Sei sulla pagina 1di 14

bs_bs_banner

Animal Science Journal (2013) 84, 369–381 doi: 10.1111/asj.12056

REV IEW AR TICLE


Kisspeptin as a master player in the central control of
reproduction in mammals: An overview of kisspeptin
research in domestic animals
Hiroaki OKAMURA, Takashi YAMAMURA and Yoshihiro WAKABAYASHI
Animal Physiology Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan

ABSTRACT
The hypothalamo-pituitary-gonadal (HPG) axis is the regulatory system for reproduction in mammals. Because secretion of
gonadotropin-releasing hormone (GnRH) into the portal vessels is the final step at which the brain controls gonadal
activities, the GnRH neuronal system had been thought to be central to the HPG axis. A newly discovered neural peptide,
kisspeptin, has opened a new era in reproductive neuroendocrinology. As shown in a variety of mammals, kisspeptin is a
potent endogenous secretagogue of GnRH, and the kisspeptin neuronal system governs both the pulsatile GnRH secretion
that drives folliculogenesis, spermatogenesis and steroidogenesis, and the GnRH surge that triggers ovulation in females.
The kisspeptin neuronal system is therefore considered a master player in the central control of mammalian reproduction,
and kisspeptin and related substances could therefore be valuable for the development of novel strategies for the
management of fertility in farm animals. To this end, the present review aimed to summarize the current research on
kisspeptin signaling with a focus on domestic animals such as sheep, goats, cattle, pigs and horses.

Key words: domestic animals, GnRH, kisspeptin, neurokinin B, reproduction.

INTRODUCTION resolved. For example, how are the two modes of


A subset of neurons in the hypothalamus synthesizes GnRH secretion generated, and how are steroid
gonadotropin-releasing hormone (GnRH) and releases hormone signals transmitted to GnRH neurons that
it into the portal vessels, which is the final mechanism lack estrogen receptor alpha (ERa) mediating the feed-
by which the brain controls gonadal activities. GnRH back action of estradiol (E2) on GnRH secretion? A
acts on the anterior pituitary gland to stimulate the newly discovered peptide, kisspeptin, provides essen-
secretion of the gonadotropins, luteinizing hormone tial clues to answering these long-lasting questions
(LH) and follicle-stimulating hormone (FSH), which and has opened a new era in reproductive endocrinol-
mediate the control of the gonads by the brain. In ogy. Here, we describe the history, anatomy and physi-
response to gonadotropins, the gonad releases steroid ology of kisspeptin and the kisspeptin neuronal system
hormones, such as estrogen and progesterone in and then offer a brief prospective for the potential
females and androgens in males, which in turn feed applications of kisspeptin and related substances to
back to the hypothalamus to alter GnRH secretion. The domestic animal production. As a number of excellent
reproductive system is regulated by this circuit, the general reviews of kisspeptin have appeared elsewhere
so-called hypothalamo-pituitary-gonadal (HPG) axis (Maeda et al. 2007; Oakley et al. 2009; Ohkura et al.
(Karsch 1984). There are two modes of GnRH secre- 2009a; Clarke 2011; Goodman & Lehman 2012; Smith
tion: the pulsatile mode, tonic gonadotropin secretion 2012; Caraty et al. 2012a), this review aimed mainly to
that drives folliculogenesis, spermatogenesis and ster- summarize the knowledge accumulated in domestic
oidogenesis, and is controlled by negative feedback via
gonadal steroids, and the surge mode, the preovula-
tory gonadotropin discharge that induces ovulation in Correspondence: Hiroaki Okamura, Animal Physiology
Research Unit, National Institute of Agrobiological Sciences,
females in response to positive feedback via estrogen. 2 Ikenodai, Tsukuba, Ibaraki 305-0901, Japan. (Email:
Although the above-described scenario had been hokamu@affrc.go.jp)
accepted as the central dogma of the control of repro- Received 20 November 2012; accepted for publication 16
duction, several critical issues had remained to be January 2013.

© 2013 Japanese Society of Animal Science


370 H. OKAMURA et al.

animals such as sheep, goats, cattle, pigs and horses. it should be noted that full-length kisspeptin, either rat
Chickens are not included because it is generally type (52 residues; Pheng et al. 2009) or human type
accepted that kisspeptin signaling is absent in birds. (54 residues; Kinoshita et al. 2005), seems to have
more potent biological activity than Kp-10 in rats.
DISCOVERY OF KISSPEPTIN
The Kiss1 gene was initially isolated as a tumor metas- BIOLOGICAL ACTIONS OF KISSPEPTIN
tasis gene by a group in Hershey, PA, USA, (Lee et al. Since the initial observations in rodents (Gottsch et al.
1996), the home of the famous Hershey’s Kiss choco- 2004; Matsui et al. 2004), the effects of centrally or
lates. Thereafter, the Kiss1 gene product, kisspeptin, peripherally administered kisspeptin on gonadotropin
was discovered to be an endogenous ligand for the secretion have been investigated in a variety of
G-protein-coupled receptor, GPR54 (Kotani et al. mammals, including domestic animals. Kisspeptin
2001; Ohtaki et al. 2001). In 2003, two groups inde- consistently stimulates gonadotropin, namely LH,
pendently found that inactivating mutations of the secretion in all mammalian species examined so far,
kisspeptin receptor gene (GPR54) were associated although its efficacy seems to depend on the species,
with idiopathic hypogonadotropic hypogonadism in age, sex, steroidal milieu, season and route of admin-
humans (de Roux et al. 2003; Seminara et al. 2003). istration involved. Several studies have indicated that
This human genetic finding was substantiated experi- kisspeptin has less effect on FSH secretion than on LH
mentally by the generation of GPR54 null mice, which secretion (Caraty et al. 2007; Lents et al. 2008; Smith
exhibit reduced gonadal size and low levels of gona- et al. 2009a).
dotropins and steroid hormones, and fail to undergo
puberty (Funes et al. 2003; Seminara et al. 2003; Stimulatory effects of kisspeptin on
Messager et al. 2005). These studies established gonadotropin secretion
kisspeptin/GPR54 signaling as a new player in the HPG Table 2 summarizes effective doses of kisspeptin on
axis and thus led to the intensive and extensive gonadotropin secretion in domestic animals, and a
research on kisspeptin performed over the past decade. representative profile of LH secretion induced by
kisspeptin administration in a steer is shown in
AMINO ACID SEQUENCE Figure 1. LH levels increased robustly after bolus intra-
OF KISSPEPTIN venous (i.v.) administration of Kp-10 and peaked
The Kiss1 gene encodes a precursor peptide that is 30 min after the injection. The stimulatory effect of
cleaved to produce the biologically active form of Kp-10 on LH secretion lasted for approximately 1 h.
kisspeptin, which is 52–54 amino acids long in most Ezzat et al. (2009) reported that i.v. injection of as low
mammals (Oakley et al. 2009). As shown in Table 1 as 3.9 nmol of Kp-10 per kg of body weight signifi-
(Ohkura et al. 2009b; Tomikawa et al. 2010), the cantly increased plasma LH and FSH concentrations in
deduced amino acid sequence of kisspeptin is well prepubertal calves of both sexes and that the same
conserved across mammals. For example, the dose of Kp-10 was also effective, although slightly less
sequence of goat kisspeptin shows 98%, 91% and potent, when administered intramuscularly to male
77% sequence identity to ovine, bovine, and porcine calves. Whitlock et al. (2008, 2010) investigated the
kisspeptin, respectively. In particular, the C-terminal effect of Kp-10 administration on LH secretion in adult
10 residues, which are the minimal core sequence for ovariectomized (OVX) cows. Bolus i.v. injection of
maximal receptor activation (Kotani et al. 2001; Kp-10 stimulated LH secretion, and a similar effect was
Ohtaki et al. 2001), are identical among the species observed in OVX cows treated with estradiol cypionate
listed with the exception that the C-terminal tyrosine and/or progesterone (Whitlock et al. 2008). Intrave-
(Y) is replaced by phenylalanine (F) in primates. nous infusion of Kp-10 over 5 h in OVX cows
The C-terminal decapeptide of kisspeptin, Kp-10, is increased plasma LH concentrations for as long as
commercially available and is routinely used to inves- 140 min following the start of infusion, but the stimu-
tigate the physiological actions of kisspeptin in verte- latory action of the peptide was not observed thereaf-
brates, including fishes (Kanda et al. 2008). However, ter (Whitlock et al. 2010).

