You are on page 1of 8

Reproduction and Breeding 1 (2021) 128–135

Contents lists available at ScienceDirect

Reproduction and Breeding


journal homepage: www.keaipublishing.com/en/journals/reproduction-and-breeding

A review on ghrelin and fish reproduction


Huan Zhong a, Yi Hu a, Fan Yu b, *
a
Hunan Research Center of Engineering Technology for Utilization of Distinctive Aquatic Resources, College of Animal Science and Technology, Hunan Agricultural
University, Changsha, 410128, China
b
Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery
Sciences, Wuxi, 214081, China

A R T I C L E I N F O A B S T R A C T

Keywords: Ghrelin is a multifunctional peptide hormone which dominantly secreted in stomach and intestine. Ghrelin in-
Hormone volves in growth control, food intake, energy balance, and reproductive process. Recent evidences suggested that
Gonad ghrelin also interacts with hypothalamic–pituitary–gonadal axis (HPG axis) and controls gametes maturation. In
Gamete maturation
addition, current clues showed that via regulating ghrelin system (acyl ghrelin: des acyl ghrelin ratio, GOAT, GHS-
Hypothalamic–pituitary–gonadal axis
Diets
R1a), glucose and weight could be regulated promising a potential way for artificial controlling of gonadal
development and gamete maturation. The ghrelin signaling system may be a new target for reproductive oper-
ation in fish artificial propagation and breeding. This review summarizes the recent studies of the function of
ghrelin in fish reproduction and the regulatory methods for ghrelin system.

1. Introduction The following clues showed the function of ghrelin in food intake and
energy balance in human and animals [14–16]. Regarding the regulation
Food intake and energy balance are two essential biological processes of ghrelin on energy homeostasis, abundant studies have been focused on
in fishes which guarantee sufficient energy for fish growth, reproduction, its role in etiology such as obesity [17], cardiovascular diseases [18], and
and other behaviors to survive [1,2]. Several regulatory networks have diabetes [19]. Additionally, numerous medicines have been developed to
been found to participate in controlling food intake and energy balance, intervene the ghrelin-GHS-R axis to cure these diseases which have ob-
such as hypothalamic–pituitary–adrenal axis (HPA axis) [3], hypothal- tained good effects [20–22]. Simultaneously, multiple biological func-
amic–pituitary–gonadal axis (HPG axis) [4], and brain-gut axis [5]. In tions associated with ghrelin-GHS-R axis have been found [23,24].
these axes, hormones are dominant factors to control food intake and Recently, ghrelin reduced the expression of autophagy-related genes in
energy balance [6,7]. For example, as a key member of HPG axis, obesity-associated type 2 diabetes, which was reported to promise a
melanin-concentrating hormone (MCH) involves in food intake regula- novel molecular function and promise a potential treatment strategy
tion, which is mainly expressed in pituitary gland [7]. [25]. In fishes, ghrelin is a concerned intestinal endocrine hormone and
Gonadotropin-releasing hormone (GnRH) as one of the known hormones has potential value for artificial control of organism growth and feed
secreted by hypothalamus is generally used for inducing fish ovulation, utilization [26]. It has been shown that ghrelin-GHS-R axis connected
which also has function in regulation of feeding by interacting with with growth hormone (GH)-insulin-like growth factor-1 (IGF-1) axis to
orexin [8]. Commonly, food intake and energy balance have been regulate fish growth [27,28]. The communication between
regarded under control of central nervous system (CNS) [9]. Besides to ghrelin-GHS-R axis and HPG axis affects gonadal development of fishes
CNS, enteroendocrine hormones also play a crucial role in regulation of [29]. Previously, we found that injection of ghrelin could increase sur-
food intake [10]. Intestinal hormones and neuropeptide hormones such vival rates of tilapia with Aeromonas hydrophila infection [30]. Ghrelin
as ghrelin have been found acting on hypothalamus and limbic system also has functions in anti-inflammation, regulating insulin release and
stimulating appetite [11,12]. glucose metabolism in fishes [31]. Thus, ghrelin is a central factor that
Ghrelin was first reported in rat and human stomach which has also regulates organism growth, food intake, energy homeostasis,
been considered as a growth hormone secretagogue (GHS) by interacting anti-pathogens and autoimmune [26]. These evidences suggest that
with its receptor growth hormone secretagogue receptor (GHS-R) [13]. digestive tract as the main secretion site of ghrelin is not only a digestive

* Corresponding author.
E-mail address: yuf@ffrc.cn (F. Yu).

https://doi.org/10.1016/j.repbre.2021.07.004
Received 23 May 2021; Received in revised form 13 July 2021; Accepted 26 July 2021
Available online 4 August 2021
2667-0712/© 2021 The Authors. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co. Ltd. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
H. Zhong et al. Reproduction and Breeding 1 (2021) 128–135

