You are on page 1of 11

REVIEW ARTICLE

published: 16 April 2013


doi: 10.3389/fnins.2013.00060

Review: regulatory mechanisms of gonadotropin-inhibitory


hormone (GnIH) synthesis and release in photoperiodic
animals
Kazuyoshi Tsutsui 1*, Takayoshi Ubuka1 , George E. Bentley 2 and Lance J. Kriegsfeld 3
1
Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, Tokyo, Japan
2
Department of Integrative Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
3
Department of Psychology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA

Edited by: Gonadotropin-inhibitory hormone (GnIH) is a novel hypothalamic neuropeptide that was
Gregoy Y. Bedecarrats, University of discovered in quail as an inhibitory factor for gonadotropin release. GnIH inhibits
Guelph, Canada
gonadotropin synthesis and release in birds through actions on gonadotropin-releasing
Reviewed by:
hormone (GnRH) neurons and gonadotropes, mediated via the GnIH receptor (GnIH-R),
Denise D. Belsham, University of
Toronto, Canada GPR147. Subsequently, GnIH was identified in mammals and other vertebrates. As
Chris Scott, Charles Sturt University, in birds, mammalian GnIH inhibits gonadotropin secretion, indicating a conserved role
Australia for this neuropeptide in the control of the hypothalamic-pituitary-gonadal (HPG) axis
Kyungjin Kim, Seoul National
University, South Korea
across species. Identification of the regulatory mechanisms governing GnIH expression
and release is important in understanding the physiological role of the GnIH system.
*Correspondence:
Kazuyoshi Tsutsui, Laboratory of A nocturnal hormone, melatonin, appears to act directly on GnIH neurons through its
Integrative Brain Sciences, receptor to induce expression and release of GnIH in quail, a photoperiodic bird. Recently,
Department of Biology, Waseda a similar, but opposite, action of melatonin on the inhibition of expression of mammalian
University, and Center for Medical
Life Science of Waseda University,
GnIH was shown in hamsters and sheep, photoperiodic mammals. These results in
2-2 Wakamatsu-cho, Shinjuku-ku, photoperiodic animals demonstrate that GnIH expression is photoperiodically modulated
Tokyo 162-8480, Japan. via a melatonin-dependent process. Recent findings indicate that GnIH may be a mediator
e-mail: k-tsutsui@waseda.jp of stress-induced reproductive disruption in birds and mammals, pointing to a broad
role for this neuropeptide in assessing physiological state and modifying reproductive
effort accordingly. This paper summarizes the advances made in our knowledge regarding
the regulation of GnIH synthesis and release in photoperiodic birds and mammals.
This paper also discusses the neuroendocrine integration of environmental signals,
such as photoperiods and stress, and internal signals, such as GnIH, melatonin, and
glucocorticoids, to control avian and mammalian reproduction.
Keywords: gonadotropin-releasing hormone (GnRH), gonadotropin-inhibitory hormone (GnIH), gonadotropins,
melatonin, glucocorticoids, photoperiod, stress, reproduction

INTRODUCTION hypothalamic neuropeptide for gonadotropin secretion was, until


The neuroendocrine integration of environmental and inter- recently, unknown, although gonadal sex steroids and inhibin
nal signals controls reproduction across vertebrate species. The can modulate gonadotropin secretion. Findings from the last
reproductive axis of vertebrates integrates information from 12 years, however, indicate that GnRH is not the sole hypotha-
a wide range of systems via direct and indirect neurochemi- lamic regulatory neuropeptide of vertebrate reproduction, with
cal inputs. Many of the neuropeptidergic pathways involved in gonadotropin-inhibitory hormone (GnIH) playing a key role in
the transduction of environmental stimuli into neuroendocrine the inhibition of reproduction.
signals have been well-studied. A hypothalamic neuropeptide, In 2000, GnIH was discovered in the avian brain. In a search for
gonadotropin-releasing hormone (GnRH), is the primary fac- novel neuropeptides regulating the release of pituitary hormones,
tor regulating secretion of both of the gonadotropins, luteinizing Tsutsui and colleagues identified a novel hypothalamic neuropep-
hormone (LH), and follicle-stimulating hormone (FSH), and tide that directly acts on the pituitary to inhibit gonadotropin
acts as a key neurohormone for vertebrate reproduction. Since release in quail and termed it GnIH (Tsutsui et al., 2000). This was
the discovery of GnRH in the brain of mammals at the begin- the first demonstration of a hypothalamic neuropeptide inhibit-
ning of 1970s (Matsuo et al., 1971; Burgus et al., 1972), several ing gonadotropin release in any vertebrate. From the past 12
other GnRHs have been identified in the brain of a variety years of research, we now know that GnIH exists in a variety
of vertebrates (King and Millar, 1982; Miyamoto et al., 1982, of avian species and regulates avian reproduction by decreas-
1984; Sherwood et al., 1983, 1986; Jimenez-Liñan et al., 1997; ing gonadotropin release and synthesis via action on the ante-
Okubo et al., 2000a,b; Yoo et al., 2000). In contrast, an inhibitory rior pituitary gland and the GnRH system, mediated via the

www.frontiersin.org April 2013 | Volume 7 | Article 60 | 1


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

GnIH receptor (GnIH-R), GPR147 (Tsutsui et al., 2000, 2005, novel hypothalamic neuropeptide having a C-terminal Arg-Phe-
2006, 2007a,b, 2010a,b, 2012; Satake et al., 2001; Bentley et al., NH2 motif (RFamide peptide) (Tsutsui et al., 2000). GnIH has a
2003, 2006, 2008, 2010; Ubuka et al., 2003, 2005, 2006, 2008a,b, previously-unreported dodecapeptide structure, Ser-Ile-Lys-Pro-
2009a,b, 2012a,b; Ukena et al., 2003; Ciccone et al., 2004; Osugi Ser-Ala-Tyr-Leu-Pro-Leu-Arg-Phe-NH2 (SIKPSAYLPLRFamide)
et al., 2004; Yin et al., 2005; Tsutsui and Ukena, 2006; Tsutsui, (Tsutsui et al., 2000). Its C-terminus is identical to chicken
2009; Tobari et al., 2010; Tsutsui and Ubuka, 2013). LPLRFamide that was reported to be the first RFamide peptide
Tsutsui and colleagues have further identified GnIH orthologs isolated in vertebrates (Dockray et al., 1983), which is likely to be
in other vertebrates including mammals (for reviews, see Ukena a degraded fragment of GnIH (for reviews, see Ukena and Tsutsui,
and Tsutsui, 2005; Tsutsui and Ukena, 2006; Tsutsui et al., 2005; Tsutsui, 2009; Tsutsui et al., 2010a,b). After the isolation of
2007a, 2010a,b; Tsutsui, 2009; Kriegsfeld et al., 2010; Smith and GnIH in the quail brain, the precursor polypeptide for GnIH was
Clarke, 2010; Tsutsui and Ubuka, 2013). As in birds, mammalian examined (Satake et al., 2001). A cDNA that encodes GnIH pre-
GnIH orthologs appear to act as key neurohormones to inhibit cursor polypeptide was identified by a combination of 3 and 5
gonadotropin release in several mammalian species (Kriegsfeld rapid amplification of cDNA ends (3 /5 RACE) in quail (Satake
et al., 2006; Johnson et al., 2007; Clarke et al., 2008; Gibson et al., et al., 2001) and other avian species, such as chickens, sparrows,
2008; Murakami et al., 2008; Kadokawa et al., 2009; Sari et al., zebra finches, and starlings (for reviews, see Tsutsui, 2009; Tsutsui
2009; Ubuka et al., 2012a). Furthermore, fish GnIH orthologs et al., 2010a,b). The GnIH precursor encodes one GnIH and two
also inhibit gonadotropin release in fish (Zhang et al., 2010; GnIH-related peptides (GnIH-RP-1 and GnIH-RP-2) possessing
Moussavi et al., 2012), indicating a conserved role for GnIH an LPXRFamide (X = L or Q) sequence at their C-termini in all
and its orthologs in the control of the hypothalamo-pituitary- avian species studied. GnIH was further isolated as a mature pep-
gonadal (HPG) axis across species. Thus, GnIH, a newly dis- tide in starlings (Ubuka et al., 2008a) and zebra finches (Tobari
covered hypothalamic neuropeptide, and its orthologs appear to et al., 2010) and GnIH-RP-2 was also identified in quail (Satake
act as a key neurohormone controlling vertebrate reproduction, et al., 2001). From the past 12 years of research, thus we now know
generally (for reviews, see Tsutsui, 2009; Tsutsui et al., 2010a,b). that GnIH exists in various avian species, and regulates avian
Following the discovery of GnIH, kisspeptin, encoded by the reproduction by decreasing gonadotropin release and synthesis
Kiss1 gene, was discovered in mammals. In contrast to GnIH, via action on gonadotropes in the anterior pituitary gland and
kisspeptin has a stimulatory effect on GnRH neurons via its recep- GnRH neurons in the hypothalamus, mediated via the GnIH-R,
tor, GPR54, causing up-regulation of the HPG axis (for reviews, GPR147 (Tsutsui et al., 2000, 2005, 2006, 2007a,b, 2010a,b; Satake
see Ukena and Tsutsui, 2005; Tsutsui and Ukena, 2006; Tsutsui, et al., 2001; Bentley et al., 2003, 2006, 2010; Ubuka et al., 2003,
2009; Tsutsui et al., 2010a). Kisspeptin and GPR54 are considered 2005, 2006, 2008a,b, 2009a,b, 2012a,b; Ukena et al., 2003; Ciccone
to be essential for puberty and subsequent fertility in mammals. et al., 2004; Osugi et al., 2004; Yin et al., 2005; Tsutsui and Ukena,
At present, the Kiss1 gene has been identified in most vertebrates, 2006; Tsutsui, 2009; Tobari et al., 2010; Tsutsui and Ubuka, 2013)
including mammals, amphibians, and fish (for reviews, see Ukena (Figure 1).
and Tsutsui, 2005; Tsutsui and Ukena, 2006; Tsutsui, 2009; Tsutsui After the discovery of GnIH in birds, GnIH orthologs have
et al., 2010a). Most recently, we found a second isoform of Kiss1, been further identified in a number of other vertebrates including
designated Kiss2, in several vertebrate groups, but not in birds and mammals (for reviews, see Tsutsui et al., 2007a, 2010a,b; Tsutsui,
rodents (Lee et al., 2009). 2009). In mammals, cDNAs that encode LPXRFamide peptides
Thus, the discovery of GnIH and kisspeptin has changed our similar to GnIH have been investigated by a gene database search
understanding of the vertebrate reproductive axis despite the (Hinuma et al., 2000). The cDNAs identified from mammalian
well-accepted dogma that reproduction is mainly under stimula- brain encode three GnIH orthologs [known as RFamide-related
tory control from GnRH. Seasonal breeders such as photoperiodic peptides (RFRPs)], RFRP-1, -2, and -3, in bovines and humans
animals rely on environmental and internal cues to initiate and/or and two GnIH orthologs, RFRP-1 and -3, in rodents (for reviews,
terminate reproduction, and these signals need to be further see Tsutsui, 2009; Tsutsui et al., 2010a,b). RFRP-1 and -3 are both
translated and integrated to generate the appropriate neuroen- LPXRFamide peptides, but RFRP-2 is not an LPXRFamide pep-
docrine response. Herein we review what is currently known tide. The mammalian GnIH orthologs, RFRP-1 and/or RFRP-3
about the regulation of GnIH synthesis and release by photope- were also identified as mature peptides in bovines (Fukusumi
riods and melatonin, a pineal hormone, in photoperiodic birds et al., 2001; Yoshida et al., 2003), rats (Ukena et al., 2002), ham-
and mammals. We also highlight the effect of stress on GnIH sters (Ubuka et al., 2012a), monkeys (Ubuka et al., 2009a), and
synthesis. Finally, we discuss the neuroendocrine integration of humans (Ubuka et al., 2009b). Subsequently, it was shown that
environmental signals, such as photoperiods and stress, and inter- RFRP-3 inhibits gonadotropin synthesis and/or release in var-
nal signals, such as GnIH, melatonin, and glucocorticoids, to ious mammalian species (Johnson et al., 2007; Clarke et al.,
control avian and mammalian reproduction. 2008; Murakami et al., 2008; Kadokawa et al., 2009; Sari et al.,
2009; Ubuka et al., 2012a). In addition, RFRP-3 was shown
STRUCTURE AND FUNCTION to inhibit GnRH-stimulated gonadotropin synthesis in mam-
It was believed for a long time that GnRH is the only hypotha- malian pituitary gonadotropes (Sari et al., 2009). Accordingly,
lamic neuropeptide regulating pituitary gonadotropin synthesis at least RFRP-3 acts as a functional ortholog of GnIH in terms
and release in vertebrates. In 2000, however, GnIH was discov- of reducing gonadotropin secretion (for reviews, see Tsutsui,
ered in the brain of the Japanese quail whilst in search of a 2009; Tsutsui et al., 2010a,b). More recently, it was found that