Table 1 Comparison of amino acid sequence organization of kisspeptin among different species
Goat 1 GAALCPS–ESSAGPRQPGPCAPRSRLIPAPRGAVLVQREKDVSAYNWNSFGLRY 53
Sheep 1 GAALCPS–ESSAGPRQPGPCAPRSRLIPAPRGAALVQREKDVSAYNWNSFGLRY 53
Cattle 1 GAALCPP–ESSAGPQRLGPCAPRSRLIPSPRGAVLVQREKDVSAYNWNSFGLRY 53
Pig 1 GTSSCQPPESSSGPQRPGLCTPRSRLIPAPRGAVLVQREKDLSAYNWNSFGLRY 54
Rat 1 –TSPCPPVENPTGHQRP–PCATRSRLIPAPRGSVLVQREKDMSAYNWNSFGLRY 52
Human 1 GTSLSPPPESSGSPQQPGLSAPHSRQIPAPQGAVLVQREKDLPNYNWNSFGLRF 54
—Kp–10—

© 2013 Japanese Society of Animal Science Animal Science Journal (2013) 84, 369–381
KISSPEPTIN IN DOMESTIC ANIMALS 371

Smith et al. (2009a, 2011)

Hashizume et al. (2010)


Whitlock et al. (2008)
Whitlock et al. (2008)

Ohkura et al. (2009b)


Smith et al. (2009a)

Magee et al. (2009)


Caraty et al. (2007)

Lents et al. (2008)


Ezzat et al. (2009)

Saito et al. (2012)


Reference

15.6 or 39 nmol/head
Effective doses of bolus i.v. administration on Kp-10 on stimulating luteinizing hormone (LH) release in various domestic animals
Dose of kisspeptin

0.77 nmol/kg BW
3.9 nmol/kg BW
0.1 nmol/kg BW
0.1 nmol/kg BW

3.9 nmol/kg BW
15.6 nmol/head

390 nmol/head

780 nmol/head
390 nmol/head
6 nmol/head

Figure 1 A representative luteinizing hormone (LH) profile


after bolus i.v. injection of the kisspeptin decapeptide Kp-10
in a steer.
Gonadal status and steroid milieu

In OVX adult ewes treated with E2, i.v. injection of


Kp-10 during the anestrous season significantly stimu-
lated LH and FSH secretion, and the human (hKp-10)
Intact (breeding season)

OVX + E2 (anestrous)

and rat (rKp-10) types of the decapeptide exerted


Intact (luteal phase)

Intact (luteal phase)

similar effects (Caraty et al. 2007). Smith et al. (2009a,


Intact (anestrous)

2011) reported that the effect of Kp-10 administration


on gonadotropin secretion varied with the stage of the
OVX + E2

Castrated

estrous cycle and the season in ewes with intact


ovaries. The LH response to Kp-10 was greater in the
Intact

Intact

Intact
OVX

late-follicular phase compared with the luteal phase


during the breeding season (Smith et al. 2011), and the
effect of Kp-10 on LH but not FSH secretion was
greater during the anestrous season than in the luteal
Male, female

phase during the breeding season (Smith et al. 2009a).


BW, body weight; E2, estradiol; OVX, ovariectomized; P, progesterone.

In male castrated goats, plasma LH concentrations


Female
Female
Female
Female
Female

Female
Female
Female

were promptly elevated after bolus i.v. administration


Male
Male
Sex

of Kp-10, and the stimulatory effect of the peptide on


LH secretion lasted for several hours (Ohkura et al.
2009b). Similar effects of Kp-10 were observed in pre-
and post-pubertal male goats (Saito et al. 2012), and
Young (pre- and post-pubertal)

cycling female goats during the luteal phase


(Hashizume et al. 2010).
In prepubertal gilts, intracerebroventricular (i.c.v.)
Young (prepubertal)

Young (prepubertal)

administration of as low as 7.8 nmol of Kp-10 signifi-


cantly elevated serum LH and FSH concentrations. On
the other hand, a dose of 780 nmol of Kp-10 was
required to increase serum LH concentrations when it
was administered peripherally, and serum FSH con-
Adult

Adult

Adult

Adult

centrations were not affected even by i.v. injection of


Age

3900 nmol of the peptide (Lents et al. 2008).


In cycling adult mares, bolus i.v. administration of
390 nmol of Kp-10 during the luteal phase signifi-
cantly increased serum LH and FSH concentrations,
Animal
Table 2

Sheep
Cattle

Horse

whereas 780 nmol of Kp-10 administration in the late


Goat

Pig

follicular phase was insufficient to induce ovulation

Animal Science Journal (2013) 84, 369–381 © 2013 Japanese Society of Animal Science
372 H. OKAMURA et al.

(Magee et al. 2009). The effect of Kp-10 on LH secre- LH secretion had no effect on plasma GH, prolactin
tion in pony mares was less potent during the late and cortisol concentrations regardless of the season or
follicular phase than during other follicular stages the stage of the estrous cycle (Smith et al. 2009a;
(Decourt et al. 2010). Whitlock et al. 2010), whereas i.c.v. administration of
Kp-10 significantly increased GH secretion in OVX
Sites of kisspeptin action on ewes (Whitlock et al. 2010). Neither i.v. nor i.c.v. injec-
gonadotropin secretion tion of Kp-10 affected GH secretion in prepubertal gilts
(Lents et al. 2008) or GH or prolactin secretion in
Several lines of evidence indicate that the action of
luteal-phase female goats (Hashizume et al. 2010).
kisspeptin on gonadotropin secretion is mediated cen-
Although it seems that the stimulation of GH secretion
trally by GnRH. First, the majority of GnRH neurons in
is the sole endocrine action of kisspeptin outside of the
mice (Irwig et al. 2004) and sheep (Smith et al. 2009b)
HPG axis observed to date, there is controversy as to
express kisspeptin receptor gene, GPR54. Second,
the conservation of this effect between species or even
peripheral administration of kisspeptin activates GnRH
within the same species, and the biological significance
neurons, as reflected by Fos (a marker of neuronal
of this action of kisspeptin remains far from clear.
activation) expression, in rats (Matsui et al. 2004).
A previous report in mice indicated that kisspeptin/
Third, kisspeptin stimulates GnRH release in vitro from
GPR54 signaling is not required for male or female
hypothalamic tissues from mice (d’Anglemont de
copulatory behavior (Kauffman et al. 2007). As far as
Tassigny et al. 2008), rats (Uenoyama et al. 2011) and
we know, there is no study published to date examin-
sheep (Smith et al. 2011). Fourth, i.v. injection of
ing the effects of kisspeptin on sexual behavior in
kisspeptin increases the concentration of GnRH in the
domestic animals.
portal vessels, with a parallel rise in peripheral LH
concentrations, in sheep (Smith et al. 2011; Li et al.
2012) and goats (Tanaka et al. 2012). Finally, the effect ANATOMY OF KISSPEPTIN NEURONS
of kisspeptin on gonadotropin secretion is blocked by Distribution
pretreatment with a GnRH receptor antagonist in
The distribution of kisspeptin-producing neurons has
rodents (Gottsch et al. 2004; Irwig et al. 2004; Matsui
been examined by in situ hybridization and/or immu-
et al. 2004) and monkeys (Plant et al. 2006) and by
nohistochemistry in sheep (Estrada et al. 2006;
hypothalamo-pituitary disconnection, which prevents
Franceschini et al. 2006; Smith et al. 2007; Merkley
the delivery of GnRH to the pituitary (Clarke et al.
et al. 2012), goats (Takase et al. 2007; Ohkura et al.
1983), in sheep (Smith et al. 2008a).
2009b), pigs (Tomikawa et al. 2010) and horses
The expression of GPR54 in the anterior pituitary in
(Decourt et al. 2008; Magee et al. 2009). Kisspeptin
some animals (Richard et al. 2009), including sheep
neurons are found almost exclusively in two discrete
(Smith et al. 2008a), suggests that kisspeptin may also
areas within the hypothalamus, rostrally in the preop-
act at the pituitary level. Indeed, in vitro studies in
tic area (POA) and caudally in the arcuate nucleus
which Kp-10 was added to pituitary cell cultures
(ARC). Alternatively, the rostral population is found in
derived from sheep (1 nmol/L; Smith et al. 2008a),
the periventricular nucleus rather than the POA in
pigs (10 nmol/L; Suzuki et al. 2008) and cattle
pigs (Tomikawa et al. 2010). This distribution pattern is
(1000 nmol/L; Suzuki et al. 2008; Ezzat et al. 2010)
similar to that in rodents, in which the rostral popu-
showed that Kp-10 increased the concentration of LH
lation of kisspeptin neurons is concentrated in the
in the culture medium. Therefore, it is possible that
anteroventral periventricular nucleus (AVPV) and
kisspeptin acts at dual sites, both the hypothalamus
associated areas (Clarkson & Herbison 2006; Adachi
and the pituitary. However, the biological significance
et al. 2007). Although kisspeptin-immunoreactive
of the latter is completely unclear at present.
neurons were also observed in several other areas of
the equine hypothalamus (Magee et al. 2009), this
Other actions of kisspeptin study may need to be interpreted with caution, as the
Peripheral administration of Kp-10 increased growth antibody used seems to cross-react with other antigens
hormone (GH) secretion in OVX cows treated with in addition to kisspeptin (Pompolo et al. 2006;
estradiol cypionate and/or progesterone (Whitlock Goodman et al. 2007; Smith et al. 2008b).
et al. 2008) but had no effect without the concomitant Although the projections of kisspeptin neurons have
steroid treatment (Whitlock et al. 2008, 2010). not been systematically examined in domestic
Kadokawa et al. (2008) reported that i.v. injection of animals, the initial study in sheep revealed that fibers
Kp-10 elevated plasma GH concentrations in prepu- containing kisspeptin are present throughout the
bertal heifers. However, another study failed to hypothalamus, being most abundant in the POA, ARC
observe any effect of Kp-10 on GH secretion in prepu- and median eminence (ME) (Franceschini et al. 2006).
bertal calves of either sex (Ezzat et al. 2009). In sheep, A more detailed analysis in mice demonstrated that
i.v. injection of doses of Kp-10 that potently stimulated kisspeptin fibers project not only to most of the