organ, but also a multifunctional organ. Table 1


Development of germ cells is crucial for lasting of germline and Several studies of the tissue distributions of ghrelin in fishes.
concerned by researchers for fish breeding and farmers [32,33]. Culti- Species Tissue Method References
vation of broodstock could promise mature gametes for breeding. Thus,
Goldfish intestine Northern blot [60]
finding strategies for inducing high-quality broodstock via nutrient Sea bass proximal intestine, stomach, brain real-time [61]
regulation may provide new approach to increase the fertility and fry quantitative
quality [34]. Up to date, the requirement of broodstock has been widely PCR
investigated and the feeding formula as well as additives [35,36]. These Zebrafish ovary, testis, brain, liver, skin, RT-PCR analysis [62]
heart, gill, muscle and gut
new diets improved the effect of broodstock cultivation. Farmed fish are Common foregut, hindgut, spleen and brain RT-PCR [63]
various and single feeding formula and additive may not satisfy the carp
requirement of different species. Therefore, understanding the nutrient Channel high levels in the stomach real-time [64]
needs and its molecular mechanism is important for cultivation of catfish moderate levels in the pancreas quantitative
and gall bladder PCR
broodstock. Digestive tract is the first object for nutrient absorption and
low levels in the hypothalamus,
sensitive to nutrients after feeding. The secreted hormones such as pituitary, gill, muscle, liver,
ghrelin from digestive tract could response to nutrient and act on CNS to spleen, trunk kidney, head kidney,
regulate the behavior of food intake [37]. In addition, ghrelin as key node antierior intestine, middle
factor had multiple physiological functions. With increasing evidences intestine, posterior intestine
Japanese stomach Northern blot [56]
indicating ghrelin plays an important role in fish reproduction, this eel analysis
article provides an overview of the update studies on basic information of Japanese brain, heart, stomach, intestines, RT-PCR analysis [56]
ghrelin, its function in reproduction and potential approach to regulated eel body kidney and head kidney
ghrelin artificially. Seabream stomach RT-PCR [59]
Atlantic stomach (low expression was qPCR [65]
salmon detected by qPCR in pyloric caeca)
2. Ghrelin Grass carp highest in the foregut real-time [66]
moderate in the muscle, liver, quantitative
2.1. Structure and expression hypothalamus, white adipose PCR
tissue, midgut, heart, and pituitary
low in hindgut, spleen, gill
Ghrelin has been regarded as a 28-amino-acid peptide when it was Blunt snout intestine (foregut, midgut, qPCR [54]
first identified in mammalian [38]. This peptide hormone is encoded by bream hindgut), muscle, liver, heart,
ghrelin and obestatin prepropeptide (GHRL) gene [39]. GHRL has four spleen, gill, kidney, pituitary,
exons in human which produces ghrelin and obestatin [40]. GHRL is hypothalamus
transcribed into proghrelin in ribosomes. Then, the proghrelin is acylated
by ghrelin O-acyltransferase (GOAT) in endoplasmic reticulum [41]. red-bellied piranha (Pygocentrus nattereri), ghrelin is a widespread gene
Finally, the acylated proghrelin is transferred into Golgi apparatus via that expresses in brain, pyloric caeca, and intestine [58]. On contrary,
vesicle transporting and then cut into mature ghrelin [42]. The mature ghrelin was only secreted in stomach in black seabream (Acanthopagrus
acylated ghrelin could be transferred into different organs through blood schlegeli) [59]. These results indicated that the expression sites of ghrelin
circulatory system and interacted with specific GHS-R [43]. With large are different depending on species.
amount of ghrelin proteins have been identified, the sequences of amino
acids show diversity in different species. In goldfish (Carassius auratus),
11 different molecular forms of ghrelin were identified [44]. Goldfish is a 2.2. GHRL and ghrelin in different species
species experienced polyploidization [45]. Therefore, it is reasonable to
contain different ghrelin genes in the duplicated genome as tetraploid. We compared the deduced GHRL protein from 7 fishes including 4
The synthesized 12 amino-acid (GTSFLSPAQKPQ) form shows similar species of Cyprinid, 1 species of Siluriformes, 1 species of Salmoniformes
effects and has been used for the function study of ghrelin in vitro [44]. and 1 species of Perciformes (The fishes contain Danio rerio, Ctenophar-
Despite of the diversity of ghrelin mature peptides, the serine in third yngodon idella, Cyprinus carpio, Carassius auratus, Ictalurus punctatus,
amino acid is conserved from fish to mammalian [46]. This residue is the Salmo salar and Sparus aurata). The prepropeptide length is ranged from
site for ghrelin acylation. Acyl ghrelin plays its biological function via 103 to 108 amino acids and the similarities covered from 35.00% to
interacting with GHS-R [47,48]. Interestingly, GHS-R contains two 89.32% (Table 2). Meanwhile, the mature ghrelin was not conversed in
different receptors including GHS-R1a and GHS-R1b. Only GHS-R1a has length either which had 19-22 amino acids in these fishes (Fig. 1A).
function while GHS-R1b is a truncated non-function receptor which Noticeably, the third residues of the mature peptide were serine in all the
could not interact with acyl ghrelin [49–51]. fishes suggesting the conserved sites for acylation [38]. The phylogenetic
Stomach and intestine are the dominant tissues for ghrelin secretion. analysis showed that the ghrelin in vertebrates grouped into five main
As reported in mammals, high expression of ghrelin was found in the clades corresponding to the classification in the evolution. The phylo-
stomach, while low expression of ghrelin was shown in small and large genetic tree indicated that Cyprinidae ghrelin clustered with other teleost
intestines [52]. Some studies suggested that ghrelin could be generated homologues, such as Siluridae and Salmonidae ghrelin (Fig. 1B). These
by CNS [53]. In stomach, gastric X/A-like endocrine cell is the main results suggested that ghrelin had the similar function in the animals. The
secretion cell types for ghrelin production. In the fish without stomach, GHRL proteins in Amphibian, Reptilian, Avian, and Mammalian had less
the ghrelin expressed in intestine [54,55]. However, the origin of the similarity with the GHRL proteins in Teleost which were not clustered
intestinal cells that secreted ghrelin is still blurred. In goldfish, a species together.
lacking stomach, it showed that mucosal cells were the primary synthe-
sizing site for ghrelin by in situ hybridization [51]. Northern blot and 2.3. Ghrelin acylation
RT-PCR analyses suggested stomach had high ghrelin expression and low
expression in other tissues including brain, intestine, kidney, and After translation into proghrelin, the protein is acylated by GOAT and
head-kidney in Japanese eel (Anguilla japonica) [56]. Meanwhile, ghrelin fragmented into mature ghrelin in Golgi apparatus [41]. In the classical
mRNA had high expression in nighttime displaying a daily rhythm [57] theory, des-acyl ghrelin has no biological function because des-acyl
(Table 1). Considering the starving status of fish at night, this finding may ghrelin could not interact with GHS-R [67]. Although the evidences are
also result from the different energy balance in the daily rhythm. In relatively scarce, des-acyl ghrelin also induces food intake, impairs