Frontiers in Neuroscience | Neuroendocrine Science April 2013 | Volume 7 | Article 60 | 2


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

LH release in non-breeding song sparrows (Osugi et al., 2004).


GnIH administration in vivo and in vitro inhibits the synthesis
of LHβ- and FSHβ-subunits within the pituitary gland of quail
and chickens (Ciccone et al., 2004; Ubuka et al., 2006), indicat-
ing a dual role for GnIH within the pituitary-acting over different
time-frames to reduce first the release of gonadotropins into the
circulation followed by inhibition of LH and FSH synthesis. Thus,
it has become clear that GnIH in birds is an important regulator
of pituitary gonadotropin synthesis in addition to gonadotropin
release (Ciccone et al., 2004; Osugi et al., 2004; Bentley et al.,
2006; Ubuka et al., 2006) (Figure 1). Despite our published data
on the distribution of GnIH in the median eminence and the
GnIH-R, GPR147 in the pituitary, there are some inconsistencies
in the literature. For example, rufous-winged sparrows do not
seem to exhibit GnIH-ir fibers in the median eminence (Small
et al., 2007). Nor do IV injections of GnIH rapidly inhibit LH
secretion in this species (Deviche et al., 2006). It may be possible
that there is another source of GnIH that can influence pituitary
gonadotropin release.
FIGURE 1 | Summary of the neuroendocrine integration of
The binding activity of GnIH-R was characterized in the quail
environmental signals and internal signals to control reproduction. The (Yin et al., 2005). The crude membrane fraction of COS-7 cells
neuroendocrine integration of environmental signals, such as photoperiods transfected with the putative GnIH-R (GPR147) cDNA specifi-
and stress, and internal signals, such as GnIH, melatonin, and cally bound GnIH and GnIH-RPs in a concentration-dependent
glucocorticoids, is important for the control of avian and mammalian
manner, indicating that GPR147 is GnIH-R (Yin et al., 2005).
reproduction. GnIH inhibits gonadotropin synthesis and release by directly
acting on the pituitary or by inhibiting the activity of GnRH neurons in birds
GPR147 is a member of the G-protein coupled receptor (GPCR)
and mammals. GnIH can also inhibit reproductive behavior by possibly family which couples to Gα and, upon activation inhibits adeny-
acting within the brain. GnIH expression is photoperiodically modulated via lyl cyclase (AC) activity, thus reducing intracellular cAMP levels
a melatonin-dependent process. Melatonin induces the expression of GnIH (Bédécarrats et al., 2009; Shimizu and Bédécarrats, 2010). Type
in quail and rats, whereas melatonin inhibits the expression of GnIH in
III GnRH receptor (GnRH-R-III) is the pituitary specific GnRH
hamsters and sheep. Stress induces the expression of GnIH in birds and
mammals. GnIH may therefore be a mediator of stress-induced receptor in the chicken (Shimizu and Bédécarrats, 2006). In
reproductive disruption. See the text for details. the chicken pituitary gland, the chicken GnRH-R-III (cGnRH-
R-III)/GnIH-R ratio changes during sexual maturation in favor
of cGnRH-R-III that appears to result in hypothalamic control
intracerebroventricular (ICV) administration of RFRP-1 also of gonadotropin secretion shifting from inhibitory to stimula-
inhibits gonadotropin release in hamsters (Ubuka et al., 2012a). tory, with corresponding changes in GnRH-induced cAMP levels
Thus, as in birds, the mammalian GnIH orthologs, RFRP-1 (Shimizu and Bédécarrats, 2010). GnIH-R mRNA quantity was
and RFRP-3, act to inhibit gonadotropin secretion in mammals significantly higher in the pituitaries of sexually immature chick-
(Kriegsfeld et al., 2006, 2010; Johnson et al., 2007; Clarke et al., ens relative to sexually mature chickens (Maddineni et al., 2008).
2008; Gibson et al., 2008; Murakami et al., 2008; Kadokawa et al., Estradiol or a combination of estradiol and progesterone treat-
2009; Ubuka et al., 2012a). Recently, an inhibitory action of fish ment caused a significant decrease in pituitary GnIH-R mRNA
GnIH orthologs (LPXRFamide peptides) on gonadotropin release (Maddineni et al., 2008). Furthermore, GnIHR-ir cells were found
was also reported in zebrafish (Zhang et al., 2010) and goldfish to be colocalized with LHβ mRNA-, or FSHβ mRNA-containing
(Moussavi et al., 2012). In general, GnIH and its orthologs seem cells, possibly mediating the inhibitory effect of GnIH on LH and
to act similarly across vertebrate species to regulate reproduction, FSH secretion (Maddineni et al., 2008) (Figure 1).
although some exceptions exist. In addition to the effects of GnIH on the pituitary, the con-
tact of GnRH neurons by GnIH axons is perhaps the most
LOCALIZATION AND MODE OF ACTION conserved property of GnIH:GnRH interactions in birds and
In birds, GnIH neurons are located in the paraventricular nucleus other vertebrates (Figure 1). Initially discovered in house spar-
(PVN) in the hypothalamus and their major projections reach rows (Bentley et al., 2003), contact of GnRH neurons by GnIH
the median eminence of the hypothalamus (Tsutsui et al., 2000; has been observed in all other vertebrates studied to date, includ-
Ubuka et al., 2003; Ukena and Tsutsui, 2005). Based on the obser- ing humans (Ubuka et al., 2009b). In birds, GnIH neurons project
vation that GnIH-immunoreactive (-ir) neurons project to the to GnRH-I and -II neurons and presumably inhibit the action of
external layer of the median eminence in quail, the first study these two types of GnRH via the GnIH-R, GPR147, in European
on GnIH (Tsutsui et al., 2000) focused on the effect of GnIH starlings (Ubuka et al., 2008a). Experimental support of this
on pituitary gonadotropin release. In songbirds, intravenous (IV) notion comes from Bentley et al. (2006), in which centrally-
injection of GnIH can rapidly reduce circulating LH in breeding infused GnIH inhibited circulating LH and reduced copula-
white-crowned sparrows in addition to inhibiting GnRH-induced tion solicitation in female white-crowned sparrows. Further,