© 2013 Japanese Society of Animal Science Animal Science Journal (2013) 84, 369–381
KISSPEPTIN IN DOMESTIC ANIMALS 373

hypothalamic nuclei (except the suprachiasmatic and treated with a low dose of E2 (Smith et al. 2007,
ventromedial nuclei) but also to extra-hypothalamic 2008b, 2009b), indicating the down-regulation of
areas such as the bed nucleus of the stria terminalis, kisspeptin expression by E2 as in other species. On the
medial amygdala, periaqueductal gray matter, and other hand, the number of kisspeptin-expressing
locus coeruleus (Clarkson & Herbison 2009), implicat- neurons in the middle and/or caudal portions of the
ing kisspeptin in a variety of physiological functions ARC is higher during the late-follicular phase than
beyond reproduction. during the luteal phase in cycling ewes (Estrada et al.
2006; Smith et al. 2009b). Interestingly, the middle
Sensitivity of kisspeptin neurons to and caudal portions of the ovine ARC contain greater
gonadal steroids numbers of kisspeptin neurons in mid-luteal-phase
Although ERa plays pivotal roles in both the negative ewes than in intact rams (Cheng et al. 2010). There-
(Dorling et al. 2003) and positive (Wintermantel et al. fore, the effect of gonadal steroids on kisspeptin
2006) feedback effects of E2 on GnRH secretion, GnRH expression in the ARC seems to vary among species,
neurons themselves do not express ERa (Lehman et al. sexes and the portions of the nucleus, and must there-
1993). Therefore, some other population of neurons fore be mediated by a complex mechanism, especially
must mediate the effect of E2 action on GnRH in sheep.
neurons. Among several subsets of neurons, kisspeptin
neurons are conspicuous in that virtually all of them Interaction of kisspeptin neurons with
express ERa in the ARC and approximately 50% of GnRH neurons
them do so in the POA in ewes (Franceschini et al. Approximately 40% to 50% of the GnRH neurons in
2006). Moreover, approximately 90% of kisspeptin the POA and medial basal hypothalamus (MBH)
neurons also contain progesterone receptor in the receive direct apposition of kisspeptin fibers on their
ovine ARC (Smith et al. 2007). cell bodies in ewes, and during the breeding season this
In rodents, E2 is well established as up-regulating percentage increases to virtually 100% in the MBH,
the expression of kisspeptin in the AVPV and down- although not in the POA (Smith et al. 2008b). Such
regulating its expression in the ARC (Smith et al. 2005; apposition occurs only in a small percentage of GnRH
Adachi et al. 2007). In domestic animals, there is cur- neurons in the POA in male goats (Matsuyama et al.
rently only fragmentary evidence for the regulation of 2011), male mice (Clarkson & Herbison 2006) and
kisspeptin expression by steroid hormones. castrated male monkeys (Ramaswamy et al. 2008). On
The rostral population of kisspeptin neurons express the other hand, kisspeptin fibers associate extensively
Kiss1 messenger RNA (mRNA) at higher levels during with GnRH axons in the ME in both females (ewes:
the late-follicular phase than during the luteal phase Smith et al. 2011; mares: Decourt et al. 2008; rats:
in ewes (Smith et al. 2009b) and up-regulate Kiss1 Uenoyama et al. 2011) and males (goats: Ohkura et al.
expression in response to high levels of E2 in pigs 2009b; Matsuyama et al. 2011; monkeys: Ramaswamy
(Tomikawa et al. 2010). In goats, a subset of neurons in et al. 2008) as shown (Fig. 2B). Electron microscopy has
the POA of mature males expresses Kiss1 and kisspep- revealed that the axon terminals of kisspeptin neurons
tin peptide (Matsuyama et al. 2011), but this expres- do in fact closely contact GnRH axon terminals in goats
sion is completely eliminated by castration (Ohkura (Matsuyama et al. 2011) and rats (Uenoyama et al.
et al. 2009b). These observations are similar to the 2011). Therefore, although kisspeptin neurons may
findings in rodents. On the other hand, the action of interact with either cell bodies or the axon terminals of
E2 in ewes remains debatable, because a similar E2 GnRH neurons, the former is unlikely to be the major
treatment increased the expression of Kiss1 in the site of kisspeptin action in males. These differences in
ovine POA in one report (Smith et al. 2008b) but not in the anatomy of the kisspeptin-GnRH interaction may
another (Smith et al. 2007). reflect different effects of kisspeptin action on GnRH
In gonadectomized goats, a large number of kisspep- secretion: the control of the pulsatile and surge modes
tin neurons can be observed in the ARC, being much of GnRH secretion.
abundant at its caudal portion in both females
(Fig. 2A) and males (Ohkura et al. 2009b), whereas
the distribution of kisspeptin neurons is extremely
CONTROL OF GnRH SECRETION BY
sparse in intact animals (Matsuyama et al. 2011), sug- KISSPEPTIN NEURONS
gesting a strong inhibition of kisspeptin expression by Central infusion of a GPR54 antagonist (Roseweir et al.
androgen throughout the ARC. In OVX pigs, the 2009) blocked or attenuated both pulsatile LH secre-
expression of Kiss1 in the ARC is decreased by E2, but tion (Roseweir et al. 2009; Smith et al. 2011; Goodman
only in the most caudal portion of the nucleus et al. 2012) and the E2-induced LH surge (Smith et al.
(Tomikawa et al. 2010). In OVX ewes, the number of 2011) in ewes. Functional inactivation of GPR54 by
kisspeptin-expressing neurons in the ARC is signifi- chronic administration of potent GPR54 agonists
cantly higher in vehicle-control animals than in those (Matsui et al. 2012) completely eliminated LH pulses in

Animal Science Journal (2013) 84, 369–381 © 2013 Japanese Society of Animal Science
374 H. OKAMURA et al.

(Dyn) in both sheep (Goodman et al. 2007; Cheng et al.


2010) and goats (Wakabayashi et al. 2010; Matsuyama
et al. 2011), and these neurons have therefore
been termed KNDy (kisspeptin/NKB/Dyn) neurons
(Lehman et al. 2010). KNDy neurons are surrounded
by their own dense network of varicose fibers in sheep
(Foradori et al. 2002, 2006) and goats (Wakabayashi
et al. 2010) and are bilaterally interconnected by NKB
axons in rats (Krajewski et al. 2010) and goats
(Wakabayashi et al. 2013). Therefore, KNDy neurons
Figure 2 The distribution of kisspeptin neurons in the goat likely form a neuronal network interconnected by
medial basal hypothalamus (MBH). (A) Immunofluorescence axon collaterals and/or dendrites. This hypothesis is
staining for kisspeptin (green) in the arcuate nucleus (ARC) supported by the physiological finding that the activity
of an ovariectomized goat. Kisspeptin neurons are densely of the GnRH pulse generator is synchronous between
packed in the caudal portion of the ARC. (B) Dual the ARCs on both sides of the brain (Wakabayashi et al.
immunofluorescence for kisspeptin (red) and
2013). Such a neuronal network would serve as a
gonadotropin-releasing hormone (GnRH; green) at the
lateral aspect of the median eminence in a castrated male
framework for synchronizing the bursting activities of
goat. Kisspeptin fibers make intimate associations with the KNDy neurons and thereby generate discrete
GnRH fibers at the external layer of the median eminence neural signals to induce pulsatile GnRH secretion.
(ME) at the level of the tuberoinfundibular sulcus. ARC, Like kisspeptin, NKB has also recently attracted con-
arcuate nucleus; MEe, external layer of the ME; MEi, siderable attention. First, Topaloglu et al. (2009) discov-
internal layer of the ME; pt, pars tuberalis; 3V, third ered that inactivating mutations of Tac3 and Tacr3,
ventricle. Scale bars, 100 mm.
which encode NKB and its receptor (NK3R), respec-
tively, produce gonadotropin deficiency and pubertal
male (Ohkura et al. 2011) and female (Wakabayashi failure in humans, suggesting the involvement of NKB/
et al. 2011) goats. Furthermore, immunoneutraliza- NK3R signaling in the control of pulsatile GnRH secre-
tion of endogenous kisspeptin activity disrupted the tion. This suggestion is supported by experimental
estrous cycle in rats (Kinoshita et al. 2005). Together observations in goats that bolus i.c.v. administration of
with earlier human genetic findings (de Roux et al. NKB immediately activates the GnRH pulse generator
2003; Seminara et al. 2003) and subsequent studies in (Wakabayashi et al. 2010) and further, that continuous
transgenic mice (Funes et al. 2003; Seminara et al. i.v. infusion of senktide, a selective NK3R agonist, pro-
2003; Dungan et al. 2007), these results clearly show duces intermittent bursts of the GnRH pulse generator
that kisspeptin plays pivotal roles in the control of both with corresponding LH pulses (Wakabayashi et al.
the pulsatile and surge modes of GnRH secretion. 2012). One possible site of this action of NKB might be
a subset of KNDy neurons, as KNDy neurons express
Control of the pulsatile GnRH secretion NK3R (Amstalden et al. 2010; Wakabayashi et al. 2012)
by kisspeptin neurons and i.c.v. administration of senktide-activated KNDy
The pulsatile discharge of GnRH into the portal vessels neurons in rats (Navarro et al. 2011a) and sheep
is governed by neural substrates, the so-called GnRH (Sakamoto et al. 2012). Moreover, NKB has been
pulse generator, that periodically fire a volley of action shown to elicit trains of action potentials in mouse brain
potentials (Knobil 1981; Kawakami et al. 1982; Mori slices containing KNDy neurons (Navarro et al. 2011b).
et al. 1991; Nishihara et al. 1994). Recent electrophysi- Collectively, it is plausible that NKB is involved in the
ological studies in goats have demonstrated that activ- generation of the intermittent bursts of the GnRH pulse
ity of the GnRH pulse generator is monitored at close generator via an auto-feedback loop of the KNDy neu-
vicinity of the ARC kisspeptin neurons in both males ronal network and that kisspeptin transmits this activ-
(Ohkura et al. 2009b) and females (Wakabayashi et al. ity to the GnRH neurons (Fig. 3) (Maeda et al. 2010;
2010). Moreover, kisspeptin has been shown in Wakabayashi et al. 2010). Although Dyn has been sug-
monkeys to be secreted episodically into the ME, and gested to terminate the burst activity (Wakabayashi
these kisspeptin pulses are temporally associated with et al. 2010), the precise role and action sites of Dyn in
pulsatile GnRH secretion (Keen et al. 2008). These the KNDy network remain unclear.
results, together with other findings, have led to the
compelling hypothesis that the population of kisspep-
tin neurons in the ARC is the intrinsic source of the
Control of the GnRH surge by
GnRH pulse generator (Navarro et al. 2009; Lehman kisspeptin neurons
et al. 2010; Maeda et al. 2010; Rance et al. 2010). The rostral population of kisspeptin neurons (in the
Virtually all kisspeptin neurons in the ARC AVPV) is well documented as playing a critical role in
co-express neurokinin B (NKB) and dynorphin A triggering the preovulatory GnRH/LH surge in rodents