129
H. Zhong et al. Reproduction and Breeding 1 (2021) 128–135

Table 2
Similarities (%) of amino acids of prepropeptide from the compared species.
Ictalurus punctatus Carassius auratus Danio rerio Ctenopharyngodon idella Cyprinus carpio Salmo salar Sparus aurata

Ictalurus punctatus 100 53.4 55.88 57.84 59.8 49.51 39.42


Carassius auratus 53.4 100 74.76 82.52 82.52 44.12 35
Danio rerio 55.88 74.76 100 84.47 80.58 48.51 38.38
Ctenopharyngodon idella 57.84 82.52 84.47 100 89.32 47.52 37.37
Cyprinus carpio 59.8 82.52 80.58 89.32 100 46.53 37.37
Salmo salar 49.51 44.12 48.51 47.52 46.53 100 57.84
Sparus aurata 39.42 35 38.38 37.37 37.37 57.84 100

orexigenic effect, decreases fat mass and increases muscle regeneration [83]. Similarly, in dairy heifers, ghrelin injection suppresses GnRH
[68]. Due to the multiple functions of des-acyl ghrelin, Delhanty and his induced preovulatory gonadotropin surge [84]. Ghrelin also was proven
colleagues proposed that des-acyl ghrelin should be regarded as a sepa- that had inhibitory effects on GnRH-induced gonadotropin secretion via
rate hormone besides of acyl ghrelin [69]. Compared to des-acyl ghrelin, acted on hypothalamic in rat [85]. Meanwhile, ghrelin also acts on pi-
acyl ghrelin interacts with GHS-R1a to regulate several biological pro- tuitary directly. In human, ghrelin suppresses FSH and LH secretion [86].
cesses. This classical pathway is the main functional system for ghrelin. These similar suppressive effects of ghrelin on LH secretion were also
Thus, as a regulator, GOAT could control ratios of acyl ghrelin/des-acyl found in rat (Rattus norvegicus) [87], ovariectomized rhesus monkey
ghrelin, which contribute to regulate the energy balance of organisms (Macaca mulatta) [88], and ovariectomized sheep (Ovis aries) [89].
[70]. Unlike ghrelin, GOAT is a wide expression gene in tissues. The two However, in rat, FSH was not changed in vivo and induced in vitro by
types of GOAT (GOAT-v1 and GOAT-v2) from goldfish were both highly ghrelin treatment. In fishes, such as goldfish and common carp (Cyprinus
expressed in gonad and intestine, but lowly expressed in other tested carpio), ghrelin has been reported as a stimulator for LH releasing
tissues [71]. These results showed that GOAT is not only expressed in the [90–92]. The ghrelin regulated Ca- and Protein kinase C (PKC)-mediated
tissues that express ghrelin, suggesting GOAT has extensive functions. pathways participate in the upregulation of LH in goldfish [93]. The
Thus, more studies are needed to elucidate the functional details of GOAT different effects of fish and mammalian may due to the different treat-
especially the functions of GOAT in other tissue rather than digestive ment conditions (dosage, time, in vivo or in vitro, etc) and species. GOAT
tract. Taken together, ghrelin, GOAT and GHS-R1a constitute the ghrelin and GHS-R1a have been shown high expressed in testes and ovaries in
system to regulate food intake and energy balance. several fishes providing clues of regulatory role of ghrelin system on
gonads in a direct way. Ishikawa and his colleagues found that ghrelin
3. Ghrelin's function in energy balance and reproduction expressed in human testis by immunohistochemistry [94]. The ghrelin
protein expression was also found in rat testis [95]. In addition, Chicken
3.1. Ghrelin's role in food intake and energy balance (Gallus gallus domesticus) ovary expressed ghrelin mRNA was reported
[96]. Ovarian follicle cells in zebrafish had ghrelin immunoreactivity
The energy metabolism regulated by ghrelin is mediated by control- signals showing protein expression [96]. In Nile tilapia (Oreochromis
ling of food intake and energy conversion. For food intake, ghrelin and its niloticus), no mRNA expression was detected in gonad but specific
receptor stimulate neuropeptide Y (NPY)/agouti-related protein (AgRP) expressed in stomach. This study also showed that females had higher
neurons via interacting with mammalian target of rapamycin complex 1 mRNA expression of ghrelin compared to males [97]. Thus, ghrelin ex-
(mTORC1)/mammalian target of rapamycin complex 1 (S6K1) pathway presses in testis and ovary depends on species, developmental stage, and
in neurons of arcuate nucleus from hypothalamus [72]. The activated sex. Meanwhile, the reported evidences indicated that ghrelin may act on
NPY/AgRP neurons express NPY and AgRP to induce appetite [73]. For testis and ovary directly [98]. Recent studies also showed that ghrelin
energy metabolism, ghrelin system regulates glucose metabolism via could be regulated by hormones from HPG axis [99,100]. All these clues
stimulating hepatic glycolysis, decreasing activity of glycogen synthase suggest that ghrelin and HPG axis constitute a network to co-regulate
kinase, promoting peroxisome proliferator-activated receptor gamma reproductive activities in fishes (Fig. 2).
coactivator 1-α (PGC-1α), and inhibiting insulin secretion [74]. New
studies also reported that ghrelin system regulates lipid metabolism [75]. 3.3. Other mechanism of Ghrelin's role in reproduction
However, the ghrelin regulated lipid metabolism in fishes have been
rarely reported. The effects of ghrelin on reproduction could also be mediated by
other mechanisms beside regulating HPG axis. The bovine ghrelin affects
3.2. Ghrelin as a regulator of HPG axis AKT Serine/Threonine Kinase 1 (Akt1) and extracellular signal-regulated
protein kinase (ERK1/2) pathways of oocytes with different effects
In fishes, as in other vertebrates, HPG axis is the central axis to control depending on the treated time [101]. Another study suggested that
gonadal development and reproductive behavior [76,77]. In this axis, the ghrelin accelerated the maturation of oocyte in vitro exerting a specific
most upstream tissue is hypothalamus. Hypothalamus is a part of brain and direct role [102]. In zebrafish, ghrelin could inhibit basal and MIH
secretes GnRH [78]. Via binding to its receptor (GnRHR) on the adeno- induced stage IV germinal vesicle breakdown (GVBD) [103]. Intraperi-
hypophysis by traveling down the anterior portion of pituitary, GnRH toneal injection with human ghrelin increased number of mature ovarian
induces the secreting cells to produce follicle-stimulating hormone (FSH) follicles but reduced the diameters of the oocyte in Barbus sharpeyi [104].
and luteinizing hormone (LH) [79,80]. After released into blood stream, These results indicate that the effects of ghrelin on gametes maturation is
FSH and LH could travel to gonad and interacted with their specific re- not only mediated by HPG axis. Unfortunately, the details of the direct
ceptors FSHR and LHR, respectively [81]. The FSH and LH signals effects of ghrelin on follicle developments are still not systematically
stimulate testis and ovary to produce sex steroid hormones such as pro- investigated. Two potential mechanisms may be participated by ghrelin
gesterone, estradiol, and testosterone [82]. The sex steroid hormones act in the gonads: 1. Ghrelin directly interacts with GHS-R1a in gonads and
on gonads and other tissues to regulate sexual behaviors, gonadal switch on downstream pathways such as Akt1 and ERK1/2 pathways to
development, and final ovulation. control the gonad maturation; 2. As a key regulator for energy meta-
In mammalian, several studies reported that ghrelin depressed HPG bolism, ghrelin control the energy utilization for gonadal development
axis. Ghrelin decreased fire activity of GnRH neurons in C57BL/6J mice (Fig. 2).

