne.12015

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Dardente H, Birnie M, Lincoln GA, Hazlerigg DG. RFamide-related peptide and ts cognate receptor in the sheep: cDNA cloning, mRNA distribution in the hypothalamus and the effect of photoperiod. J Neuroendocrinol. (2008) 20:1252–9. doi: 10.1111/j.1365-2826.2008.01784.x

CrossRef Full Text | Google Scholar

151. Smith JT, Coolen LM, Kriegsfeld LJ, Sari IP, Jaafarzadehshirazi MR, Maltby M, et al. Variation in kisspeptin and RFamide-related peptide (RFRP) expression and terminal connections to gonadotropin-releasing hormone neurons in the brain: a novel medium for seasonal breeding in the sheep. Endocrinology (2008) 149:5770–82. doi: 10.1210/en.2008-0581

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Gingerich S, Wang X, Lee PK, Dhillon SS, Chalmers JA, Koletar MM, et al. The generation of an array of clonal, immortalized cell models from the rat hypothalamus: analysis of melatonin effects on kisspeptin and gonadotropin-inhibitory hormone neurons. Neuroscience (2009) 162:1134–40. doi: 10.1016/j.neuroscience.2009.05.026

PubMed Abstract | CrossRef Full Text | Google Scholar

153. McGuire NL, Bentley GE. A functional neuropeptide system in vertebrate gonads: Gonadotropin-inhibitory hormone and its receptor in testes of field-caught house sparrow (Passer domesticus). Gen Comp Endocrinol. (2010) 166:565–72. doi: 10.1016/j.ygcen.2010.01.010

PubMed Abstract | CrossRef Full Text | Google Scholar

154. McGuire NL, Kangas K, Bentley GE. Effects of melatonin on peripheral reproductive function: regulation of testicular GnIH and testosterone. Endocrinology (2011) 152:3461–70. doi: 10.1210/en.2011-1053

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Chand D, Lovejoy DA. Stress and reproduction: controversies and challenges. Gen Comp Endocrinol. (2011) 171:253–7. doi: 10.1016/j.ygcen.2011.02.022

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Kirby ED, Geraghty AC, Ubuka T, Bentley GE, Kaufer D. Stress increases putative gonadotropin inhibitory hormone and decreases luteinizing hormone in male rats. Proc Natl Acad Sci USA (2009) 106:11324–9. doi: 10.1073/pnas.0901176106

PubMed Abstract | CrossRef Full Text | Google Scholar

157. Son YL, Ubuka T, Narihiro M, Fukuda Y, Hasunuma I, Yamamoto K, et al. Molecular basis for the activation of gonadotropin-inhibitory hormone gene transcription by corticosterone. Endocrinology (2014) 155:1817–26. doi: 10.1210/en.2013-2076

PubMed Abstract | CrossRef Full Text | Google Scholar

158. Breuner CW, Greenberg AL, Wingfield JC. Noninvasive corticosterone treatment rapidly increases activity in gambel's white-crowned sparrows (Zonotrichia leucophrys gambelii). Gen Comp Endocrinol. (1998) 111:386–94.

PubMed Abstract | Google Scholar

159. Breuner CW, Wingfield JC. Rapid behavioral response to corticosterone varies with photoperiod and dose. Horm Behav. (2000) 37:23–30. doi: 10.1006/hbeh.1999.1554

PubMed Abstract | CrossRef Full Text | Google Scholar

160. McGuire NL, Koh A, Bentley GE. (2013). The direct response of the gonads to cues of stress in a temperate songbird species is season-dependent. PeerJ. 1:e139. doi: 10.7717/peerj.139

CrossRef Full Text

161. Kaewwongse M, Takayanagi Y, Onaka T. Effects of RFamide-related peptide (RFRP)-1 and RFRP-3 on oxytocin release and anxiety-related behaviour in rats. J Neuroendocrinol. (2011) 23:20–7. doi: 10.1111/j.1365-2826.2010.02077.x

PubMed Abstract | CrossRef Full Text | Google Scholar

162. Ullah R, Batool A, Wazir M, Naz R, Rahman TU, Wahab F, et al. Gonadotropin-inhibitory hormone and RF9 stimulate hypothalamic-pituitary-adrenal axis in adult male rhesus monkeys. Neuropeptides (2017) 66:1–7. doi: 10.1016/j.npep.2017.07.005

PubMed Abstract | CrossRef Full Text | Google Scholar

163. Calisi RM, Díaz-Muñoz SL, Wingfield JC, Bentley GE. Social and breeding status are associated with the expression of GnIH. Genes Brain Behav. (2011) 10:557–64. doi: 10.1111/j.1601-183X.2011.00693.x