© 2013 Japanese Society of Animal Science Animal Science Journal (2013) 84, 369–381
KISSPEPTIN IN DOMESTIC ANIMALS 375

Figure 3 A schematic illustration of the novel hypothalamic-pituitary-gonadal (HPG) axis. The two populations of kisspeptin
neurons in the preoptic area (POA) and arcuate nucleus (ARC) are suggested to be located upstream of the
gonadotropin-releasing hormone (GnRH) neurons in the HPG axis and to control the surge and pulse modes of
GnRH/luteinizing hormone (LH) secretion, respectively. Neurokinin B (NKB) likely plays a role in the pulse-generating
mechanism of the ARC kisspeptin neurons. ME, median eminence; och, optic chiasm; Pit, pituitary; pt, pars tuberalis.

(Oakley et al. 2009; Tsukamura et al. 2010; Khan & neurons in the POA are involved in the LH
Kauffman 2012). First, E2 treatment dramatically surge. OVX sows subjected to the E2 treatment suf-
enhances the expression of kisspeptin in the AVPV ficient to induce an LH surge and estrous behavior
(Smith et al. 2005; Adachi et al. 2007; Tomikawa et al. comparable to those observed in intact animals,
2012). Second, the expression of kisspeptin in the exhibited significantly increased expression of
AVPV is highest at the time of the LH surge (Smith kisspeptin in the rostral (periventricular nucleus) but
et al. 2006; Adachi et al. 2007). Third, the E2 treatment not the caudal (ARC) population of kisspeptin
that induces the LH surge in OVX animals also acti- neurons (Tomikawa et al. 2010). In OVX ewes, the
vates the majority of the kisspeptin neurons in the expression of kisspeptin in the POA was markedly
AVPV (Smith et al. 2006; Adachi et al. 2007). Mating enhanced by even a low dose of E2, which produces
stimulus has been shown to activate POA kisspeptin circulating E2 levels during the luteal phase (Smith
neurons to induce ovulation in musk shrews, which et al. 2008b). Hoffman et al. (2011) demonstrated
are reflex ovulators (Inoue et al. 2011). Fourth, a brief using an excellent synchronized follicular phase
application of Kp-10 induces prolonged activation of model in cycling ewes that Fos was induced con-
GnRH neurons in mouse brain slices (Han et al. 2005; comitantly in kisspeptin neurons and in GnRH
Pielecka-Fortuna et al. 2008). neurons in the POA, but not in kisspeptin neurons in
In domestic animals, on the other hand, the role of the ARC, at the time of the GnRH/LH surge. These
kisspeptin in evoking the LH surge has not been fully results suggest that the POA kisspeptin-GnRH system
evaluated. A few reports in several animal models is involved in the mechanism of GnRH surge genera-
have suggested that, as in rodents, the kisspeptin tion in ewes (Fig. 3).

Animal Science Journal (2013) 84, 369–381 © 2013 Japanese Society of Animal Science
376 H. OKAMURA et al.

Interestingly, another group showed that Fos was injections of Kp-10 (15.6 nmol) for 24 h increased LH
induced in kisspeptin neurons not only in the POA but secretion to the levels comparable to those of the surge
also in the ARC during the LH surge in cycling ewes in cycling adult ewes; this was followed by a rise in
(Merkley et al. 2012). Moreover, the number of progesterone secretion within 5 days after the treat-
kisspeptin-expressing neurons in the ARC increases ment. These results indicate that intermittent Kp-10
during the late-follicular phase in this species (Estrada administration, which might mimic increasing activity
et al. 2006; Smith et al. 2009b). Therefore, a subset of of the kisspeptin neuronal system during the prepu-
kisspeptin neurons in the ARC may also participate in bertal period, induces ovulation and formation of the
the induction of the GnRH/LH surge in ewes (Caraty corpus luteum, suggesting that kisspeptin has potential
et al. 1998; Merkley et al. 2012). However, ARC for advancing puberty in domestic animals. Because
kisspeptin neurons seem to be homogeneous in terms the duration of the induced luteal activity was shorter
of molecular identity: virtually all of them express than that of the normal luteal phase, the method
kisspeptin, NKB, Dyn, NK3R, ERa and progesterone employed in this study might require further modifi-
receptor (Oakley et al. 2009; Lehman et al. 2010; cation before being applied in the field.
Goodman & Lehman 2012). Further studies are
required to clarify whether there is a specific subpopu- Development of effective
lation of kisspeptin neurons in the ovine ARC and kisspeptin analogues
whether the mechanism by which kisspeptin controls
Kp-10 is clearly a potent GnRH secretagogue. Never-
the GnRH surge differs between rodents and sheep.
theless, relatively large amounts of kisspeptin, com-
pared with those of GnRH analogues, were necessary
APPLICATIONS OF KISSPEPTIN AND to obtain the expected outcomes in the aforemen-
RELATED SUBSTANCES: PROSPECTIVE tioned studies in sheep (Caraty et al. 2007), pigs (Lents
Studies investigating potential applications of kisspep- et al. 2008) and horses (Magee et al. 2009; Caraty et al.
tin to the management of fertility in domestic animals 2012a). Therefore kisspeptin analogues with more
have just begun. Although any conclusions would be powerful GnRH/LH secretion-inducing activity would
premature at this point, a few promising results have be highly desirable for practical applications in domes-
nevertheless been demonstrated (Caraty et al. 2010, tic animals. Several groups have recently reported the
2012a). development of novel kisspeptin analogues (Tomita
et al. 2008; Curtis et al. 2010; Matsui et al. 2010, 2012).
Synchronization of ovulation Future studies of the effects of these analogues on LH
by kisspeptin secretion in domestic animals are highly anticipated.
To investigate the effect of Kp-10 on the induction of
the LH surge, Kp-10 was infused intravenously at a Mode of kisspeptin administration
rate of 480 nmol/h for 8 h in progesterone-primed The action of kisspeptin on gonadotropin secretion
cyclic ewes (Caraty et al. 2007). The LH surge occurred seems to largely depend on the mode of its adminis-
within 2 h after the start of the infusion in the Kp-10- tration. Continuous i.v. infusion of Kp-10 increased E2
treated group but later and with variable timing in secretion and induced the LH surge in seasonally
vehicle control-treated animals. Subsequent examina- anestrous ewes (Caraty et al. 2007; Sébert et al. 2010),
tion showed no difference in the number of corpora suggesting that kisspeptin can be utilized to stimulate
lutea between the Kp-10 and vehicle-treated groups. the gonadal activity. However, it should be noted that
These results strongly suggest that kisspeptin could be continuous exposure of the gonadotrophs to GnRH
used as a new tool for synchronization of ovulation causes desensitization of the GnRH receptors, which
and thereby improve the efficiency of artificial insemi- results in the abrogation of gonadotropin secretion
nation and in vitro fertilization programs (Caraty et al. (Knobil 1980). Likewise, continuous administration of
2010, 2012a). kisspeptin or its analogues has been shown to suppress
However, in pony mares a similar treatment with the secretion of gonadotropins and/or gonadal steroid
Kp-10 (infusion for 3 days at a rate of 2370 nmol/h) in hormones in monkeys (Seminara et al. 2006), rats
the late follicular phase failed to advance or synchro- (Matsui et al. 2012), sheep (Messager et al. 2005) and
nize the LH surge (Caraty et al. 2012a) despite the fact goats (Ohkura et al. 2011). Therefore, the optimal dose
that Kp-10 can stimulate gonadotropin secretion in of kisspeptin and duration of the treatment might be
mares (Magee et al. 2009; Decourt et al. 2010). carefully determined for a given species, when con-
tinuous administration of kisspeptin is applied to
Acceleration of puberty by kisspeptin improve reproductive efficacy in domestic animals.
Redmond et al. (2011) investigated whether kisspeptin Caraty et al. (2012b) have demonstrated in ewes
activates the HPG axis in prepubertal ewe lambs. that intermittent Kp-10 administration leads to inter-
Treatment of lambs (28 weeks of age) with hourly i.v. mittent rises in GnRH and LH secretion (Caraty et al.