www.frontiersin.org April 2013 | Volume 7 | Article 60 | 3


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

rhodaminated GnIH was shown to bind to putative GnRH-II In rodents, GnIH cell bodies are clustered in the dorsomedial
neurons and, in a later study on European starlings, GnRH-I nucleus of the hypothalamus (DMH) with extensive projections
and -II neurons were shown to express GnIH-R (GPR147) mRNA to hypothothamic and limbic structures (Johnson et al., 2007;
(Ubuka et al., 2008a). Thus, physiological, behavioral and histo- Kriegsfeld et al., 2010; Ubuka et al., 2012a). In other species,
logical evidence in several species combine to indicate a direct such as sheep, GnIH neurons are located in both the PVN and
role for GnIH in regulation of GnRH in the brain of songbirds more widely distributed throughout the mediobasal hypothala-
(Figure 1). In contrast to the highly-clustered cell bodies in the mus (Smith et al., 2008). With regard to reproduction, GnIH
PVN, GnIH-ir fibers are widely distributed in diencephalic and neurons have been shown to project to the external layer of
mesencephalic regions in birds, suggesting the regulation of other, the median eminence of Syrian hamsters, with the expression
non-reproductive behaviors (Bentley et al., 2003; Ukena et al., of GnIH-R (GPR147) observed in pituitary in the same inves-
2003; Ubuka et al., 2008a). For example, central administration tigation (Gibson et al., 2008). These findings suggested that,
of GnIH stimulates feeding behavior in chickens (Tachibana et al., as in birds, GnIH may act at the pituitary level to suppress
2005). gonadotropin secretion (Figure 1). In sheep, GnIH administra-
To identify the mechanism of GnIH neurons in the regula- tion down-regulates LHβ- and FSHβ-subunit expression (Sari
tion of behavior, Ubuka et al. (2012b) investigated the effect et al., 2009) and results in marked reductions in LH pulse ampli-
of RNA interference (RNAi) of the GnIH gene on the behav- tude without affecting pulse frequency (Clarke et al., 2008). In
ior of white-crowned sparrows, a highly social songbird species. cows, repeated IV administration of GnIH leads to reduced LH
Administration of small interfering RNA against GnIH precursor pulse frequency (Kadokawa et al., 2009), indicating the possibility
mRNA into the third ventricle of male and female birds reduced of different modes of action across species. Taken together, these
resting time, spontaneous production of complex vocalizations, findings provide convincing evidence that the function of GnIH
and stimulated brief agonistic vocalizations. GnIH RNAi fur- is conserved in mammals as well as in birds.
ther enhanced song production of short duration in male birds Smith et al. (2012) measured GnIH-3 (RFRP-3) concentra-
when they were challenged by playbacks of novel male songs. tions in hypophyseal portal blood in ewes during the non-
These behaviors resembled those of breeding birds during territo- breeding (anestrous) season and during the luteal and follicular
rial defense. The overall results suggest that GnIH gene silencing phases of the estrous cycle in the breeding season. Pulsatile GnIH-
induces arousal. In addition, the activities of male and female 3 secretion was observed in the portal blood of all animals. Mean
birds were negatively correlated with GnIH mRNA expression in GnIH-3 pulse amplitude and pulse frequency were higher dur-
the PVN. Density of GnIH neuronal fibers in the ventral tegmen- ing the non-breeding season. GnIH-3 was virtually undetectable
tal area was decreased by GnIH RNAi treatment in female birds, in peripheral blood plasma. To determine the role of secreted
and the number of GnRH neurons that received close apposi- GnIH-3, they further examined its effects on GnRH-stimulated
tions of GnIH neuronal fiber terminals was negatively correlated LH secretion in hypothalamo-pituitary-disconnected ewes, and
with the activity of male birds. In summary, GnIH may decrease found a significant reduction in the LH response to GnRH.
arousal level resulting in the inhibition of specific motivated GPR147 was expressed in fractions enriched for gonadotropes
behavior, such as in reproductive contexts (Ubuka et al., 2012b). somatotropes, and lactotropes (Smith et al., 2012). IV administra-
As in birds, GnIH has been found in the brains of all mam- tion of RF9 to anestrous acyclic ewes induced a sustained increase
malian species studied to date, including humans (Fukusumi in LH plasma concentrations, and the increase in LH plasma
et al., 2001; Ukena et al., 2002; Yoshida et al., 2003; Clarke concentrations induced by RF9 was blocked by previous adminis-
et al., 2009; Ubuka et al., 2009a,b, 2012a; Kriegsfeld et al., tration of GnRH antagonist, Teverelix (Caraty et al., 2012). These
2010). As described previously, the cDNA for GnIH encodes two data clearly show that GnIH-3 is secreted into portal blood to act
LPXRFamide peptides, RFRP-1 and RFRP-3, in mammals. Across on pituitary gonadotropes, reducing the action of GnRH in sheep.
mammals, administration of GnIH suppresses gonadotropin Some disparity across studies and species has drawn into
secretion (Kriegsfeld et al., 2006; Clarke et al., 2008; Kadokawa question the generality of the actions of GnIH directly on the
et al., 2009; Sari et al., 2009; Pineda et al., 2010a,b; Ubuka et al., pituitary. In one study in rats, for example, GnIH fibers were not
2012a). Most studies to date have applied RFRP-3 as the mam- detected in the external layer of the median eminence (Rizwan
malian ortholog of avian GnIH (Clarke et al., 2008; Kadokawa et al., 2009). In the same study, peripheral injections of the retro-
et al., 2009; Pineda et al., 2010a). Our recent study further showed grade tracer, Fluorogold, did not label GnIH cell bodies whereas
a strong suppressive effect of RFRP-1 on LH in Siberian hamsters a majority of cells bodies were labeled following central injec-
(Ubuka et al., 2012a). Consistent with results for GnIH across tions. Finally, in this study, IV injections of GnIH did not affect
mammalian species, ICV injections of the GnIH-R (GPR147) basal LH concentrations, but GnRH-induced LH secretion was
antagonist, RF9, result in rapid and sustained, dose-dependent modestly suppressed. This latter finding is consistent with work
increases in LH in male and female mice and rats (Pineda et al., by another group indicating that GnIH had minimal impact
2010b) and the ewe (Caraty et al., 2012). Together, these findings on LH secretion in cultured rat pituitary, but significantly sup-
suggest that RFRP-1 and/or -3 operate as GnIH in mammalian pressed GnRH-induced LH secretion in vitro (Murakami et al.,
species. Whether RFRP-1 and -3 operate synergistically in mam- 2008). Curiously, in this study, IV injections of GnIH led to
mals to inhibit HPG axis activity or whether each peptide dom- gross reductions in LH at 120 min post injection whereas central
inates differentially across species remains an interesting avenue injections did not impact LH concentrations. In another study
for further investigation. of rats, intraperitoneal (IP) injections of the GnIH-R (GRP147)

Frontiers in Neuroscience | Neuroendocrine Science April 2013 | Volume 7 | Article 60 | 4


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

antagonist, RF9, led to modest increases in LH in vivo, whereas involved in steroid biosynthesis and gamete maturation indicates
application of RF9 to pituitary cultures was without effect (Pineda the likely importance of GnIH as a paracrine/autocrine regula-
et al., 2010b). In contrast, central injections of this receptor tor within reproductive tissues (for a review, see McGuire and
antagonist markedly stimulated LH release. Another study using Bentley, 2010a). In house sparrows, testosterone secretion from
ovariectomized female rats treated with a low dose of estrogen, gonadotropin-stimulated cultured house sparrow testes was sig-
found no effect of central GnIH injections on LH (Anderson et al., nificantly decreased by the application of GnIH in vitro (McGuire
2009). Together, these findings make it difficult to reconcile the and Bentley, 2010b). As is the case in birds, GnIH and its recep-
specific impact of GnIH on the pituitary, at least in rats. The dis- tor are robustly expressed in Syrian hamster testis (Zhao et al.,
parity across studies likely results from differences in the time 2010), indicating that a role for GnIH in gonadal function may
course of sample collection, antibody, peptide, and tissue prepa- be common to all vertebrates. In hamsters, GnIH expression is
ration, and other differences in experimental procedures and the confined to seminiferous tubules, with spermatocytes and sper-
reproductive state of the animals investigated. matids expression the GnIH-R, GPR147, suggesting an impor-
GnIH-R (GPR147) couples to Gαi to inhibit AC (Hinuma tant role in spermatogenesis. In macaque, GnIH is expressed
et al., 2000). A recent study of ours using immortalized mouse in Leydig cells, Sertoli cells, spermatogonia, and spermatocytes
gonadotrope cell line (LβT2 cells) has demonstrated that the (McGuire and Bentley, 2010b), suggesting roles in steroidoge-
inhibitory action of mouse GnIHs (RFRPs) on gonadotropin nesis, spermatogenesis, and sperm production. GnIH is also
gene expression is mediated by an inhibition of AC/cAMP/cAMP- found in the granulosa cells of mouse ovarian follicles during
dependent protein kinase (PKA)-dependent extracellular signal- proestrus and estrus and in the luteal cells during diestrus 1 and
regulated kinase (ERK) pathway (Son et al., 2012). In the 2 (Singh et al., 2011a), and macaque granulosa cells and oocytes
sheep, GnIH can inhibit both GnRH-induced intracellular Ca2+ (McGuire and Bentley, 2010b). In mice, GnIH was closely asso-
increase and ERK phosphorylation, impacting GnRH-induced ciated with GnRH expression in mouse ovarian tissue (Singh
gonadotropin release and synthesis (Sari et al., 2009). et al., 2011b), suggesting that a coordinated balance between
As in birds, GnIH neurons project monosynaptically to GnRH GnRH and GnIH activity plays an important role in follicular
neurons (Johnson et al., 2007; Smith et al., 2008; Kriegsfeld development and atresia throughout the ovulatory cycle. More
et al., 2010) and GnRH neurons express the GnIH-R, GPR147 recently, GnIH was shown to suppress gonadotropin-induced
in mammals (Rizwan et al., 2012; Ubuka et al., 2012a). GPR74, progesterone production in human granulosa cells (Oishi et al.,
which GnIH binds with lower affinity (Bonini et al., 2000), is not 2012).
expressed on GnRH neurons (Rizwan et al., 2012). Direct appli-
cation of GnIH inhibits neuronal firing in GnRH cells and this REGULATION OF GnIH SYNTHESIS AND RELEASE BY
inhibitory action of GnIH persists when amino acid transmis- PHOTOPERIODS AND MELATONIN IN PHOTOPERIODIC
sion is blocked, providing strong evidence for a direct inhibitory ANIMALS
role of GnIH on the GnRH neuronal system (Ducret et al., Identification of the regulatory mechanisms governing GnIH
2009; Wu et al., 2009). Similarly, ICV administration of GnIH synthesis and release is important in understanding the phys-
decreases the activational state of GnRH neurons (Anderson et al., iological role of the GnIH system. The mechanisms regulating
2009). Together, these findings, combined with those indicating GnIH synthesis have been investigated in birds. In general, the
suppressive action of central GnIH administration or stimula- annual changes in pineal melatonin secretion drive the repro-
tory action of central RF9 administration on LH concentrations ductive responses of photoperiodic mammals (Bronson, 1989).
(Kriegsfeld et al., 2006; Anderson et al., 2009; Rizwan et al., 2009; However, several studies suggested that melatonin is not respon-
Pineda et al., 2010a,b), point to a potent role for GnIH in GnRH sible for changes in seasonal reproduction in birds (e.g., Wilson,
inhibition (Figure 1). 1991; Juss et al., 1993). Despite information to the contrary,
In addition, vertebrate gonads are the sites of synthesis and there are data available on regulation of seasonal processes by
action of GnIH. The gonadal GnIH system was first discovered melatonin, including but not limited to that of gonadal activ-
in European starlings and Japanese quail (Bentley et al., 2008). ity and gonadotropin secretion (Ohta et al., 1989; Bentley et al.,
GnIH-ir was observed in the interstitial and germ cells in the 1999; Bentley and Ball, 2000; Guyomarc’h et al., 2001; Rozenboim
quail testis (Bentley et al., 2008). GnIH-ir was also observed in et al., 2002). More recently there has been the suggestion that
the columnar epithelial cells in the quail epididymis (Bentley the avian hypothalamus can synthesize melatonin de novo (Kang
et al., 2008). GnIH-R mRNA was localized by in situ hybridiza- et al., 2007). These researchers demonstrated the presence of tryp-
tion in the germ cells and interstitial cells in the testis and tophan hydroxylase 1 and 5-HT N-acetyl-transferase, two key
columnar epithelial cells in the epididymis in quail (Bentley enzymes in melatonin biosynthesis, along with melatonin. This
et al., 2008). In situ hybridization of GnIH in house sparrow finding could explain the lack of effect of pinealectomy on the
testis produced a strong reaction product localized to interstitial avian reproductive system; day length information could still be
cells, and GnIH-R to interstitial cells and secondary spermato- encoded by the hypothalamic melatonin system. Considering the
cytes (McGuire and Bentley, 2010a). In female birds, GnIH-ir inhibitory effects of GnIH on gonadotropin release and synthe-
substance was observed in the thecal and granulose layers in sis (Tsutsui et al., 2000; Ciccone et al., 2004; Osugi et al., 2004;
starling ovary (Bentley et al., 2008). The presence of transcripts Ubuka et al., 2006) and the inhibitory effects of short-day (SD)
for GnIH and GnIH-R in the gonads and associated tissues, photoperiods, we manipulated melatonin levels in quail, a highly
and the expression of GnIH and GnIH-R by specific cell types photoperiodic bird species, by removing sources of melatonin and