130
H. Zhong et al. Reproduction and Breeding 1 (2021) 128–135

Fig. 1. Phylogenetic analysis of preproghrelin in vertebrates. A. Multiple sequence alignment of preproghrelin peptides of 7 fishes. The red box showed mature ghrelin
peptide. B. Neighbor Joining phylogenetic tree in vertebrates based on the amino acid sequences by MEGA4 (http://www.megasoftware.net/). The numbers at the
branches were bootstrap values (1000 repetitions). All the sequences were downloaded from NCBI GenBank database: Danio rerio: NP_001077341.1; Cyprinus carpio:
BAF95542.1; Sinocyclocheilus anshuiensis: XP_016314450.1; Ctenopharyngodon idella: AFI98391.1; Carassius auratus: XP_026069130.1; Anabarilius grahami:
ROL50687.1; Labeo rohita: BAX76678.1; Astyanax mexicanus: XP_022531474.1; Ictalurus punctatus: NP_001187244.1; Salmo salar: NP_001133057.1; Clupea harengus:
XP_031423948.1; Oncorhynchus mykiss: XP_021423083.2; Pangasianodon hypophthalmus: XP_026801445.2; Piaractus brachypomus: AJO68701.1; Salvelinus alpinus:
XP_023851722.1; Silurus meridionalis: AER12037.1; Tachysurus fulvidraco: XP_026999190.1; Acanthopagrus latus: XP_036957504.1; Anarrhichthys ocellatus:
XP_031704024.1; Morone saxatilis: XP_035517613.1; Anguilla anguilla: XP_035237098.1; Lepisosteus oculatus: XP_015202865.1; Sparus aurata: XP_030274358.1; Bufo
japonicus: BAM29298.1; Bufo bufo: XP_040264397.1; Rhinatrema bivittatum: XP_029453476.1; Dermochelys coriacea: XP_038265762.1; Trachemys scripta elegans:
XP_034632246.1; Nothoprocta pentlandii: NWX88017.1; Gallus gallus: NP_001001131.1; Homo sapiens: NP_001128413.1; Mus musculus: NP_001273333.1; Rattus nor-
vegicus: NP_067701.1.

131
H. Zhong et al. Reproduction and Breeding 1 (2021) 128–135

Fig. 2. Representation of the ghrelin-HPG axis crosstalk and its role in reproduction. “?” represented the key mechanism that is still under investigation.