© 2013 Japanese Society of Animal Science Animal Science Journal (2013) 84, 369–381
KISSPEPTIN IN DOMESTIC ANIMALS 377

2012b) mimicking the pulsatile nature of physiological ACKNOWLEDGMENTS


secretion of GnRH/LH in vivo (Karsch 1984). The result This work was supported in part by a grant from the
clearly indicate that intermittent repeated administra- Ministry of Agriculture, Forestry and Fisheries of Japan
tion of Kp-10 avoids desensitization of kisspeptin (Research Program on Innovative Technologies for
receptors as well as GnRH receptors, and suggests that Animal Breeding, Reproduction and Vaccine Develop-
this mode of administration would be an alternative ment, REP-2001), and by a Grant-in Aid for Young
protocol for the kisspeptin application. Indeed, it has Scientists (B), 24780275 (TY) and 23780285 (YW) from
been reported that hourly i.v. injection of Kp-10 main- the Japan Society for the Promotion of Science.
tained a train of LH discharges for 48 h in juvenile
monkeys (Plant et al. 2006) and induced ovulation in
REFERENCES
prepubertal lambs (Redmond et al. 2011).
Adachi S, Yamada S, Takatsu Y, Matsui H, Kinoshita M,
Takase K, Sugiura H, Ohtaki T, Matsumoto H, Uenoyama
Application of NK3R agonists to Y, Tsukamura H, Inoue K, Maeda K-I. 2007. Involvement
facilitate pulsatile GnRH/LH secretion of anteroventral periventricular metastin/kisspeptin
neurons in estrogen positive feedback action on luteiniz-
As shown in Figure 3, the studies on the neural ing hormone release in female rats. The Journal of Repro-
mechanism of the generation of the GnRH pulse duction and Development 53, 367–378.
have proposed the hypothesis that NKB plays a Allouche J, Bennet A, Barbe P, Plantavid M, Caron P, Louvet
pivotal role in the GnRH pulse generation cascade JP. 1991. LH pulsatility and in vitro bioactivity in women
with anorexia nervosa-related hypothalamic amenor-
(Maeda et al. 2010; Rance et al. 2010; Wakabayashi
rhea. Acta Endocrinologica 125, 614–620.
et al. 2010). This hypothesis has been experimentally Amstalden M, Coolen LM, Hemmerle AM, Billings HJ,
addressed in a preliminary study using OVX+E2 Connors JM, Goodman RL, Lehman MN. 2010. Neuroki-
goats (Yamamura et al. 2012). Continuous subcutane- nin 3 receptor immunoreactivity in the septal region,
ous infusion of senktide (1440 nmol/h) for 24 h preoptic area and hypothalamus of the female sheep:
increased the frequency of the GnRH pulse generator colocalisation in neurokinin B cells of the arcuate nucleus
activity by approximately 1.5-fold over the pre- but not in gonadotrophin-releasing hormone neurones.
Journal of Neuroendocrinology 22, 1–12.
infusion value, and the effect of senktide was main- Caraty A, Decourt C, Briant C, Beltramo M. 2012a.
tained throughout the infusion period. The results Kisspeptins and the reproductive axis: potential applica-
suggest, although the dose of senktide used in that tions to manage reproduction in farm animals. Domestic
study was still very high, that high-frequency pulsa- Animal Endocrinology 43, 95–102.
tile LH secretion can be produced by continuous Caraty A, Fabre-Nys C, Delaleu B, Locatelli A, Bruneau G,
administration of senktide, a method more preferable Karsch FJ, Herbison A. 1998. Evidence that the medioba-
sal hypothalamus is the primary site of action of estradiol
over intermittent repeated administration in the field
in inducing the preovulatory gonadotropin releasing
of animal management. hormone surge in the ewe. Endocrinology 139, 1752–1760.
There are several situations in which an insufficient Caraty A, Franceschini I, Hoffman GE. 2010. Kisspeptin and
LH pulse frequency causes reproductive disorders, the preovulatory gonadotrophin-releasing hormone/
such as domestic animals with undernutrition or a luteinising hormone surge in the ewe: basic aspects and
negative energy balance (Schillo 1992) and women potential applications in the control of ovulation. Journal
with anorexia nervosa (Allouche et al. 1991) or exer- of Neuroendocrinology 22, 710–715.
Caraty A, Lomet D, Sébert ME, Guillaume D, Beltramo M,
cise amenorrhea (Veldhuis et al. 1985). The LH pulse-
Evans NP. 2012b. GnRH release into the hypophyseal
stimulating action of senktide implies that NK3R portal blood of the ewe mirrors both pulsatile and con-
agonists may hold promise as novel therapeutic drugs tinuous intravenous infusion of kisspeptin: insights into
to accelerate or improve gonadal activity in humans as kisspeptin mechanism of action. 2nd world conference of
well as domestic animals. kisspeptin signaling in the brain, Tokyo, Japan, P-40.
Caraty A, Smith JT, Lomet D, Ben Saïd S, Morrissey A,
Cognie J, Doughton B, Baril G, Briant C, Clarke IJ. 2007.
Conclusion Kisspeptin synchronizes preovulatory surges in cyclical
ewes and causes ovulation in seasonally acyclic ewes.
Emerging studies over the last decade revealed that the Endocrinology 148, 5258–5267.
kisspeptin neuronal system plays an essential role as Cheng G, Coolen LM, Padmanabhan V, Goodman RL,
the gatekeeper of reproduction and thereby governs Lehman MN. 2010. The kisspeptin/neurokinin
both the pulsatile and surge modes of GnRH secretion B/dynorphin (KNDy) cell population of the arcuate
(Fig. 3). Although translational and field studies by a nucleus: sex differences and effects of prenatal testoster-
few groups have just begun, the reported biological one in sheep. Endocrinology 151, 301–311.
Clarke IJ. 2011. Control of GnRH secretion: one step back.
actions of kisspeptin and NKB strongly suggest that Frontiers in Neuroendocrinology 32, 367–375.
analogues of kisspeptin and NKB would be useful in Clarke IJ, Cummins JT, de Kretser DM. 1983. Pituitary gland
the development of novel strategies for the manage- function after disconnection from direct hypothalamic
ment of fertility in domestic animals. influences in the sheep. Neuroendocrinology 36, 376–384.

Animal Science Journal (2013) 84, 369–381 © 2013 Japanese Society of Animal Science
378 H. OKAMURA et al.