www.frontiersin.org April 2013 | Volume 7 | Article 60 | 5


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

investigating the action of melatonin on GnIH expression in the In addition, studies on other avian species with breeding
quail brain (Ubuka et al., 2005). strategies different from that of quail are required for us to deter-
The pineal gland and eyes are the major sources of melatonin mine whether or not the melatonin effects on GnIH we observe
in quail (Underwood et al., 1984). Ubuka et al. (2005) found in quail are universal in photoperiodic avian species. Unlike quail,
that pinealectomy combined with orbital enucleation (Px+Ex) which terminate reproduction during decreasing day lengths,
decreased the expression of GnIH precursor mRNA and the many passerine species terminate their reproductive activities
mature GnIH peptide in the diencephalon. Melatonin adminis- while day lengths are still increasing, so in those species presum-
tration to Px+Ex birds caused a dose-dependent increase in the ably the effects of melatonin on GnIH synthesis and release would
expression of GnIH precursor mRNA and production of mature also be decreasing. There is also some evidence in sparrow species
peptide (Ubuka et al., 2005). Furthermore, the expression of that the GnIH system is active during the breeding season (LD,
GnIH increased under SD (Ubuka et al., 2005), when the noc- short duration of melatonin secretion) and is responsible for the
turnal duration of melatonin secretion increases (Cockrem and temporary cessation of reproductive activities in response to envi-
Follett, 1985). Mel1c , a melatonin receptor subtype, was expressed ronmental stimuli such as stress (Bentley et al., 2003; Calisi et al.,
by GnIH-ir neurons in the PVN, thus indicating a direct action of 2011). How the regulation of GnIH by melatonin fits into the
melatonin on GnIH neurons (Ubuka et al., 2005). Finally, mela- seasonal reproductive profile of these species has not yet been
tonin receptor autoradiography further revealed specific binding determined.
of melatonin in the PVN (Ubuka et al., 2005). Taken together, In mammals, GnIH also appears to play an important role
these data indicate that melatonin acts directly on GnIH neu- in monitoring internal and external status and integrating this
rons via its receptor to induce GnIH expression (Figure 1). This information to control reproductive functioning precisely and
was the first demonstration in any vertebrate that melatonin can maximize reproductive success. In hamsters, reproductive status
directly induce synthesis of a neuropeptide, and indicates that is primarily driven by day lengths (photoperiods). If GnIH is
GnIH is capable of transducing photoperiodic information to involved in the control of reproductive functions in hamsters, the
the avian reproductive axis via changes in the melatonin signal expressions of GnIH precursor mRNA and GnIH peptides should
(Figure 1). be regulated by photoperiod and melatonin. We found that
Melatonin is likely a key factor controlling GnIH neural func- expression of GnIH precursor mRNA, GnIH-immunoreactivity
tion. Therefore, we further investigated the role of melatonin in GnIH-ir perikarya and GnIH-ir fiber density decreased under
in the regulation of GnIH release and the correlation of GnIH SD photoperiod compared to LD in Siberian hamsters, a highly
release with LH release in quail. Melatonin administration dose- photoperiodic mammalian species (Ubuka et al., 2012a). This
dependently increased GnIH release from hypothalamic explants inhibitory effect of SD was not seen if the hamsters were pinealec-
in vitro (Chowdhury et al., 2010). Furthermore, GnIH release was tomized, and melatonin administration inhibited the expressions
photoperiodically controlled in quail with diurnal changes neg- of GnIH precursor mRNA, GnIH-immunoreactivity in GnIH-ir
atively correlated with plasma LH concentrations (Chowdhury perikarya and GnIH-ir fiber density. It was further shown that
et al., 2010). GnIH release from hypothalamic explants in quail the percentage of GnRH-ir neurons receiving GnIH-ir fiber ter-
exposed to long-day (LD) photoperiods was greater in tissues minals decreased in SD photoperiod. These results are in line
collected during the dark period than during the light period with the results in Syrian hamsters (Revel et al., 2008; Mason
(Chowdhury et al., 2010). Conversely, plasma LH concentra- et al., 2010) and sheep (Dardente et al., 2008). These findings
tions decreased during the dark period. In contrast to LD, GnIH are consistent with the possibility that the activity of GnIH neu-
release from hypothalamic explants increased in quail under SD, rons decreases in SD by the inhibitory action of pineal melatonin
when the duration of nocturnal secretion of melatonin increases in hamsters. The functional significance of this counterintuitive
(Chowdhury et al., 2010). These results indicate that melatonin relationship between photoperiod and GnIH is not straightfor-
plays a role in stimulating not only GnIH synthesis but also ward and is discussed further below. On the other hand, it was
GnIH release, thus inhibiting plasma LH concentrations in quail shown that melatonin directly induces GnIH mRNA expression in
(Figure 1). the rat GnIH cell line (rHypoE-7) (Gingerich et al., 2009). These
Although it is well-established that GnRH-I controls repro- results demonstrate that the expression of GnIH is photoperiodi-
ductive function in birds (Sharp et al., 1990), no direct associa- cally modulated via a melatonin-dependent process in mammals,
tion between the gonadal regression observed during SD and a like in birds, although how it is related to seasonal reproduction
decrease in hypothalamic GnRH-I has been made in quail (Foster is not yet clear (Figure 1).
et al., 1988; Dawson et al., 2001). In Chowdhury et al. (2010), There are different actions of melatonin on GnIH expres-
we are the first to show that the release of GnIH from hypotha- sion across species (Figure 1). Melatonin increases the expression
lamic explants in quail previously exposed to SD was increased, of GnIH in quail, a photoperiodic bird, (Ubuka et al., 2005;
and this increase was associated with a decrease in plasma LH lev- Chowdhury et al., 2010). A similar, but opposite, action of mela-
els and gonadal regression. Because melatonin stimulates GnIH tonin on the inhibition of the expression of GnIH was shown
synthesis and release and GnIH inhibits gonadotropin synthe- in photoperiodic mammals, such as Syrian and Siberian ham-
sis and release in quail, it could be assumed that the gonadal sters (Revel et al., 2008; Mason et al., 2010; Ubuka et al., 2012a)
regression occurring in birds exposed to SD is a direct conse- and sheep (Dardente et al., 2008). Quail is a LD breeder, which
quence of the melatonin/GnIH system on circulating LH levels activates its reproductive activity in LD and suppresses its repro-
(Figure 1). ductive activity in SD. It is understandable that the expression of

Frontiers in Neuroscience | Neuroendocrine Science April 2013 | Volume 7 | Article 60 | 6