4. Regulation of ghrelin system in reproduction regulation of ghrelin system promises a potential approach to artificial
control of fish reproduction.
4.1. Agonists and inhibitors injection
4.2. Regulation of ghrelin system via diets
Until now, peptide hormones are well investigated because they
could be key targets for regulating biological process. To intervene
Diet control is an easier way to operate in fish farming. Diet regulated
growth performance, energy metabolism, diseases, gonadal develop-
function could be changed by the feeding dosage, feeding times and di-
ment, and reproductive behaviors, several agonists and inhibitors for the
etary formula which contains multiple variables. Therefore, it is easy to
hormones along with their regulated factors and receptors have been
change the feeding strategies. For example, ketogenic diet decreased
developed. In application of aquaculture, the artificial control of HPG
ghrelin and des-acyl ghrelin plasma levels [110]. As a sensor of food
axis is an example. Human chorionic gonadotropin (hCG), luteinizing
supply, abundant feeding inhibits and starvation stimulates ghrelin sys-
hormone-releasing hormone agonist (LHRH-A) and domperidone (DOM)
tem. However, feeding conditions (feeding, fasting, fasting-refeeding)
injections change the activities of HPG axis to regulate gonadal matu-
affects multiple molecular pathways and biological processes. Intake of
ration and ovulation which have made a great economic value for world
medium-chain fatty acids could increase the acyl modification of ghrelin
aquaculture [105]. Several agonists and inhibitors have been developed
[111]. This finding gave a new thought to control levels of acyl ghrelin
in recent years. The original intention for developing these factors were
via changing feeding formula. Caprylic acid (C8:0) supplement might
controlling human diseases such as diabetes and obesity. Injection with
provide octanoyl-CoA for ghrelin acylation and changed the ratios of acyl
ghrelin have been used for studying its function. Meanwhile, ghrelin has
ghrelin/des-acyl ghrelin [112]. Acyl ghrelin inhibited meiosis of bovine
also been employed to control growth by inducing GH releasing. Acyl
oocytes and resulted in simultaneous maturation of oocytes [101].
ghrelin is the dominate part of ghrelin to play the function, thus, syn-
Although the mechanism of bovine oocyte maturation may be different
thesized acyl ghrelin is also used for stimulating appetite of goldfish
from fish, the approach of caprylic acid intake changes acyl ghrelin levels
[106]. The synthetic peptide (growth hormone releasing peptide 6
may also be used for regulating maturation of fish gametes.
(GHRP-6)) and non-peptide (capromorelin) GHS-R agonists were deter-
mined as regulators for gastric motor [107]. Inhibitors of ghrelin and its
5. Conclusion
receptors were also used for controlling adipogenesis and diabetes. In
addition, GOAT antagonist GO-CoA-Tat, a peptide-based bisubstrate
Ghrelin system is a critical system for energy metabolism and food
analog was designed to depress production of acyl ghrelin [108] which
intake which also plays a key role in fish reproductive processes. Ghrelin
was used for controlling glucose and weight. In male ob/ob mouse which
interacting with hormones from HPG axis representing ghrelin-HPG axis
is an animal model of type II diabetes, inhibition of ghrelin led to improve
crosstalk to regulate gonadal development and gamete maturation.
the phenotypes of reproduction [109]. In contrast to mammalian, ghrelin
However, the gene regulatory networks and functional details are far
in several fishes has positive effects on gonadal maturation. However, the
from systematically understood. As a key node for energy balance and
different effects in animals derived by ghrelin are still needed to be
HPG axis, the feedback of sexual hormones on ghrelin in fish is unveiled.
elucidate to guide the operation. As key regulator system, hormonal
Meanwhile, the energy metabolism regulated by ghrelin during