Clarkson J, Herbison AE. 2006. Postnatal development of Foradori CD, Coolen LM, Fitzgerald ME, Skinner DC,
kisspeptin neurons in mouse hypothalamus; sexual Goodman RL, Lehman MN. 2002. Colocalization of pro-
dimorphism and projections to gonadotropin-releasing gesterone receptors in parvicellular dynorphin neurons of
hormone neurons. Endocrinology 147, 5817–5825. the ovine preoptic area and hypothalamus. Endocrinology
Clarkson J, Herbison AE. 2009. Oestrogen, kisspeptin, 143, 4366–4374.
GPR54 and the pre-ovulatory luteinising hormone surge. Franceschini I, Lomet D, Cateau M, Delsol G, Tillet Y, Caraty
Journal of Neuroendocrinology 21, 305–311. A. 2006. Kisspeptin immunoreactive cells of the ovine
Curtis AE, Cooke JH, Baxter JE, Parkinson JRC, Bataveljic A, preoptic area and arcuate nucleus co-express estrogen
Ghatei MA, Bloom SR, Murphy KG. 2010. A receptor alpha. Neuroscience Letters 401, 225–230.
kisspeptin-10 analog with greater in vivo bioactivity than Funes S, Hedrick JA, Vassileva G, Markowitz L, Abbondanzo
kisspeptin-10. American Journal of Physiology Endocrinology S, Golovko A, Yang S, Monsma FJ, Gustafson EL. 2003.
and Metabolism 298, E296–E303. The KiSS-1 receptor GPR54 is essential for the develop-
d’Anglemont de Tassigny X, Fagg LA, Carlton MBL, Colledge ment of the murine reproductive system. Biochemical and
WH. 2008. Kisspeptin can stimulate gonadotropin- Biophysical Research Communications 312, 1357–1363.
releasing hormone (GnRH) release by a direct action at Goodman RL, Lehman MN. 2012. Kisspeptin neurons from
GnRH nerve terminals. Endocrinology 149, 3926–3932. mice to men: similarities and differences. Endocrinology
de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, 153, 5105–5118.
Milgrom E. 2003. Hypogonadotropic hypogonadism due Goodman RL, Lehman MN, Smith JT, Coolen LM, de
to loss of function of the KiSS1-derived peptide receptor Oliveira CVR, Jafarzadehshirazi MR, Pereira A, Iqbal J,
GPR54. Proceedings of the National Academy of Sciences of the Caraty A, Ciofi P, Clarke IJ. 2007. Kisspeptin neurons in
United States of America 100, 10972–10976. the arcuate nucleus of the ewe express both dynorphin A
Decourt C, Tillet Y, Caraty A, Franceschini I, Briant C. 2008. and neurokinin B. Endocrinology 148, 5752–5760.
Kisspeptin immunoreactive neurons in the equine Goodman RL, Maltby MJ, Millar RP, Hileman SM, Nestor CC,
hypothalamus Interactions with GnRH neuronal system. Whited B, Tseng AS, Coolen LM, Lehman MN. 2012.
Journal of Chemical Neuroanatomy 36, 131–137. Evidence that dopamine acts via kisspeptin to hold GnRH
Decourt CV, Merzouki Y, Duchamp G, Bruneau B, pulse frequency in check in anestrous ewes. Endocrinology
Caraty A, Briant C. 2010. Effects of constant intravenous 153, 5918–5927.
kisspeptin administration on luteinizing hormone release Gottsch ML, Cunningham MJ, Smith JT, Popa SM, Acohido
in cyclic mares. Animal Reproduction Science 121, S65– BV, Crowley WF, Seminara S, Clifton DK, Steiner RA.
S67. 2004. A role for kisspeptins in the regulation of gonado-
Dorling AA, Todman MG, Korach KS, Herbison AE. 2003. tropin secretion in the mouse. Endocrinology 145, 4073–
Critical role for estrogen receptor alpha in negative feed- 4077.
back regulation of gonadotropin-releasing hormone Han SK, Gottsch ML, Lee KJ, Popa SM, Smith JT, Jakawich
mRNA expression in the female mouse. Neuroendocrinol- SK, Clifton DK, Steiner RA, Herbison AE. 2005. Activa-
ogy 78, 204–209. tion of gonadotropin-releasing hormone neurons by
Dungan HM, Gottsch ML, Zeng H, Gragerov A, Bergmann JE, kisspeptin as a neuroendocrine switch for the onset of
Vassilatis DK, Clifton DK, Steiner RA. 2007. The role puberty. The Journal of Neuroscience 25, 11349–11356.
of kisspeptin-GPR54 signaling in the tonic regulation Hashizume T, Saito H, Sawada T, Yaegashi T, Ezzat AA, Sawai
and surge release of gonadotropin-releasing hormone/ K, Yamashita T. 2010. Characteristics of stimulation of
luteinizing hormone. The Journal of Neuroscience 27, 12088– gonadotropin secretion by kisspeptin-10 in female goats.
12095. Animal Reproduction Science 118, 37–41.
Estrada KM, Clay CM, Pompolo S, Smith JT, Clarke IJ. Hoffman GE, Le WW, Franceschini I, Caraty A, Advis JP.
2006. Elevated KiSS-1 expression in the arcuate nucleus 2011. Expression of fos and in vivo median eminence
prior to the cyclic preovulatory gonadotrophin-releasing release of LHRH identifies an active role for preoptic area
hormone/lutenising hormone surge in the ewe suggests a kisspeptin neurons in synchronized surges of LH and
stimulatory role for kisspeptin in oestrogen-positive feed- LHRH in the ewe. Endocrinology 152, 214–222.
back. Journal of Neuroendocrinology 18, 806–809. Inoue N, Sasagawa K, Ikai K, Sasaki Y, Tomikawa J, Oishi S,
Ezzat AA, Saito H, Sawada T, Yaegashi T, Goto Y, Nakajima Y, Fujii N, Uenoyama Y, Ohmori Y, Yamamoto N, Hondo E,
Jin J, Yamashita T, Sawai K, Hashizume T. 2010. The role Maeda K-I TH. 2011. Kisspeptin neurons mediate reflex
of sexual steroid hormones in the direct stimulation by ovulation in the musk shrew (Suncus murinus). Proceedings
kisspeptin-10 of the secretion of luteinizing hormone, of the National Academy of Sciences of the United States of
follicle-stimulating hormone and prolactin from bovine America 108, 17527–17532.
anterior pituitary cells. Animal Reproduction Science 121, Irwig MS, Fraley GS, Smith JT, Acohido BV, Popa SM,
267–272. Cunningham MJ, Gottsch ML, Clifton DK, Steiner RA.
Ezzat AA, Saito H, Sawada T, Yaegashi T, Yamashita T, Hirata 2004. Kisspeptin activation of gonadotropin releasing
TI, Sawai K, Hashizume T. 2009. Characteristics of the hormone neurons and regulation of KiSS-1 mRNA in the
stimulatory effect of kisspeptin-10 on the secretion of male rat. Neuroendocrinology 80, 264–272.
luteinizing hormone, follicle-stimulating hormone and Kadokawa H, Matsui M, Hayashi K, Matsunaga N,
growth hormone in prepubertal male and female cattle. Kawashima C, Shimizu T, Kida K, Miyamoto A. 2008.
The Journal of Reproduction and Development 55, 650–654. Peripheral administration of kisspeptin-10 increases
Foradori CD, Amstalden M, Goodman RL, Lehman MN. plasma concentrations of GH as well as LH in prepubertal
2006. Colocalisation of dynorphin A and neurokinin B Holstein heifers. The Journal of Endocrinology 196, 331–334.
immunoreactivity in the arcuate nucleus and median Kanda S, Akazome Y, Matsunaga T, Yamamoto N, Yamada S,
eminence of the sheep. Journal of Neuroendocrinology 18, Tsukamura H, Maeda K-I, Oka Y. 2008. Identification of
534–541. KiSS-1 product kisspeptin and steroid-sensitive sexually

© 2013 Japanese Society of Animal Science Animal Science Journal (2013) 84, 369–381
KISSPEPTIN IN DOMESTIC ANIMALS 379

dimorphic kisspeptin neurons in medaka (Oryzias latipes). Li Q, Roa A, Clarke IJ, Smith JT. 2012. Seasonal variation in
Endocrinology 149, 2467–2476. the gonadotropin-releasing hormone response to kisspep-
Karsch FJ. 1984. The hypothalamus and anterior pituitary tin in sheep: possible kisspeptin regulation of the kisspep-
gland. In: Austin CR, Short RV (eds), Reproduction in tin receptor. Neuroendocrinology 96, 212–221.
Mammals, Vol. 3, Hormonal Control of Reproduction, 2nd edn, Maeda K-I, Adachi S, Inoue K, Ohkura S, Tsukamura H.
pp. 1–20. Cambridge University Press, Cambridge. 2007. Metastin/kisspeptin and control of estrous cycle in
Kauffman AS, Park JH, McPhie-Lalmansingh AA, Gottsch rats. Reviews in Endocrine and Metabolic Disorders 8, 21–
ML, Bodo C, Hohmann JG, Pavlova MN, Rohde AD, 29.
Clifton DK, Steiner RA, Rissman EF. 2007. The kisspeptin Maeda K-I, Ohkura S, Uenoyama Y, Wakabayashi Y, Oka Y,
receptor GPR54 is required for sexual differentiation of the Tsukamura H, Okamura H. 2010. Neurobiological mecha-
brain and behavior. The Journal of Neuroscience 27, 8826– nisms underlying GnRH pulse generation by the hypotha-
8835. lamus. Brain Research 1364, 103–115.
Kawakami M, Uemura T, Hayashi R. 1982. Electrophysi- Magee C, Foradori CD, Bruemmer JE, Arreguin-Arevalo JA,
ological correlates of pulsatile gonadotropin release in McCue PM, Handa RJ, Squires EL, Clay CM. 2009. Bio-
rats. Neuroendocrinology 35, 63–67. logical and anatomical evidence for kisspeptin regulation
Keen KL, Wegner FH, Bloom SR, Ghatei MA, Terasawa E. of the hypothalamic-pituitary-gonadal axis of estrous
2008. An increase in kisspeptin-54 release occurs with horse mares. Endocrinology 150, 2813–2821.
the pubertal increase in luteinizing hormone-releasing Matsui H, Takatsu Y, Kumano S, Matsumoto H, Ohtaki T.
hormone-1 release in the stalk-median eminence of 2004. Peripheral administration of metastin induces
female rhesus monkeys in vivo. Endocrinology 149, 4151– marked gonadotropin release and ovulation in the rat.
4157. Biochemical and Biophysical Research Communications 320,
Khan AR, Kauffman AS. 2012. The role of kisspeptin and 383–388.
RFamide-related peptide-3 neurones in the circadian- Matsui H, Takatsu Y, Tanaka A, Asami T, Nishizawa N, Kiba A,
timed preovulatory luteinising hormone surge. Journal of Kumano S, Suzuki A, Kusaka M, Ohtaki T. 2010. Potent
Neuroendocrinology 24, 131–143. and efficient testosterone suppression by chronic adminis-
Kinoshita M, Tsukamura H, Adachi S, Matsui H, Uenoyama tration of novel metastin analogues, TAK-448 and TAK-
Y, Iwata K, Yamada S, Inoue K, Ohtaki T, Matsumoto H, 683, in male rats. 21st Meeting of the European Association for
Maeda K-I. 2005. Involvement of central metastin in the Cancer Research; 26–29 June 2010, Oslo, Norway. p. 66.
regulation of preovulatory luteinizing hormone surge and Matsui H, Tanaka A, Yokoyama K, Takatsu Y, Ishikawa K,
estrous cyclicity in female rats. Endocrinology 146, 4431– Asami T, Nishizawa N, Suzuki A, Kumano S, Terada M,
4436. Kusaka M, Kitada C, Ohtaki T. 2012. Chronic administra-
Knobil E. 1980. The neuroendocrine control of the men- tion of the metastin/kisspeptin analog KISS1-305 or the
strual cycle. Recent Progress in Hormone Research 36, 53– investigational agent TAK-448 suppresses hypothalamic
88. pituitary gonadal function and depletes plasma testoster-
Knobil E. 1981. Patterns of hypophysiotropic signals and one in adult male rats. Endocrinology 153, 5297–5308.
gonadotropin secretion in the rhesus monkey. Biology of Matsuyama S, Ohkura S, Mogi K, Wakabayashi Y, Mori Y,
Reproduction 24, 44–49. Tsukamura H, Maeda K-I, Ichikawa M, Okamura H. 2011.
Kotani M, Detheux M, Vandenbogaerde A, Communi D, Morphological evidence for direct interaction between
Vanderwinden JM, Le Poul E, Brézillon S, Tyldesley R, kisspeptin and gonadotropin-releasing hormone neurons
Suarez-Huerta N, Vandeput F, Blanpain C, Schiffmann at the median eminence of the male goat: an immuno-
SN, Vassart G, Parmentier M. 2001. The metastasis sup- electron microscopic study. Neuroendocrinology 94, 323–
pressor gene KiSS-1 encodes kisspeptins, the natural 332.
ligands of the orphan G protein-coupled receptor GPR54. Merkley CM, Porter KL, Coolen LM, Hileman SM, Billings
The Journal of Biological Chemistry 276, 34631–34636. HJ, Drews S, Goodman RL, Lehman MN. 2012. KNDy
Krajewski SJ, Burke MC, Anderson MJ, McMullen NT, (kisspeptin/neurokinin B/dynorphin) neurons are acti-
Rance NE. 2010. Forebrain projections of arcuate neuro- vated during both pulsatile and surge secretion of LH in
kinin B neurons demonstrated by anterograde tract- the ewe. Endocrinology 153, 5406–5414.
tracing and monosodium glutamate lesions in the rat. Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D,
Neuroscience 166, 680–697. Dixon J, Thresher RR, Malinge I, Lomet D, Carlton MBL,
Lee JH, Miele ME, Hicks DJ, Phillips KK, Trent JM, Weissman Colledge WH, Caraty A, Aparicio SA. 2005. Kisspeptin
BE, Welch DR. 1996. KiSS-1, a novel human malignant directly stimulates gonadotropin-releasing hormone
melanoma metastasis-suppressor gene. Journal of the release via G protein-coupled receptor 54. Proceedings of
National Cancer Institute 88, 1731–1737. the National Academy of Sciences of the United States of America
Lehman MN, Coolen LM, Goodman RL. 2010. Minireview: 102, 1761–1766.
kisspeptin/neurokinin B/dynorphin (KNDy) cells of the Mori Y, Nishihara M, Tanaka T, Shimizu T, Yamaguchi M,
arcuate nucleus: a central node in the control of Takeuchi Y, Hoshino K. 1991. Chronic recording of elec-
gonadotropin-releasing hormone secretion. Endocrinology trophysiological manifestation of the hypothalamic
151, 3479–3489. gonadotropin-releasing hormone pulse generator activity
Lehman MN, Ebling FJ, Moenter SM, Karsch FJ. 1993. Dis- in the goat. Neuroendocrinology 53, 392–395.
tribution of estrogen receptor-immunoreactive cells in the Navarro VM, Castellano JM, McConkey SM, Pineda R,
sheep brain. Endocrinology 133, 876–886. Ruiz-Pino F, Pinilla L, Clifton DK, Tena-Sempere M,
Lents CA, Heidorn NL, Barb CR, Ford JJ. 2008. Central Steiner RA. 2011a. Interactions between kisspeptin and
and peripheral administration of kisspeptin activates neurokinin B in the control of GnRH secretion in the
gonadotropin but not somatotropin secretion in prepu- female rat. American Journal of Physiology Endocrinology and
bertal gilts. Reproduction 135, 879–887. Metabolism 300, E202–E210.