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

GnIH is stimulated by melatonin, a nocturnal hormone, and SD a significant increase in GnIH positive cells was seen in stressed
when the duration of melatonin secretion is long. Accordingly, birds in spring. These data imply an influence of stress upon the
it was hypothesized that the increase of GnIH expression may GnIH system. It is likely that this effect of immobilization stress is
inhibit reproductive activity in SD in quail (Ubuka et al., 2005). mediated directly via glucocorticoid action on GnIH neurons via
The opposite but similar thoughts can be applied to sheep. Sheep glucocorticoid receptor (GR).
is a SD breeder that activates its reproductive activity in SD and Kirby et al. (2009) also investigated how acute immobilization
suppresses its reproductive activity in LD. It is also understand- stress alters GnIH mRNA and protein levels in adult male rats.
able that the expression of GnIH is inhibited by melatonin, a GnIH mRNA and protein levels increased immediately follow-
nocturnal hormone, and SD when the duration of melatonin ing stress but returned to a lower level 24 h after stress. Thus,
secretion is long. In contrast, the expression of GnIH increases GR expression by GnIH neurons is evident in birds and mam-
in LD in sheep. Accordingly, it was hypothesized that the increase mals, and is likely to be an evolutionarily conserved phenomenon.
of GnIH expression may inhibit reproductive activity in LD in GnIH may therefore be a mediator of stress-induced reproduc-
sheep (Dardente et al., 2008). On the other hand, it was diffi- tive disruption in birds and mammals (Figure 1). The role of the
cult to understand the inhibitory effect of melatonin or SD on GnIH system in suppressing reproduction during times of stress
GnIH expression in hamsters, because hamsters are LD breeders may not be common across species; stress does not affect GnIH
(Revel et al., 2008; Mason et al., 2010). Some clarity was garnered peptide or mRNA expression in sheep (Papargiris et al., 2011).
from a recent report showing that GnIH may have a stimula-
tory effect on gonadotropin secretion in SD in Siberian hamsters NEUROENDOCRINE INTEGRATION OF ENVIRONMENTAL
(Ubuka et al., 2012a). Long duration of melatonin secretion in AND INTERNAL SIGNALS
SD may need to decrease GnIH expression to inhibit reproductive In birds, termination of breeding is coincident with long-duration
activities of Siberian hamsters in SD. Recently, Ancel et al. (2012) melatonin secretion in the fall. Although the termination of
reported stimulatory effects of RFRP-3 on the gonadotrophic axis breeding results from refractoriness to melatonin stimulation,
in the male Syrian hamster (Ancel et al., 2012), underscoring given the actions on melatonin on GnIH (Ubuka et al., 2005;
the importance of considering sex and photoperiodic conditions Chowdhury et al., 2010), it is likely that this neuropeptide is
when evaluating the role of GnIH. Further studies are required involved in avian reproductive termination in response to the
to understand the role of melatonin controlling GnIH expres- nocturnal melatonin signal. More recently, melatonin implants
sion in photoperiodic animals (see the section of Neuroendocrine have been shown to delay the onset of clutch initiation in female
Integration of Environmental and Internal Signals). great tits but without affecting clutch size, body mass, or timing
Several lines of evidence indicate that kisspeptin and GnIH of onset of activity (Greives et al., 2012). In other species, GnIH
might work in concert to guide seasonal changes in reproduction and GnRH expression are positively correlated (Bentley et al.,
across species (reviewed in Simonneaux et al., 2012). In Syrian 2003; Calisi and Bentley, 2009). Such a relationship may appear
hamsters, for example, Kiss1 mRNA levels are lower in the Arc counter-intuitive: why should an inhibitory hormone increase
of SD hamsters and melatonin appeared to be necessary for the during the breeding season (Dawson et al., 2001)? We hypothesize
seasonal change; pineal-gland ablation prevents the SD-induced that in some species, GnIH acts to induce a temporary pause in
down-regulation of Kiss1 expression (Revel et al., 2006). In Syrian reproductive effort without full HPG axis suppression and regres-
hamsters, short day lengths lead to decreased kisspeptin-ir in sion, which would cause the individual to miss the opportunity
the anteroventral periventricular nucleus (AVPV) and increased to breed in that year. Such a missed opportunity in a single year
expression in the Arc (Greives et al., 2007). In Siberian hamsters, could markedly reduce reproductive success in short-lived song-
chronic administration of kisspeptin restored testicular activity birds. In their natural environments, this temporary cessation of
of SD hamsters despite persisting photoinhibitory conditions. reproduction occurs in response to unpredictable environmen-
The authors propose that photoperiod, via melatonin, modulates tal cues, such as stress, or mate/nest availability. Some support
Kiss1 signaling to drive the reproductive axis (Revel et al., 2006). for this possibility comes from an experiment on European star-
In Syrian hamsters, chronic administration of kisspeptin does lings in which nesting opportunities were limited (Calisi et al.,
not restore reproductive competence to SD animals, likely due to 2011). Under these conditions, birds that outcompeted others
downregulation of kisspeptin receptor (Greives et al., 2008). Data for nest boxes (“winners”) had significantly fewer numbers of
from these studies suggest that GnIH and kisspeptin may both GnIH-producing cells than those without nest boxes (“losers”)
be direct or indirect targets of melatonin, decoding photoperiod, at the beginning of the breeding season (Calisi et al., 2011).
and appropriately signaling the reproductive axis based on time This relationship changed with breeding stage; once “winners”
of year. had begun to incubate eggs, their GnIH content increased sig-
nificantly above that of “losers.” Thus, whereas birds appeared
EFFECT OF STRESS ON GnIH SYNTHESIS reproductively capable across treatments, these findings indicate
Calisi et al. (2008) hypothesized that suppressive effects of stress that hypothalamic GnIH may serve as a short-term modula-
upon reproduction are mediated by the hypothalamic GnIH tor of reproductive behaviors in response to social environment.
system in birds. The effects of capture-handling stress on the Consequently, hypothalamic GnIH may serve as a regulator of
numbers of GnIH neurons in the hypothalamus of adult male reproductive behaviors in response to social environment (Calisi
and female house sparrows were tested. There were more GnIH- et al., 2011). How this pattern of GnIH expression during LD in a
positive cells in birds in fall vs. those sampled in the spring, and naturally-housed songbird relates to the increased expression and

www.frontiersin.org April 2013 | Volume 7 | Article 60 | 7


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

release of GnIH by SD-induced melatonin action in laboratory mammalian GnIH is found in hamsters and sheep. The vari-
quail (Ubuka et al., 2005; Chowdhury et al., 2010) remains to be ety of melatonin actions and modes of regulation of GnIH raise
seen. Studies on wild quail might provide some valuable answers interesting questions as to the evolution of GnIH and its adap-
to this question (see Calisi and Bentley, 2009). tive significance. Recent studies have further demonstrated that
As in birds, mammals in their natural environments are faced stress induces GnIH expressions in birds and mammals. GnIH
with the challenge of maximizing reproductive success while con- may therefore be a mediator of stress-induced reproductive dis-
sidering obstacles to survival such as reduced food availability ruption. Thus, the neuroendocrine integration of environmental
and inclement weather (Bronson, 1989). As a result, animals must signals, such as photoperiods and stress, and internal signals, such
have a mechanism for transducing environmental conditions into as GnIH, melatonin, and glucocorticoids, is important for the
neural signals to guide sexual motivation and reproductive axis control of avian and mammalian reproduction.
activity. The GnIH system is well-situated to act as an inter-
mediary between the environment and the reproductive axis. ACKNOWLEDGMENTS
For example, GnIH neurons project to neuropeptide-Y, pro- This work was supported by Grants-in-Aid for Scientific
opiomelanocortin, orexin and melanin concentrating cells in the Research from the Ministry of Education, Science and Culture,
ovine brain (Clarke et al., 2009; Qi et al., 2009), providing a neural Japan (22132004 and 22227002 to Kazuyoshi Tsutsui), National
circuit capable of driving motivation to feed over sex during unfa- Institutes of Health Grant (HD-050470 to Lance J. Kriegsfeld),
vorable energetic conditions. Consistent with this notion, admin- and National Science Foundation Integrative Organismal Systems
istration of GnIH increases feeding across mammalian species Grants (0641188, 0956338, and 0920753 to George E. Bentley).
(Johnson et al., 2007). Likewise, in Syrian hamsters, NPY cells We are grateful to the following collaborators, J. C. Wingfield,
project to GnIH cells in the DMH, providing a neural pathway for P. J. Sharp, I. J. Clarke, H. Vaudry, R. P. Millar, A. Sower, G.
interpretation of energetic status and initiation of GnIH-induced Bedecarrats, T. Osugi, V. S. Chowdhury, E. Saigoh, H. Yin, Y.
inhibition of reproductive function during times of reduced food Tobari, Y. L. Son, K. Inoue, Y. Muneoka, K. Ukena, H. Teranishi,
availability (Klingerman et al., 2011). In agreement with this Y. Fujisawa, S. Ishii, M. Ueno, H. Minakata, H. Satake, M. Hisada,
possibility, GnIH neuronal activity is increased following food T. Kawada, N. L. McGuire, R. Calisi, N. Perfito, S. O’Brien, I. T.
deprivation in this species (Klingerman et al., 2011). 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,
CONCLUSION T. W. Small, M. A. Ottinger, T. Tachibana, M. Furuse, D. F. Mei,
The discovery of GnIH has fundamentally changed our under- A. Mason, E. M. Gibson, S. A. Humber, S. Jain, W. P. Williams III,
standing of hypothalamic control of reproduction. Recent studies S. Zhao, I. P. Sari, Y. Qi, J. T. Smith, H. C. Parkington, J. Iqbal,
on photoperiodic birds and mammals have demonstrated that Q. Li, A. Tilbrook, K. Morgan, A. J. Pawson, M. Murakami, T.
GnIH expression is photoperiodically modulated via a melatonin- Matsuzaki, T. Iwasa, T. Yasui, M. Irahara, M. A. Johnson, G. S.
dependent process. Melatonin, a nocturnal hormone, appears to Fraley, M. Binns, P. A. Cadigan, H. Lai, M. Kitani, A. Suzuuchi, V.
act directly on GnIH neurons through its receptor to induce Pham, S. Kikuyama, K. Yamamoto, A. Koda, K. Sawada, P. Singh,
expression and release of GnIH in quail and rats. A similar, but A. Krishna, R. Sridaran, M. Amano, S. Moriyama, M. Iigo, and
opposite, action of melatonin on the inhibition of expression of H. Kawauchi.

REFERENCES Bentley, G. E., and Ball, G. F. (2000). studies of gonadotropin-inhibitory Bronson, F. H. (1989). Mammalian
Ancel, C., Bentsen, A. H., Photoperiod-dependent and hormone (GnIH) in the mam- Reproductive Biology. Chicago:
Sébert, M. E., Tena-Sempere, M., -independent regulation of mela- malian hypothalamus, pituitary University of Chicago Press.
Mikkelsen, J. D., and Simonneaux, tonin receptors in the forebrain of and gonads. Brain Res. 1364, Burgus, R., Butcher, M., Amoss, M.,
V. (2012). Stimulatory effect of songbirds. J. Neuroendocrinol. 12, 62–71. Ling, N., Monahan, M., Rivier,
RFRP-3 on the gonadotrophic 745–752. Bentley, G. E., Ubuka, T., McGuire, N. J., et al. (1972). Primary struc-
axis in the male Syrian ham- Bentley, G. E., Jensen, J. P., Kaur, L., Chowdhury, V. S., Morita, ture of the ovine hypothalamic
ster: the exception proves G. J., Wacker, D. W., Tsutsui, K., Y., Yano, T., et al. (2008). luteinizing hormone-releasing
the rule. Endocrinology 153, and Wingfield, J. C. (2006). Rapid Gonadotropin-inhibitory hor- factor (LRF) (LH-hypothalamus-
1352–1363. inhibition of female sexual behav- mone and its receptor in the avian LRF - gas - chromatography - mass -
Anderson, G. M., Relf, H. L., Rizwan, ior by gonadotropin-inhibitory hor- reproductive system. Gen. Comp. spectrometry - decapeptide - Edman
M. Z., and Evans, J. J. (2009). mone (GnIH). Horm. Behav. 49, Endocrinol. 156, 34–43. degradation). Proc. Natl. Acad. Sci.
Central and peripheral effects of 550–555. Bentley, G. E., Van’t Hof, T. J., and U.S.A. 69, 278–282.
RFamide-related peptide-3 on Bentley, G. E., Perfito, N., Ukena, Ball, G. F. (1999). Seasonal neu- Calisi, R. M., and Bentley, G. E. (2009).
luteinizing hormone and prolactin K., Tsutsui, K., and Wingfield, J. roplasticity in the songbird telen- Lab and field experiments: are they
secretion in rats. Endocrinology 150, C. (2003). Gonadotropin-inhibitory cephalon: a role for melatonin. the same animal? Horm. Behav. 56,
1834–1840. peptide in song sparrows (Melospiza Proc. Natl. Acad. Sci. U.S.A. 96, 1–10.
Bédécarrats, G. Y., McFarlane, H., melodia) in different reproductive 4674–4679. Calisi, R. M., Díaz-Muñoz, S. L.,
Maddineni, S. R., and conditions, and in house spar- Bonini, J. A., Jones, K. A., Adham, Wingfield, J. C., and Bentley, G. E.
Ramachandran, R. (2009). rows (Passer domesticus) relative N., Forray, C., Artymyshyn, R., (2011). Social and breeding status
Gonadotropin-inhibitory hor- to chicken-gonadotropin-releasing Durkin, M. M., et al. (2000). are associated with the expression
mone receptor signaling and its hormone. J. Neuroendocrinol. 15, Identification and characterization of GnIH. Genes Brain Behav. 10,
impact on reproduction in chick- 794–802. of two G protein-coupled receptors 557–564.
ens. Gen. Comp. Endocrinol. 163, Bentley, G. E., Tsutsui, K., and for neuropeptide FF. J. Biol. Chem. Calisi, R. M., Rizzo, N. O., and
7–11. Kriegsfeld, L. J. (2010). Recent 275, 39324-39331. Bentley, G. E. (2008). Seasonal