132
H. Zhong et al. Reproduction and Breeding 1 (2021) 128–135

reproductive cycles in fish is not investigated yet. By elucidation these [24] D. Nikolopoulos, S. Theoharis, G. Kouraklis, Ghrelin, another factor affecting bone
metabolism, Med. Sci. Mon. Int. Med. J. Exp. Clin. Res. 16 (7) (2010)
questions, it is possible to precisely control reproductive cycles, gametes
RA147–RA162.
maturation, and ovulation of fishes. [25] M.-J. Yuan, T. Wang, The new mechanism of Ghrelin/GHSR-1a on autophagy
regulation, Peptides 126 (2020) 170264.
Declaration of competing interest [26] K.S. Kang, S. Yahashi, K. Matsuda, Central and peripheral effects of ghrelin on
energy balance, food intake and lipid metabolism in teleost fish, Peptides 32 (11)
(2011) 2242–2247.
The authors declare no competing financial interests. [27] J. Zhang, W. Ma, Y. He, J. Wu, F.U. Dawar, F. Ren, X. Zhao, J. Mei, Sex biased
expression of ghrelin and GHSR associated with sexual size dimorphism in yellow
catfish, Gene 578 (2) (2016) 169–176.
Acknowledgments [28] E.M. Hevrøy, C. Azpeleta, M. Shimizu, A. Lanzen, H. Kaiya, M. Espe, P.A. Olsvik,
Effects of short-term starvation on ghrelin, GH-IGF system, and IGF-binding
This research was supported by National Natural Science Foundation proteins in Atlantic salmon, Fish Physiol. Biochem. 37 (1) (2011) 217–232.
[29] S.A. Dar, P.P. Srivastava, Ghrelin and its role in reproductive physiology of fish, in:
of China (31760756) and the Natural Science Foundation of Hunan Recent Updates in Molecular Endocrinology and Reproductive Physiology of Fish,
Province (2021JJ20033). Springer, 2021, pp. 171–179.
[30] Z. Han, Y. Zhou, X. Zhang, J. Yan, J. Xiao, Y. Luo, H. Zheng, H. Zhong, Ghrelin
modulates the immune response and increases resistance to Aeromonas
References hydrophila infection in hybrid tilapia, Fish Shellfish Immunol. 98 (2020)
100–108.
[1] M. Conde-Sieira, J.L. Soengas, Nutrient sensing systems in fish: impact on food [31] K.R. Upton, L.G. Riley, Acute stress inhibits food intake and alters ghrelin
intake regulation and energy homeostasis, Front. Neurosci. 10 (2017) 603. signaling in the brain of tilapia (Oreochromis mossambicus), Domest. Anim.
[2] S. Saravanan, J.W. Schrama, A.C. Figueiredo-Silva, S.J. Kaushik, J.A.J. Verreth, Endocrinol. 44 (3) (2013) 157–164.
I. Geurden, Constraints on energy intake in fish: the link between diet [32] R.S. McBride, S. Somarakis, G.R. Fitzhugh, A. Albert, N.A. Yaragina,
composition, energy metabolism, and energy intake in rainbow trout, PloS One 7 M.J. Wuenschel, A. Alonso-Fernandez, G. Basilone, Energy acquisition and
(4) (2012), e34743. allocation to egg production in relation to fish reproductive strategies, Fish Fish.
[3] N. Bazhan, D. Zelena, Food-intake regulation during stress by the hypothalamo- 16 (1) (2015) 23–57.
pituitary-adrenal axis, Brain Res. Bull. 95 (2013) 46–53. [33] C.C. Mylonas, A. Fostier, S. Zanuy, Broodstock management and hormonal
[4] J. Xu, W. Huang, C. Zhong, D. Luo, S. Li, Z. Zhu, W. Hu, Defining Global gene manipulations of fish reproduction, Gen. Comp. Endocrinol. 165 (3) (2010)
expression changes of the hypothalamic-pituitary-gonadal axis in female sgnrh- 516–534.
antisense transgenic common carp (Cyprinus carpio), PloS One 6 (6) (2011), [34] J. Bobe, C. Labbe, Egg and sperm quality in fish, Gen. Comp. Endocrinol. 165 (3)
e21057. (2010) 535–548.
[5] A. Agustí, M.P. García-Pardo, I. Lopez-Almela, I. Campillo, M. Maes, M. Romaní- [35] W.-K. Ng, Y. Wang, Inclusion of crude palm oil in the broodstock diets of female
Perez, Y. Sanz, Interplay between the gut-brain Axis, obesity and cognitive Nile tilapia, Oreochromis niloticus, resulted in enhanced reproductive performance
function, Front. Neurosci. 12 (2018) 155. compared to broodfish fed diets with added fish oil or linseed oil, Aquaculture 314
[6] J.I. Bertucci, A.M. Blanco, L. Sundarrajan, J.J. Rajeswari, C. Velasco, (1) (2011) 122–131.
S. Unniappan, Nutrient regulation of endocrine factors influencing feeding and [36] H. Migaud, G. Bell, E. Cabrita, B. McAndrew, A. Davie, J. Bobe, M.P. Herraez,
growth in fish, Front. Endocrinol. 10 (2019) 83. M. Carrillo, Gamete quality and broodstock management in temperate fish, Rev.
[7] M. L opez, C.V. Alvarez, R. Nogueiras, C. Dieguez, Energy balance regulation by Aquacult. 5 (s1) (2013) S194–S223.
thyroid hormones at central level, Trends Mol. Med. 19 (7) (2013) 418–427. [37] D.P. Begg, S.C. Woods, The endocrinology of food intake, Nat. Rev. Endocrinol. 9
[8] L.J. Hoskins, M. Xu, H. Volkoff, Interactions between gonadotropin-releasing (10) (2013) 584–597.
hormone (GnRH) and orexin in the regulation of feeding and reproduction in [38] T. Sato, Y. Nakamura, Y. Shiimura, H. Ohgusu, K. Kangawa, M. Kojima, Structure,
goldfish (Carassius auratus), Horm. Behav. 54 (3) (2008) 379–385. regulation and function of ghrelin, J. Biochem. 151 (2) (2012) 119–128.
[9] M.G. Myers, D.P. Olson, Central nervous system control of metabolism, Nature [39] T.D. Ghaleh, S.S. Skandari, R. Najafipour, Z. Rashvand, M. Darabi, M. Sahmani,
491 (7424) (2012) 357–363. Association of 604G/A and 501A/C ghrelin and obestatin prepropeptide gene