Animal Science Journal (2013) 84, 369–381 © 2013 Japanese Society of Animal Science
380 H. OKAMURA et al.

Navarro VM, Gottsch ML, Chavkin C, Okamura H, Clifton immunofluorescence and confocal microscopy. Endo-
DK, Steiner RA. 2009. Regulation of gonadotropin- crinology 149, 4387–4395.
releasing hormone secretion by kisspeptin/dynorphin/ Rance NE, Krajewski SJ, Smith MA, Cholanian M, Dacks PA.
neurokinin B neurons in the arcuate nucleus of the 2010. Neurokinin B and the hypothalamic regulation of
mouse. The Journal of Neuroscience 29, 11859–11866. reproduction. Brain Research 1364, 116–128.
Navarro VM, Gottsch ML, Wu M, García-Galiano D, Hobbs SJ, Redmond JS, Macedo GG, Velez IC, Caraty A, Williams GL,
Bosch MA, Pinilla L, Clifton DK, Dearth A, Ronnekleiv OK, Amstalden M. 2011. Kisspeptin activates the
Braun RE, Palmiter RD, Tena-Sempere M, Alreja M, hypothalamic-adenohypophyseal-gonadal axis in prepu-
Steiner RA. 2011b. Regulation of NKB pathways and their bertal ewe lambs. Reproduction 141, 541–548.
roles in the control of Kiss1 neurons in the arcuate nucleus Richard N, Corvaisier S, Camacho E, Kottler M-L. 2009.
of the male mouse. Endocrinology 152, 4265–4275. KiSS-1 and GPR54 at the pituitary level: overview and
Nishihara M, Sano A, Kimura F. 1994. Cessation of the recent insights. Peptides 30, 123–129.
electrical activity of gonadotropin-releasing hormone Roseweir AK, Kauffman AS, Smith JT, Guerriero KA,
pulse generator during the steroid-induced surge of lutei- Morgan K, Pielecka-Fortuna J, Pineda R, Gottsch ML,
nizing hormone in the rat. Neuroendocrinology 59, 513–519. Tena-Sempere M, Moenter SM, Terasawa E, Clarke IJ,
Oakley AE, Clifton DK, Steiner RA. 2009. Kisspeptin signal- Steiner RA, Millar RP. 2009. Discovery of potent kisspep-
ing in the brain. Endocrine Reviews 30, 713–743. tin antagonists delineate physiological mechanisms of
Ohkura S, Takase K, Matsuyama S, Mogi K, Ichimaru T, gonadotropin regulation. The Journal of Neuroscience 29,
Wakabayashi Y, Uenoyama Y, Mori Y, Steiner RA, 3920–3929.
Tsukamura H, Maeda K-I OH. 2009b. Gonadotrophin- Saito H, Sawada T, Yaegashi T, Goto Y, Jin J, Sawai K,
releasing hormone pulse generator activity in the Hashizume T. 2012. Kisspeptin-10 stimulates the release
hypothalamus of the goat. Journal of Neuroendocrinology of luteinizing hormone and testosterone in pre- and post-
21, 813–821. pubertal male goats. Animal Science Journal 83, 487–492.
Ohkura S, Tanaka T, Kuroiwa T, Wakabayashi Y, Ohtaki T, Sakamoto K, Murata K, Wakabayashi Y, Yayou K-I, Ohkura
Kusaka M, Okamura H. 2011. Effects of metastin/ S, Takeuchi Y, Mori Y, Okamura H. 2012. Central admin-
kisspeptin analog, TAK-683, on luteinizing hormone istration of neurokinin B activates kisspeptin/NKB
secretion in peripheral plasma, and gonadotropin- neurons in the arcuate nucleus and stimulates luteinizing
releasing hormone secretion in the pituitary portal circu- hormone secretion in ewes during the non-breeding
lation in goats. Endocrine Reviews 32, P2–273. season. The Journal of Reproduction and Development 58,
Ohkura S, Uenoyama Y, Yamada S, Homma T, Takase K, 700–706.
Inoue N, Maeda K-I, Tsukamura H. 2009a. Physiological Schillo KK. 1992. Effects of dietary energy on control of
role of metastin/kisspeptin in regulating gonadotropin- luteinizing hormone secretion in cattle and sheep. Journal
releasing hormone (GnRH) secretion in female rats. Pep- of Animal Science 70, 1271–1282.
tides 30, 49–56. Sébert M-E, Lomet D, Saïd SB, Monget P, Briant C,
Ohtaki T, Shintani Y, Honda S, Matsumoto H, Hori A, Scaramuzzi RJ, Caraty A. 2010. Insights into the mecha-
Kanehashi K, Terao Y, Kumano S, Takatsu Y, Masuda Y, nism by which kisspeptin stimulates a preovulatory LH
Ishibashi Y, Watanabe T, Asada M, Yamada T, Suenaga M, surge and ovulation in seasonally acyclic ewes: potential
Kitada C, Usuki S, Kurokawa T, Onda H, Nishimura O, role of estradiol. Domestic Animal Endocrinology 38, 289–
Fujino M. 2001. Metastasis suppressor gene KiSS-1 298.
encodes peptide ligand of a G-protein-coupled receptor. Seminara SB, Dipietro MJ, Ramaswamy S, Crowley WF,
Nature 411, 613–617. Plant TM. 2006. Continuous human metastin 45–54 infu-
Pheng V, Uenoyama Y, Homma T, Inamoto Y, Takase K, sion desensitizes G protein-coupled receptor 54-induced
Yoshizawa-Kumagaye K, Isaka S, Watanabe TX, Ohkura gonadotropin-releasing hormone release monitored indi-
S, Tomikawa J, Maeda K-I, Tsukamura H. 2009. Potencies rectly in the juvenile male Rhesus monkey (Macaca
of centrally- or peripherally-injected full-length kisspep- mulatta): a finding with therapeutic implications. Endo-
tin or its C-terminal decapeptide on LH release in intact crinology 147, 2122–2126.
male rats. The Journal of Reproduction and Development 55, Seminara SB, Messager S, Chatzidaki EE, Thresher RR,
378–382. Acierno JS, Shagoury JK, Bo-Abbas Y, Kuohung W,
Pielecka-Fortuna J, Chu Z, Moenter SM. 2008. Kisspeptin Schwinof KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB,
acts directly and indirectly to increase gonadotropin- Slaugenhaupt SA, Gusella JF, O’Rahilly S, Carlton MB,
releasing hormone neuron activity and its effects are Crowley WF Jr, Araricio SA, Colledge WH. 2003. The
modulated by estradiol. Endocrinology 149, 1979–1986. GPR54 gene as a regulator of puberty. The New England
Plant TM, Ramaswamy S, Dipietro MJ. 2006. Repetitive acti- Journal of Medicine 349, 1614–1627.
vation of hypothalamic G protein-coupled receptor 54 Smith JT. 2012. The role of kisspeptin and gonadotropin
with intravenous pulses of kisspeptin in the juvenile inhibitory hormone in the seasonal regulation of repro-
monkey (Macaca mulatta) elicits a sustained train of duction in sheep. Domestic Animal Endocrinology 43, 75–84.
gonadotropin-releasing hormone discharges. Endocrinol- Smith JT, Clay CM, Caraty A, Clarke IJ. 2007. KiSS-1 mes-
ogy 147, 1007–1013. senger ribonucleic acid expression in the hypothalamus of
Pompolo S, Pereira A, Estrada KM, Clarke IJ. 2006. Colocali- the ewe is regulated by sex steroids and season. Endo-
zation of kisspeptin and gonadotropin-releasing hormone crinology 148, 1150–1157.
in the ovine brain. Endocrinology 147, 804–810. Smith JT, Coolen LM, Kriegsfeld LJ, Sari IP,
Ramaswamy S, Guerriero KA, Gibbs RB, Plant TM. 2008. Jaafarzadehshirazi MR, Maltby M, Bateman K, Goodman
Structural interactions between kisspeptin and GnRH RL, Tilbrook AJ, Ubuka T, Bentley GE, Clarke IJ, Lehman
neurons in the mediobasal hypothalamus of the male MN. 2008b. Variation in kisspeptin and RFamide-related
rhesus monkey (Macaca mulatta) as revealed by double peptide (RFRP) expression and terminal connections to