Frontiers in Neuroscience | Neuroendocrine Science April 2013 | Volume 7 | Article 60 | 8


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

differences in hypothalamic EGR-1 carpalis. Gen. Comp. Endocrinol. Deregnaucourt, S. (2001). Effect of Front. Endocrinol. 2:101. doi:
and GnIH expression follow- 149, 226–235. melatonin supplementation on the 10.3389/fendo.2011.00101
ing capture-handling stress in Dockray, G. J., Reeve, J. R. Jr., Shively, sexual development in European Kriegsfeld, L. J., Gibson, E. M.,
house sparrows (Passer domesti- J., Gayton, R. J., and Barnard, C. quail (Coturnix coturnix). Behav. Williams, W. P. 3rd., Zhao,
cus). Gen. Comp. Endocrinol. 157, S. (1983). A novel active pentapep- Processes 53, 121–130. S., Mason, A. O., Bentley, G.
283–287. tide from chicken brain identified by Hinuma, S., Shintani, Y., Fukusumi, S., E., et al. (2010). The roles of
Caraty, A., Blomenröhr, M., Vogel, antibodies to FMRFamide. Nature Iijima, N., Matsumoto, Y., Hosoya, RFamide-related peptide-3 in
G. M., Lomet, D., Briant, C., and 305, 328–330. M., et al. (2000). New neuropep- mammalian reproductive function
Beltramo, M. (2012). RF9 pow- Ducret, E., Anderson, G. M., and tides containing carboxy-terminal and behaviour. J. Neuroendocrinol.
erfully stimulates gonadotrophin Herbison, A. E. (2009). RFamide- RFamide and their receptor 22, 692–700.
secretion in the ewe: evidence for related peptide-3, a mammalian in mammals. Nat. Cell Biol. 2, Kriegsfeld, L. J., Mei, D. F., Bentley, G.
a seasonal threshold of sensitivity. gonadotropin-inhibitory hormone 703–708. E., Ubuka, T., Mason, A. O., Inoue,
J. Neuroendocrinol. 24, 725–736. ortholog, regulates gonadotropin- Jimenez-Liñan, M., Rubin, B. S., and K., et al. (2006). Identification and
Chowdhury, V. S., Yamamoto, releasing hormone neuron firing King, J. C. (1997). Examination characterization of a gonadotropin-
K., Ubuka, T., Bentley, G. E., in the mouse. Endocrinology 150, of guinea pig luteinizing hormone- inhibitory system in the brains of
Hattori, A., and Tsutsui, K. (2010). 2799–2804. releasing hormone gene reveals a mammals. Proc. Natl. Acad. Sci.
Melatonin stimulates the release Foster, R. G., Panzica, G. C., Parry, unique decapeptide and existence U.S.A. 103, 2410–2415.
of gonadotropin-inhibitory hor- D. M., and Viglietti-Panzica, C. of two transcripts in the brain. Lee, Y. R., Tsunekawa, K., Moon,
mone by the avian hypothalamus. (1988). Immunocytochemical stud- Endocrinology 138, 4123–4130. M. J., Um, H. N., Hwang, J. I.,
Endocrinology 151, 271–280. ies on the LHRH system of the Johnson, M. A., Tsutsui, K., and Fraley, Osugi, T., et al. (2009). Molecular
Ciccone, N. A., Dunn, I. C., Boswell, Japanese quail: influence by pho- G. S. (2007). Rat RFamide-related evolution of multiple forms of
T., Tsutsui, K., Ubuka, T., Ukena, toperiod and aspects of sexual dif- peptide-3 stimulates GH secretion, kisspeptins and GPR54 receptors
K., et al. (2004). Gonadotrophin ferentiation. Cell Tissue Res. 253, inhibits LH secretion, and has vari- in vertebrates. Endocrinology 150,
inhibitory hormone depresses 327–335. able effects on sex behavior in the 2837–2846.
gonadotrophin alpha and follicle- Fukusumi, S., Habata, Y., Yoshida, H., adult male rat. Horm. Behav. 51, Maddineni, S., Ocón-Grove, O. M.,
stimulating hormone beta Iijima, N., Kawamata, Y., Hosoya, 171–180. Krzysik-Walker, S. M., Hendricks,
subunit expression in the pitu- M., et al. (2001). Characteristics Juss, T. S., Meddle, S. L., Servant, R. S., G. L. 3rd., Proudman, J. A.,
itary of the domestic chicken. and distribution of endoge- and King, V. M. (1993). Melatonin and Ramachandran, R. (2008).
J. Neuroendocrinol. 16, 999–1006. nous RFamide-related peptide-1. and photoperiodic time measure- Gonadotrophin-inhibitory hor-
Clarke, I. J., Qi, Y., Puspita Sari, Biochim. Biophys. Acta 1540, ment in Japanese quail (Coturnix mone receptor expression in the
I., and Smith, J. T. (2009). 221–232. coturnix japonica). Proc. Biol. Sci. chicken pituitary gland: potential
Evidence that RF-amide related Gibson, E. M., Humber, S. A., Jain, 254, 21–28. influence of sexual maturation and
peptides are inhibitors of repro- S., Williams, W. P. 3rd., Zhao, Kadokawa, H., Shibata, M., Tanaka, Y., ovarian steroids. J. Neuroendocrinol.
duction in mammals. Front. S., Bentley, G. E., et al. (2008). Kojima, T., Matsumoto, K., Oshima, 20, 1078–1088.
Neuroendocrinol. 30, 371–378. doi: Alterations in RFamide-related pep- K., et al. (2009). Bovine C-terminal Mason, A. O., Duffy, S., Zhao, S.,
10.1016/j.yfrne.2009.04.001 tide expression are coordinated with octapeptide of RFamide-related Ubuka, T., Bentley, G. E., Tsutsui,
Clarke, I. J., Sari, I. P., Qi, Y., Smith, the preovulatory luteinizing hor- peptide-3 suppresses luteinizing K., et al. (2010). Photoperiod
J. T., Parkington, H. C., Ubuka, mone surge. Endocrinology 149, hormone (LH) secretion from the and reproductive condition
T., et al. (2008). Potent action 4958–4969. pituitary as well as pulsatile LH are associated with changes in
of RFamide-related peptide-3 on Gingerich, S., Wang, X., Lee, P. K., secretion in bovines. Domest. Anim. RFamide-related peptide (RFRP)
pituitary gonadotropes indicative Dhillon, S. S., Chalmers, J. A., Endocrinol. 36, 219–224. expression in Syrian hamsters
of a hypophysiotropic role in the Koletar, M. M., et al. (2009). The Kang, S. W., Thayananuphat, A., (Mesocricetus auratus). J. Biol.
negative regulation of gonadotropin generation of an array of clonal, Bakken, T., and El Halawani, M. Rhythms. 25, 176–185.
secretion. Endocrinology 149, immortalized cell models from E. (2007). Dopamine-melatonin Matsuo, H., Baba, Y., Nair, R. M.,
5811–5821. the rat hypothalamus: analysis of neurons in the avian hypothalamus Arimura, A., and Schally, A. V.
Cockrem, J. F., and Follett, B. K. (1985). melatonin effects on kisspeptin controlling seasonal reproduction. (1971). Structure of the porcine
Circadian rhythm of melatonin in and gonadotropin-inhibitory hor- Neuroscience 150, 223–233. LH- and FSH-releasing hormone. I.
the pineal gland of the Japanese mone neurons. Neuroscience 162, King, J. A., and Millar, R. P. (1982). The proposed amino acid sequence.
quail (Coturnix coturnix japonica). 1134–1140. Structure of chicken hypothalamic Biochem. Biophys. Res. Commun. 43,
J. Endocrinol. 107, 317–324. Greives, T. J., Kingma, S. A., Beltrami, luteinizing hormone-releasing hor- 1334–1339.
Dardente, H., Birnie, M., Lincoln, G. G., and Hau, M. (2012). Melatonin mone. I. Structural determination McGuire, N. L., and Bentley, G. E.
A., and Hazlerigg, D. G. (2008). delays clutch initiation in a wild on partially purified material. J. Biol. (2010a). A functional neuropep-
RFamide-related peptide and its songbird. Biol. Lett. 8, 330–332. Chem. 257, 10722–10728. tide system in vertebrate gonads:
cognate receptor in the sheep: cDNA Greives, T. J., Kriegsfeld, L. J., Kirby, E. D., Geraghty, A. C., Ubuka, gonadotropin-inhibitory hormone
cloning, mRNA distribution in the and Demas, G. E. (2008). T., Bentley, G. E., and Kaufer, and its receptor in testes of field-
hypothalamus and the effect of pho- Exogenous kisspeptin does not D. (2009). Stress increases putative caught house sparrow (Passer
toperiod. J. Neuroendocrinol. 20, alter photoperiod-induced gonadal gonadotropin inhibitory hormone domesticus). Gen. Comp. Endocrinol.
1252–1259. regression in Siberian hamsters and decreases luteinizing hormone 166, 565–572.
Dawson, A., King, V. M., Bentley, (Phodopus sungorus). Gen. Comp. in male rats. Proc. Natl. Acad. Sci. McGuire, N. L., and Bentley, G. E.
G. E., and Ball, G. F. (2001). Endocrinol. 156, 552–558. U.S.A. 106, 11324–11329. (2010b). Neuropeptides in the
Photoperiodic control of seasonal- Greives, T. J., Mason, A. O., Scotti, Klingerman, C. M., Williams, W. P. gonads: from evolution to phar-
ity in birds. J. Biol. Rhythms 16, M. A., Levine, J., Ketterson, 3rd., Simberlund, J., Brahme, N., macology. Front. Pharmacol. 1:114.
365–380. E. D., Kriegsfeld, L. J., et al. Prasad, A., Schneider, J. E., et al. doi: 10.3389/fphar.2010.00114
Deviche, P., Small, T., Sharp, P., and (2007). Environmental control of (2011). Food restriction-induced Miyamoto, K., Hasegawa, Y., Minegishi,
Tsutsui, K. (2006). Control of kisspeptin: implications for seasonal changes in gonadotropin-inhibiting T., Nomura, M., Takahashi, Y.,
luteinizing hormone and testos- reproduction. Endocrinology 148, hormone cells are associated with Igarashi, M., et al. (1982). Isolation
terone secretion in a flexibly 1158–1166. changes in sexual motivation and and characterization of chicken
breeding male passerine, the Guyomarc’h, C., Lumineau, S., food hoarding, but not sexual hypothalamic luteinizing hormone-
Rufous-winged Sparrow, Aimophila Vivien-Roels, B., Richard, J., and performance and food intake. releasing hormone. Biochem.