[10] R. Latorre, C. Sternini, R. De Giorgio, B. Greenwood-Van Meerveld, polymorphisms with polycystic ovary syndrome, Biochem. Genet. 56 (1) (2018)
Enteroendocrine cells: a review of their role in brain–gut communication, Neuro 116–127.
Gastroenterol. Motil. 28 (5) (2016) 620–630. [40] J.V. Zhang, P.G. Ren, O. Avsian-Kretchmer, C.W. Luo, R. Rauch, C. Klein,
[11] L. Rui, Brain regulation of energy balance and body weight, Rev. Endocr. Metab. A.J.W. Hsueh, Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin’s
Disord. 14 (4) (2013) 387–407. effects on food intake, Science 310 (5750) (2005) 996–999.
[12] N. Delzenne, J. Blundell, F. Brouns, K. Cunningham, K. De Graaf, A. Erkner, [41] M.S. Taylor, Y. Hwang, P.Y. Hsiao, J.D. Boeke, P.A. Cole, Chapter thirteen - ghrelin
A. Lluch, M. Mars, H.P.F. Peters, M. Westerterp-Plantenga, Gastrointestinal targets O-acyltransferase assays and inhibition, in: M. Kojima, K. Kangawa (Eds.),
of appetite regulation in humans, Obes. Rev. 11 (3) (2010) 234–250. Methods in Enzymology, Academic Press, 2012, pp. 205–228.
[13] M. Nakazato, N. Murakami, Y. Date, M. Kojima, H. Matsuo, K. Kangawa, [42] M.P. Cornejo, E.R. Mustafa, D. Cassano, J.L. Baneres, J. Raingo, M. Perello, The
S. Matsukura, A role for ghrelin in the central regulation of feeding, Nature 409 ups and downs of growth hormone secretagogue receptor signaling, FEBS J.
(6817) (2001) 194–198. (2021), https://doi.org/10.1111/febs.15718.
[14] H. Kaiya, M. Furuse, M. Miyazato, K. Kangawa, Current knowledge of the roles of [43] Y. Shuto, T. Shibasaki, K. Wada, I. Parhar, J. Kamegai, H. Sugihara, S. Oikawa,
ghrelin in regulating food intake and energy balance in birds, Gen. Comp. I. Wakabayashi, Generation of polyclonal antiserum against the growth hormone
Endocrinol. 163 (1) (2009) 33–38. secretagogue receptor (GHS-R): evidence that the GHS-R exists in the
[15] Y. Sun, N.F. Butte, J.M. Garcia, R.G. Smith, Characterization of adult ghrelin and hypothalamus, pituitary and stomach of rats, Life Sci. 68 (9) (2001) 991–996.
ghrelin receptor knockout mice under positive and negative energy balance, [44] T. Miura, K. Maruyama, H. Kaiya, M. Miyazato, K. Kangawa, M. Uchiyama,
Endocrinology 149 (2) (2008) 843–850. S. Shioda, K. Matsuda, Purification and properties of ghrelin from the intestine of
[16] E. J€onsson, The role of ghrelin in energy balance regulation in fish, Gen. Comp. the goldfish, Carassius auratus, Peptides 30 (4) (2009) 758–765.
Endocrinol. 187 (2013) 79–85. [45] J. Luo, J. Chai, Y. Wen, M. Tao, G. Lin, X. Liu, L. Ren, Z. Chen, S. Wu, S. Li,
[17] P. Wiedmer, R. Nogueiras, F. Broglio, D. D'Alessio, M.H. Tsch€ op, Ghrelin, obesity Y. Wang, Q. Qin, S. Wang, Y. Gao, F. Huang, L. Wang, C. Ai, X. Wang, L. Li, C. Ye,
and diabetes, Nat. Clin. Pract. Endocrinol. Metabol. 3 (10) (2007) 705–712. H. Yang, M. Luo, J. Chen, H. Hu, L. Yuan, L. Zhong, J. Wang, J. Xu, Z. Du, Z. Ma,
[18] T. Tokudome, K. Otani, M. Miyazato, K. Kangawa, Ghrelin and the heart, Peptides R.W. Murphy, A. Meyer, J. Gui, P. Xu, J. Ruan, Z.J. Chen, S. Liu, X. Lu, Y.-p. Zhang,
111 (2019) 42–46. From asymmetrical to balanced genomic diversification during rediploidization:
[19] A.-L. Poher, M.H. Tsch€ op, T.D. Müller, Ghrelin regulation of glucose metabolism, subgenomic evolution in allotetraploid fish, Science Advances 6 (22) (2020),
Peptides 100 (2018) 236–242. eaaz7677.
[20] J.H. Lee, C. Fang, X. Li, C.S. Wu, J.Y. Noh, X. Ye, R.S. Chapkin, K. Sun, Y. Sun, [46] K. Nagi, A.M. Habib, Biased signaling: a viable strategy to drug ghrelin receptors
GHS-R suppression in adipose tissues protects against obesity and insulin for the treatment of obesity, Cell. Signal. 83 (2021) 109976.
resistance by regulating adipose angiogenesis and fibrosis, Int. J. Obes. 45 (7) [47] E.T. Vestergaard, N. Jessen, N. Møller, J.O.L. Jørgensen, Acyl ghrelin induces
(2021) 1565–1575. insulin resistance independently of GH, cortisol, and free fatty acids, Sci. Rep. 7
[21] C.S. Wu, O.Y.N. Bongmba, J.H. Lee, E. Tuchaai, Y. Zhou, D.P. Li, B. Xue, Z. Chen, (1) (2017) 42706.
Y. Sun, Ghrelin receptor in agouti-related peptide neurones regulates metabolic [48] R. Stark, A. Reichenbach, S.H. Lockie, C. Pracht, Q. Wu, A. Tups, Z.B. Andrews,
adaptation to calorie restriction, J. Neuroendocrinol. 31 (7) (2019), e12763. Acyl ghrelin acts in the brain to control liver function and peripheral glucose
[22] R. Mosa, L. Huang, H. Li, M. Grist, D. LeRoith, C. Chen, Long-term treatment with homeostasis in male mice, Endocrinology 156 (3) (2015) 858–868.
the ghrelin receptor antagonist [d-Lys3]-GHRP-6 does not improve glucose [49] W.J. Cai, X.C. Yuan, Y.C. Yuan, S.Q. Xie, Y. Gong, H. Su, Y. Qiao, Sequence,
homeostasis in nonobese diabetic MKR mice, Am. J. Physiol. Regul. Integr. Comp. genomic organization and expression of ghrelin receptor in grass carp,
Physiol. 314 (1) (2018) R71–R83. Ctenopharyngodon idellus. Comparative Biochemistry and Physiology Part A:
[23] D. Nikolopoulos, S. Theocharis, G. Kouraklis, Ghrelin: a potential therapeutic Molecular & Integrative Physiology 179 (2015) 54–61.
target for cancer, Regul. Pept. 163 (1) (2010) 7–17. [50] P. Yu, Y. Zhou, X. Qi, H. Fan, K. Zhang, X. Zhang, Y. Li, H. Wen, Identification,
expression analysis, and functional characterization of ghrelin and its receptors in