PubMed Abstract | CrossRef Full Text | Google Scholar

164. Cornil CA, Stevenson TJ, Ball GF. Are rapid changes in gonadal testosterone release involved in the fast modulation of brain estrogen effects? Gen Comp Endocrinol. (2009) 163:298–305. doi: 10.1016/j.ygcen.2009.04.029

PubMed Abstract | CrossRef Full Text | Google Scholar

165. Delville Y, Sulon J, Hendrick JC, Balthazart J. Effect of the presence of females on the pituitary-testicular activity in male Japanese quail (Coturnix coturnix japonica). Gen Comp Endocrinol. (1984) 55:295–305. doi: 10.1016/0016-6480(84)90115-1

PubMed Abstract | CrossRef Full Text | Google Scholar

166. Anasti JN, Flack MR, Froehlich J, Nelson LM, Nisula BC. A potential novel mechanism for precocious puberty in juvenile hypothyroidism. J Clin Endocrinol Metab. (1995) 80:276–27.

PubMed Abstract | Google Scholar

167. Dittricha R, Beckmanna MW, Oppelta PG, Hoffmanna I, Lotza L, Kuwertb T, et al. Thyroid hormone receptors and reproduction. J Reprod Immunol. (2011) 90:58–66. doi: 10.1016/j.jri.2011.02.009

CrossRef Full Text | Google Scholar

168. Niedziela M, Korman E. Severe hypothyroidism due to autoimmune atrophic thyroiditis-predicted target height and a plausible mechanism for sexual precocity. J Pediatr Endocrinol Metab. (2001) 14:901–7. doi: 10.1515/JPEM.2001.14.7.901

PubMed Abstract | CrossRef Full Text | Google Scholar

169. Kiyohara M, Son YL, Tsutsui K. (2017). Involvement of gonadotropin-inhibitory hormonein pubertal disorders induced by thyroid status. Sci. Rep. 7:1042. doi: 10.1038/s41598-017-01183-8

PubMed Abstract | CrossRef Full Text | Google Scholar

170. Haraguchi S, Hara S, Ubuka T, Mita M, Tsutsui K. Possible role of pineal allopregnanolone in Purkinje cell survival. Proc Natl Acad Sci USA (2012) 109:21110–5. doi: 10.1073/pnas.1210804109

PubMed Abstract | CrossRef Full Text | Google Scholar

171. Haraguchi S, Yamamoto Y, Suzuki Y, Chang JH, Sato M, Mita M, Ueda. H. and Tsutsui K. 7α-Hydroxypregnenolone, a key neuronal modulator of locomotion, stimulates upstream migration by means of the dopaminergic system in salmon. Sci Rep. (2015). 5:12546. doi: 10.1038/srep12546

PubMed Abstract | CrossRef Full Text | Google Scholar

172. Hatori M, Hirota T, Iitsuka M, Kurabayashi N, Haraguchi S, Kokame K, et al. Light-dependent and circadian clock-regulated activation of sterol regulatory element-binding protein, X-box-binding protein 1, and heat shock factor pathways. Proc Natl Acad Sci USA (2011) 108:4864–9. doi: 10.1073/pnas.1015959108

PubMed Abstract | CrossRef Full Text | Google Scholar

173. Tsutsui K, Haraguchi S, Fukada Y, Vaudry H. Review: brain and pineal 7α-hydroxypregnenolone stimulating locomotor activity: identification, mode of action and regulation of biosynthesis. Front Neuroendocrinol. (2013) 34:179–89. doi: 10.1016/j.yfrne.2013.05.002

CrossRef Full Text | Google Scholar

174. Tsutsui K, Haraguchi S, Hatori M, Hirota T, Fukada Y. Review: biosynthesis and biological actions of pineal neurosteroids in domestic birds. Neuroendocrinology (2013) 98:97–105. doi: 10.1159/000353782

CrossRef Full Text | Google Scholar

Keywords: gonadotropin-inhibitory hormone (GnIH), gonadotropin-releasing hormone (GnRH), gonadotropins, melatonin, glucocorticoid, norepinephrine, thyroid hormone, reproduction

Citation: Tsutsui K and Ubuka T (2018) How to Contribute to the Progress of Neuroendocrinology: Discovery of GnIH and Progress of GnIH Research. Front. Endocrinol. 9:662. doi: 10.3389/fendo.2018.00662

Received: 05 June 2018; Accepted: 23 October 2018;
Published: 12 November 2018.

Edited by:

Honoo Satake, Suntory Foundation for Life Sciences, Japan

Reviewed by:

Yudong Zhou, Zhejiang University, China
Rafael Henrique Nóbrega, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Brazil

Copyright © 2018 Tsutsui and Ubuka. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Kazuyoshi Tsutsui, k-tsutsui@waseda.jp
Takayoshi Ubuka, takayoshi.ubuka@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.