www.frontiersin.org April 2013 | Volume 7 | Article 60 | 9


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

Biophys. Res. Commun. 107, gonadotrophin secretion in ovariec- luteinizing hormone and follicle- (Aimophila carpalis). Brain Behav.
820–827. tomised ewes. J. Neuroendocrinol. stimulating hormone synthesis Evol. 71, 127–142.
Miyamoto, K., Hasegawa, Y., Nomura, 23, 208–215. and secretion in ovine pituitary Smith, J. T., and Clarke, I. J. (2010).
M., Igarashi, M., Kangawa, K., and Pineda, R., Garcia-Galiano, D., gonadotropes. Endocrinology 150, Gonadotropin inhibitory hormone
Matsuo, H. (1984). Identification of Sanchez-Garrido, M. A., Romero, 5549–5556. function in mammals. Trends
the second gonadotropin-releasing M., Ruiz-Pino, F., Aguilar, E., et al. Satake, H., Hisada, M., Kawada, T., Endocrinol. Metab. 21, 255–260.
hormone in chicken hypothala- (2010a). Characterization of the Minakata, H., Ukena, K., and Smith, J. T., Coolen, L. M., Kriegsfeld,
mus: evidence that gonadotropin inhibitory roles of RFRP3, the Tsutsui, K. (2001). Characterization L. J., Sari, I. P., Jaafarzadehshirazi,
secretion is probably controlled mammalian ortholog of GnIH, in of a cDNA encoding a novel M. R., Maltby, M., et al. (2008).
by two distinct gonadotropin- the control of gonadotropin secre- avian hypothalamic neuropeptide Variation in kisspeptin and
releasing hormones in avian species. tion in the rat: in vivo and in vitro exerting an inhibitory effect on RFamide-related peptide (RFRP)
Proc. Natl. Acad. Sci. U.S.A. 81, studies. Am. J. Physiol. Endocrinol. gonadotropin release. Biochem. expression and terminal connec-
3874–3878. Metab. 299, E39–E46. J. 354, 379–385. tions to gonadotropin-releasing
Moussavi, M., Wlasichuk, M., Chang, Pineda, R., Garcia-Galiano, D., Sharp, P. J., Talbot, R. T., Main, G. hormone neurons in the brain: a
J. P., and Habibi, H. R. (2012). Sanchez-Garrido, M. A., Romero, M., Dunn, I. C., Fraser, H. M., novel medium for seasonal breeding
Seasonal effect of GnIH on M., Ruiz-Pino, F., Aguilar, E., et al. and Huskisson, N. S. (1990). in the sheep. Endocrinology 149,
gonadotrope functions in the (2010b). Characterization of the Physiological roles of chicken 5770–5782.
pituitary of goldfish. Mol. Cell. potent gonadotropin-releasing LHRH-I and -II in the control Smith, J. T., Young, I. R., Veldhuis,
Endocrinol. 350, 53–60. activity of RF9, a selective antago- of gonadotrophin release in the J. D., and Clarke, I. J. (2012).
Murakami, M., Matsuzaki, T., Iwasa, nist of RF-amide-related peptides domestic chicken. J. Endocrinol. Gonadotropin-inhibitory hor-
T., Yasui, T., Irahara, M., Osugi, T., and neuropeptide FF receptors: 124, 291–299. mone (GnIH) secretion into the
et al. (2008). Hypophysiotropic role physiological and pharmacological Sherwood, N., Eiden, L., Brownstein, ovine hypophyseal portal system.
of RFamide-related peptide-3 in the implications. Endocrinology 151, M., Spiess, J., Rivier, J., and Vale, Endocrinology 153, 3368–3375.
inhibition of LH secretion in female 1902–1913. W. (1983). Characterization of a Son, Y. L., Ubuka, T., Millar, R. P.,
rats. J. Endocrinol. 199, 105–112. Qi, Y., Oldfield, B. J., and Clarke, I. J. teleost gonadotropin-releasing hor- Kanasaki, H., and Tsutsui, K.
Ohta, M., Kadota, C., and Konishi, H. (2009). Projections of RFamide- mone. Proc. Natl. Acad. Sci. U.S.A. (2012). Gonadotropin-inhibitory
(1989). A role of melatonin in the related peptide-3 neurones in the 80, 2794–2798. hormone inhibits GnRH-
initial stage of photoperiodism in ovine hypothalamus, with special Sherwood, N. M., Sower, S. A., induced gonadotropin subunit
the Japanese quail. Biol. Reprod. 40, reference to regions regulating Marshak, D. R., Fraser, B. A., and gene transcriptions by inhibit-
935–941. energy balance and reproduction. Brownstein, M. J. (1986). Primary ing AC/cAMP/PKA-dependent
Oishi, H., Klausen, C., Bentley, G. J. Neuroendocrinol. 21, 690–697. structure of gonadotropin-releasing ERK pathway in LβT2 cells.
E., Osugi, T., Tsutsui, K., Gilks, Revel, F. G., Saboureau, M., Masson- hormone in lamprey brain. J. Biol. Endocrinology 153, 2332–2343.
C. B., et al. (2012). The human Pévet, M., Pévet, P., Mikkelsen, J. Chem. 15, 4812–4819. Tachibana, T., Sato, M., Takahashi,
gonadotropin-inhibitory hormone D., and Simonneaux, V. (2006). Shimizu, M., and Bédécarrats, G. H., Ukena, K., Tsutsui, K., and
ortholog RFamide-related peptide- Kisspeptin mediates the photope- Y. (2006). Identification of a Furuse, M. (2005). Gonadotropin-
3 suppresses gonadotropin-induced riodic control of reproduction in novel pituitary-specific chicken inhibiting hormone stimulates feed-
progesterone production in human hamsters. Curr. Biol. 16, 1730–1735. gonadotropin-releasing hormone ing behavior in chicks. Brain Res.
granulosa cells. Endocrinology 153, Revel, F. G., Saboureau, M., Pévet, P., receptor and its splice variants. Biol. 1050, 94–100.
3435–3445. Simonneaux, V., and Mikkelsen, J. Reprod. 75, 800–808. Tobari, Y., Iijima, N., Tsunekawa, K.,
Okubo, K., Amano, M., Yoshiura, Y., D. (2008). RFamide-related peptide Shimizu, M., and Bédécarrats, G. Y. Osugi, T., Okanoya, K., Tsutsui,
Suetake, H., and Aida, K. (2000a). gene is a melatonin-driven pho- (2010). Activation of the chicken K., et al. (2010). Identification of
A novel form of gonadotropin- toperiodic gene. Endocrinology 149, gonadotropin-inhibitory hormone gonadotropin-inhibitory hormone
releasing hormone in the medaka, 902–912. receptor reduces gonadotropin in the zebra finch (Taeniopygia
Oryzias latipes. Biochem. Biophys. Rizwan, M. Z., Poling, M. C., Corr, releasing hormone receptor signal- guttata): peptide isolation, cDNA
Res. Commun. 16, 298–303. M., Cornes, P. A., Augustine, R. ing. Gen. Comp. Endocrinol. 167, cloning and brain distribution.
Okubo, K., Suetake, H., Usami, T., A., Quennell, J. H., et al. (2012). 331–337. Peptides 31, 816–826.
and Aida, K. (2000b). Molecular RFamide-related peptide-3 receptor Simonneaux, V., Bur, I., Ancel, C., Tsutsui, K. (2009). Review: a new key
cloning and tissue-specific expres- gene expression in GnRH and Ansel, L., and Klosen, P. (2012). A neurohormone controlling repro-
sion of a gonadotropin-releasing kisspeptin neurons and GnRH- kiss for daily and seasonal reproduc- duction, gonadotropin-inhibitory
hormone receptor in the Japanese dependent mechanism of action. tion. Prog. Brain Res. 199, 423–437. hormone (GnIH): biosynthesis,
eel. Gen. Comp. Endocrinol. 119, Endocrinology 153, 3770–3779. Singh, P., Krishna, A., Sridaran, mode of action and functional
181–192. Rizwan, M. Z., Porteous, R., Herbison, R., and Tsutsui, K. (2011a). significance. Prog. Neurobiol. 88,
Osugi, T., Ukena, K., Bentley, G. A. E., and Anderson, G. M. (2009). Immunohistochemical localization 76–88.
E., O’Brien, S., Moore, I. T., Cells expressing RFamide-related of GnRH and RFamide-related Tsutsui, K., Bentley, G. E., Bedecarrats,
Wingfield, J. C., et al. (2004). peptide-1/3, the mammalian peptide-3 in the ovaries of mice G., Osugi, T., Ubuka, T., and
Gonadotropin-inhibitory hor- gonadotropin-inhibitory hormone during the estrous cycle. J. Mol. Kriegsfeld, L. J. (2010a). Review:
mone in Gambel’s white-crowned orthologs, are not hypophysiotropic Histol. 42, 371–381. gonadotropin-inhibitory hor-
sparrow (Zonotrichia leucophrys neuroendocrine neurons in the rat. Singh, P., Krishna, A., and Tsutsui, K. mone (GnIH) and its control
gambelii): cDNA identification, Endocrinology 150, 1413–1420. (2011b). Effects of gonadotropin- of central and peripheral
transcript localization and func- Rozenboim, I., Aharony, T., and Yahav, inhibitory hormone on folliculoge- reproductive function. Front.
tional effects in laboratory and field S. (2002). The effect of mela- nesis and steroidogenesis of cyclic Neuroendocrinol. 31, 284–295. doi:
experiments. J. Endocrinol. 182, tonin administration on circulat- mice. Fertil. Steril. 95, 1397–1404. 10.1016/j.yfrne.2010.03.001
33–42. ing plasma luteinizing hormone Small, T. W., Sharp, P. J., Bentley, Tsutsui, K., Bentley, G. E., Kriegsfeld,
Papargiris, M. M., Rivalland, E. T., concentration in castrated White G. E., Millar, R. P., Tsutsui, K., L. J., Osugi, T., Seong, J. Y., and
Clarke, I. J., Smith, J. T., Pereira, A., Leghorn roosters. Poult. Sci. 81, Mura, E., et al. (2007). Photoperiod- Vaudry, H. (2010b). Review:
and Tilbrook, A. J. (2011). Evidence 1354–1359. independent hypothalamic regula- discovery and evolutionary his-
that RF-amide related peptide-3 is Sari, I. P., Rao, A., Smith, J. T., Tilbrook, tion of luteinizing hormone secre- tory of gonadotrophin-inhibitory
not a mediator of the inhibitory A. J., and Clarke, I. J. (2009). Effect tion in a free-living sonoran desert hormone and kisspeptin: new
effects of psychosocial stress on of RF-amide-related peptide-3 on bird, the Rufous-winged Sparrow key neuropeptides controlling