© 2013 Japanese Society of Animal Science Animal Science Journal (2013) 84, 369–381
KISSPEPTIN IN DOMESTIC ANIMALS 381

gonadotropin-releasing hormone neurons in the brain: a neurokinin B in the central control of reproduction.
novel medium for seasonal breeding in the sheep. Endo- Nature Genetics 41, 354–358.
crinology 149, 5770–5782. Tsukamura H, Homma T, Tomikawa J, Uenoyama Y, Maeda
Smith JT, Cunningham MJ, Rissman EF, Clifton DK, Steiner K-I. 2010. Sexual differentiation of kisspeptin neurons
RA. 2005. Regulation of Kiss1 gene expression in the responsible for sex difference in gonadotropin release in
brain of the female mouse. Endocrinology 146, 3686–3692. rats. Annals of the New York Academy of Sciences 1200,
Smith JT, Li Q, Pereira A, Clarke IJ. 2009b. Kisspeptin 95–103.
neurons in the ovine arcuate nucleus and preoptic area Uenoyama Y, Inoue N, Pheng V, Homma T, Takase K,
are involved in the preovulatory luteinizing hormone Yamada S, Ajiki K, Ichikawa M, Okamura H, Maeda K-I,
surge. Endocrinology 150, 5530–5538. Tsukamura H. 2011. Ultrastructural evidence of
Smith JT, Li Q, Yap KS, Shahab M, Roseweir AK, Millar RP, kisspeptin-gonadotrophin-releasing hormone (GnRH)
Clarke IJ. 2011. Kisspeptin is essential for the full preo- interaction in the median eminence of female rats: impli-
vulatory LH surge and stimulates GnRH release from the cation of axo-axonal regulation of GnRH release. Journal
isolated ovine median eminence. Endocrinology 152, of Neuroendocrinology 23, 863–870.
1001–1012. Veldhuis JD, Evans WS, Demers LM, Thorner MO, Wakat D,
Smith JT, Popa SM, Clifton DK, Hoffman GE, Steiner RA. Rogol AD. 1985. Altered neuroendocrine regulation of
2006. Kiss1 neurons in the forebrain as central processors gonadotropin secretion in women distance runners. The
for generating the preovulatory luteinizing hormone Journal of Clinical Endocrinology and Metabolism 61, 557–
surge. The Journal of Neuroscience 26, 6687–6694. 563.
Smith JT, Rao A, Pereira A, Caraty A, Millar RP, Clarke IJ. Wakabayashi Y, Nakada T, Murata K, Ohkura S, Mogi K,
2008a. Kisspeptin is present in ovine hypophysial portal Navarro VM, Clifton DK, Mori Y, Tsukamura H, Maeda
blood but does not increase during the preovulatory lutei- K-I, Steiner RA, Okamura H. 2010. Neurokinin B and
nizing hormone surge: evidence that gonadotropes are dynorphin A in kisspeptin neurons of the arcuate nucleus
not direct targets of kisspeptin in vivo. Endocrinology 149, participate in generation of periodic oscillation of neural
1951–1959. activity driving pulsatile gonadotropin-releasing hormone
Smith JT, Saleh SNH, Clarke IJ. 2009a. Seasonal and cyclical secretion in the goat. The Journal of Neuroscience 30, 3124–
change in the luteinizing hormone response to kisspeptin 3132.
in the ewe. Neuroendocrinology 90, 283–291. Wakabayashi Y, Yamamura T, Ohkura S, Homma T,
Suzuki S, Kadokawa H, Hashizume T. 2008. Direct Sakamoto K, Mori Y, Okamura H. 2012. Senktide, a neu-
kisspeptin-10 stimulation on luteinizing hormone secre- rokinin B receptor agonist, stimulates pulsatile LH secre-
tion from bovine and porcine anterior pituitary cells. tion through a mechanism mediated by the GnRH pulse
Animal Reproduction Science 103, 360–365. generator in goats. Program of the 42nd Society for Neuro-
Takase K, Ohkura S, Imamura T, Uenoyama Y, Tsukamura H, science Annual Meeting; 13–17 October 2012, New Orleans,
Maeda KI, Okamura H. 2007. Expression of metastin/ USA, Abstract 90-07.
kisspeptin, a product of KiSS-1 gene, in the goat hypotha- Wakabayashi Y, Yamamura T, Ohkura S, Tanaka T, Kusaka
lamus. Program of the 37th Society for Neuroscience Annual M, Okamura H. 2011. Effects of chronic administration of
Meeting; 12–16 November 2007, San Diego, USA, Abstract a metastin analog, TAK-683, on the GnRH pulse genera-
194.110. tor activity in ovariectomized goats. Endocrine Reviews 32,
Tanaka T, Ohkura S, Wakabayashi Y, Okamura H. 2012. P2–264.
Effect of peripherally administered kisspeptin-10 on Wakabayashi Y, Yamamura T, Sakamoto K, Mori Y, Okamura
GnRH neurosecretion into the hypophyseal portal circu- H. 2013. Electrophysiological and morphological evidence
lation in ovariectomized goat does. Small Ruminant for synchronized GnRH pulse generator activity among
Research 105, 273–276. kisspeptin/neurokinin B/dynorphin A (KNDy) neurons in
Tomikawa J, Homma T, Tajima S, Shibata T, Inamoto Y, goats. The Journal of Reproduction and Development 59,
Takase K, Inoue N, Ohkura S, Uenoyama Y, Maeda K-I, 40–48.
Tsukamura H. 2010. Molecular characterization and Whitlock BK, Daniel JA, Wilborn RR, Maxwell HS, Steele BP,
estrogen regulation of hypothalamic KISS1 gene in the Sartin JL. 2010. Interaction of kisspeptin and the soma-
pig. Biology of Reproduction 82, 313–319. totropic axis. Neuroendocrinology 92, 178–188.
Tomikawa J, Uenoyama Y, Ozawa M, Fukanuma T, Takase K, Whitlock BK, Daniel JA, Wilborn RR, Rodning SP, Maxwell
Goto T, Abe H, Ieda N, Minabe S, Deura C, Inoue N, HS, Steele BP, Sartin JL. 2008. Interaction of estrogen and
Sanbo M, Tomita K, Hirabayashi M, Tanaka S, Imamura T, progesterone on kisspeptin-10-stimulated luteinizing
Okamura H, Maeda K-I, Tsukamura H. 2012. Epigenetic hormone and growth hormone in ovariectomized cows.
regulation of Kiss1 gene expression mediating estrogen- Neuroendocrinology 88, 212–215.
positive feedback action in the mouse brain. Proceedings of Wintermantel TM, Campbell RE, Porteous R, Bock D, Gröne
the National Academy of Sciences of the United States of America H-J, Todman MG, Korach KS, Greiner E, Pérez CA, Schütz
109, E1294–E1301. G, Herbison AE. 2006. Definition of estrogen receptor
Tomita K, Oishi S, Ohno H, Peiper SC, Fujii N. 2008. Devel- pathway critical for estrogen positive feedback to
opment of novel G-protein-coupled receptor 54 agonists gonadotropin-releasing hormone neurons and fertility.
with resistance to degradation by matrix metalloprotein- Neuron 52, 271–280.
ase. Journal of Medicinal Chemistry 51, 7645–7649. Yamamura T, Wakabayashi Y, Noguchi T, Oishi S, Fujii N,
Topaloglu AK, Reimann F, Guclu M, Yalin AS, Kotan LD, Ohkura S, Okamura H. 2012. Continuous administration
Porter KM, Serin A, Mungan NO, Cook JR, Ozbek MN, of senktide, a neurokinin B receptor agonist, facilitates
Imamoglu S, Akalin NS, Yuksel B, O’Rahilly S, Semple intermittent activities of the GnRH pulse generator for 24
RK. 2009. TAC3 and TACR3 mutations in familial h in goats. 2nd World Conference of Kisspeptin Signaling in the
hypogonadotropic hypogonadism reveal a key role for Brain; 6–9 November 2012, Tokyo, Japan, p. 55.

Animal Science Journal (2013) 84, 369–381 © 2013 Japanese Society of Animal Science
Copyright of Animal Science Journal is the property of Wiley-Blackwell and its content may not be copied or
emailed to multiple sites or posted to a listserv without the copyright holder's express written permission.
However, users may print, download, or email articles for individual use.

Potrebbero piacerti anche