Frontiers in Neuroscience | Neuroendocrine Science April 2013 | Volume 7 | Article 60 | 10


Tsutsui et al. Regulatory mechanisms of GnIH synthesis and release

reproduction. J. Neuroendocrinol. K. (2005). Melatonin induces (2006). Gonadotropin-inhibitory hormone ([Trp8] GnRH) in frog
22, 716–727. the expression of gonadotropin- hormone inhibits gonadal devel- brain. Mol. Cell. Endocrinol. 164,
Tsutsui, K., Bentley, G. E., and Ciccone, inhibitory hormone in the avian opment and maintenance by 197–204.
N. (2005). “Structure, action and brain. Proc. Natl. Acad. Sci. U.S.A. decreasing gonadotropin syn- Yoshida, H., Habata, Y., Hosoya, M.,
functional significance of GnIH,” in 102, 3052–3057. thesis and release in male quail. Kawamata, Y., Kitada, C., and
Functional Avian Endocrinology, eds Ubuka, T., Inoue, K., Fukuda, Y., Endocrinology 147, 1187–1194. Hinuma, S. (2003). Molecular
A. Dawson and P. J. Sharp (New Mizuno, T., Ukena, K., Kriegsfeld, Ukena, K., Iwakoshi, E., Minakata, H., properties of endogenous RFamide-
Delhi: Narosa Publishing House), L. J., et al. (2012a). Identification, and Tsutsui, K. (2002). A novel related peptide-3 and its interaction
73–82. expression, and physiological rat hypothalamic RFamide-related with receptors. Biochim. Biophys.
Tsutsui, K., Bentley, G. E., Ubuka, functions of Siberian hamster peptide identified by immunoaffin- Acta 1593, 151–157.
T., Saigoh, E., Yin, H., Osugi, T., gonadotropin-inhibitory hormone. ity chromatography and mass Zhang, Y., Li, S., Liu, Y., Lu, D.,
et al. (2007a). Review: the gen- Endocrinology 153, 373–385. spectrometry. FEBS Lett. 512, Chen, H., Huang, X., et al.
eral and comparative biology of Ubuka, T., Mukai, M., Wolfe, J., 255–258. (2010). Structural diversity of
gonadotropin-inhibitory hormone Beverly, R., Clegg, S., Wang, A., Ukena, K., and Tsutsui, K. (2005). A the GnIH/GnIH receptor system
(GnIH). Gen. Comp. Endocrinol. et al. (2012b). RNA interference new member of the hypothalamic in teleost: its involvement in early
153, 365–370. of gonadotropin-inhibitory hor- RF-amide peptide family, LPXRF- development and the negative
Tsutsui, K., Ubuka, T., Yin, H., Ukena, mone gene induces arousal in amide peptides: structure, localiza- control of LH release. Peptides 31,
K., Bentley, G., Sharp, P., et al. songbirds. PLoS ONE 7:e30202. doi: tion, and function. Mass. Spectrom. 1034–1043.
(2007b). Review: discovery of 10.1371/journal.pone.0030202 Rev. 24, 469–486. Zhao, S., Zhu, E., Yang, C., Bentley, G.
gonadotropin-inhibitory hormone Ubuka, T., Kim, S., Huang, Y. C., Reid, Ukena, K., Ubuka, T., and Tsutsui, E., Tsutsui, K., and Kriegsfeld, L.
in a domesticated bird, and its J., Jiang, J., Osugi, T., et al. (2008a). K. (2003). Distribution of a novel J. (2010). RFamide-related peptide
mode of action and functional Gonadotropin-inhibitory hormone avian gonadotropin-inhibitory hor- and messenger ribonucleic acid
significance. J. Ornithol. 147, 53–54. neurons interact directly with mone in the quail brain. Cell Tissue expression in mammalian testis:
Tsutsui, K., Saigoh, E., Ukena, K., gonadotropin-releasing hormone- Res. 312, 73–79. association with the spermato-
Teranishi, H., Fujisawa, Y., Kikuchi, I and -II neurons in European Underwood, H., Binkley, S., Siopes, T., genic cycle. Endocrinology 151,
M., et al. (2000). A novel avian starling brain. Endocrinology 149, and Mosher, K. (1984). Melatonin 617–627.
hypothalamic peptide inhibiting 268–278. rhythms in the eyes, pineal bod-
gonadotropin release. Biochem. Ubuka, T., McGuire, N. L., Calisi, R. ies, and blood of Japanese quail Conflict of Interest Statement: The
Biophys. Res. Commun. 275, M., Perfito, N., and Bentley, G. (Coturnix coturnix japonica). Gen. authors declare that the research
661–667. E. (2008b). The control of repro- Comp. Endocrinol. 56, 70–81. was conducted in the absence of any
Tsutsui, K., and Ubuka, T. (2013). ductive physiology and behavior by Wilson, F. E. (1991). Neither retinal commercial or financial relationships
“Gonadotropin-inhibitory hor- gonadotropin-inhibitory hormone. nor pineal photoreceptors mediate that could be construed as a potential
mone,” in Handbook of Biologically Integr. Comp. Biol. 48, 560–569. photoperiodic control of seasonal conflict of interest.
Active Peptides. Section on Brain Ubuka, T., Lai, H., Kitani, M., reproduction in American tree spar-
Peptides, eds A. J. Kastin and H. Suzuuchi, A., Pham, V., Cadigan, P. rows (Spizella arborea). J. Exp. Zool. Received: 27 November 2012; paper
Vaudry (London: Academic Press), A., et al. (2009a). Gonadotropin- 259, 117–127. pending published: 01 February 2013;
802–811. inhibitory hormone identification, Wu, M., Dumalska, I., Morozova, E., accepted: 01 April 2013; published
Tsutsui, K., Ubuka, T., Bentley, G. E., cDNA cloning, and distribution van den Pol, A. N., and Alreja, M. online: 16 April 2013.
and Kriegsfeld, L. J. (2012). Review: in rhesus macaque brain. J. Comp. (2009). Gonadotropin inhibitory Citation: Tsutsui K, Ubuka T, Bentley
gonadotropin-inhibitory hormone Neurol. 517, 841–855. hormone inhibits basal forebrain GE and Kriegsfeld LJ (2013) Review:
(GnIH): discovery, progress and Ubuka, T., Morgan, K., Pawson, vGluT2 - gonadotropin - releasing regulatory mechanisms of gonadotropin-
prospect. Gen. Comp. Endocrinol. A. J., Osugi, T., Chowdhury, V. hormone neurons via a direct inhibitory hormone (GnIH) synthesis
177, 305–314. S., Minakata, H., et al. (2009b). postsynaptic mechanism. J. Physiol. and release in photoperiodic animals.
Tsutsui, K., Ubuka, T., Yin, H., Osugi, Identification of human GnIH 587(Pt 7), 1401–1411. Front. Neurosci. 7:60. doi: 10.3389/fnins.
T., Ukena, K., Bentley, G. E., homologs, RFRP-1 and RFRP-3, Yin, H., Ukena, K., Ubuka, T., and 2013.00060
et al. (2006). Mode of action and and the cognate receptor, GPR147 Tsutsui, K. (2005). A novel G This article was submitted to Frontiers
functional significance of avian in the human hypothalamic pitu- protein-coupled receptor for in Neuroendocrine Science, a specialty of
gonadotropin-inhibitory hormone itary axis. PLoS ONE 4:e8400. doi: gonadotropin-inhibitory hormone Frontiers in Neuroscience.
(GnIH): a review. J. Exp. Zool. 305A, 10.1371/journal.pone.0008400 in the Japanese quail (Coturnix Copyright © 2013 Tsutsui, Ubuka,
801–806. Ubuka, T., Ueno, M., Ukena, K., and japonica): identification, expression Bentley and Kriegsfeld. This is an open-
Tsutsui, K., and Ukena, K. (2006). Tsutsui, K. (2003). Developmental and binding activity. J. Endocrinol. access article distributed under the terms
Review: hypothalamic LPXRF- changes in gonadotropin-inhibitory 184, 257–266. of the Creative Commons Attribution
amide peptides in vertebrates: hormone in the Japanese quail Yoo, M. S., Kang, H. M., Choi, H. S., License, which permits use, distribution
identification, localization and (Coturnix japonica) hypothalamo- Kim, J. W., Troskie, B. E., Millar, and reproduction in other forums, pro-
hypophysiotropic activity. Peptides hypophysial system. J. Endocrinol. R. P., et al. (2000). Molecular vided the original authors and source
27, 1121–1129. 178, 311–318. cloning, distribution and phar- are credited and subject to any copy-
Ubuka, T., Bentley, G. E., Ukena, Ubuka, T., Ukena, K., Sharp, P. J., macological characterization of right notices concerning any third-party
K., Wingfield, J. C., and Tsutsui, Bentley, G. E., and Tsutsui, K. a novel gonadotropin-releasing graphics etc.

www.frontiersin.org April 2013 | Volume 7 | Article 60 | 11

You might also like