133
H. Zhong et al. Reproduction and Breeding 1 (2021) 128–135

spotted sea bass (Lateolabrax maculatus), Marine Life Science & Technology 2 (4) [76] H. Zhong, J. Hu, Y. Zhou, Transcriptomic evidence of luteinizing hormone-
(2020) 349–359. releasing hormone agonist (LHRH-A) regulation on lipid metabolism in grass carp
[51] A. Sanchez-Breta~ no, A.M. Blanco, S. Unniappan, O. Kah, M.-M. Gueguen, (Ctenopharyngodon idella), Genomics 113 (1, Part 2) (2021) 1265–1271.
 Alonso-G
J.I. Bertucci, A.L. omez, A.I. Valenciano, E. Isorna, M.J. Delgado, In situ [77] A. Hatef, S. Unniappan, Metabolic hormones and the regulation of
localization and rhythmic expression of ghrelin and ghs-r1 ghrelin receptor in the spermatogenesis in fishes, Theriogenology 134 (2019) 121–128.
brain and gastrointestinal tract of goldfish (Carassius auratus), PloS One 10 (10) [78] X. Qi, W. Zhou, S. Li, D. Lu, S. Yi, R. Xie, X. Liu, Y. Zhang, H. Lin, Evidences for the
(2015), e0141043. regulation of GnRH and GTH expression by GnIH in the goldfish, Carassius auratus,
[52] N. Eissa, J.E. Ghia, Immunomodulatory effect of ghrelin in the intestinal mucosa, Mol. Cell. Endocrinol. 366 (1) (2013) 9–20.
Neuro Gastroenterol. Motil. 27 (11) (2015) 1519–1527. [79] J.A. Mu~ noz-Cueto, N. Zmora, J.A. Paullada-Salmer on, M. Marvel, E. Ma~nanos,
[53] S. Ghelardoni, V. Carnicelli, S. Frascarelli, S. Ronca-Testoni, R. Zucchi, Ghrelin Y. Zohar, The gonadotropin-releasing hormones: lessons from fish, Gen. Comp.
tissue distribution: comparison between gene and protein expression, Endocrinol. 291 (2020) 113422.
J. Endocrinol. Invest. 29 (2) (2006) 115–121. [80] Y. Zhou, Y. Niu, M. Tao, X. Deng, S. Liu, Y. Liu, J. Li, Molecular cloning,
[54] W. Ji, H.-C. Ping, K.-J. Wei, G.-R. Zhang, Z.-C. Shi, R.-B. Yang, G.-W. Zou, W.- characterization and expression of FSH and LH beta subunits from grass carp
M. Wang, Ghrelin, neuropeptide Y (NPY) and cholecystokinin (CCK) in blunt (Ctenopharyngodon idella), Fish Physiol. Biochem. 36 (2) (2010) 213–221.
snout bream (Megalobrama amblycephala): cDNA cloning, tissue distribution and [81] S. Burow, N. Mizrahi, G. Maugars, K. von Krogh, R. Nourizadeh-Lillabadi,
mRNA expression changes responding to fasting and refeeding, Gen. Comp. L. Hollander-Cohen, M. Shpilman, I. Atre, F.-A. Weltzien, B. Levavi-Sivan,
Endocrinol. 223 (2015) 108–119. Characterization of gonadotropin receptors Fshr and Lhr in Japanese medaka,
[55] C. Zhou, J. Zheng, L. Lei, D. Yuan, C. Zhu, H. Ye, C. Zhang, D. Wang, M. Yang, Oryzias latipes, Gen. Comp. Endocrinol. 285 (2020) 113276.
J. Wu, L. Zhu, B. Zeng, Evidence that ghrelin may be associated with the food [82] P. Thomas, G. Dressing, Y. Pang, H. Berg, C. Tubbs, A. Benninghoff, K. Doughty,
intake of gibel carp (Carassius auratus gibelio), Fish Physiol. Biochem. 42 (6) Progestin, estrogen and androgen G-protein coupled receptors in fish gonads,
(2016) 1637–1646. Steroids 71 (4) (2006) 310–316.
[56] H. Kaiya, M. Kojima, H. Hosoda, L.G. Riley, T. Hirano, E. Grau, K. Kangawa, [83] I. Farkas, C. Vastagh, M. Sarvari, Z. Liposits, Ghrelin decreases firing activity of
Amidated fish ghrelin: purification, cDNA cloning in the Japanese eel and its gonadotropin-releasing hormone (GnRH) neurons in an estrous cycle and
biological activity, J. Endocrinol. 176 (3) (2003) 415–423. endocannabinoid signaling dependent manner, PloS One 8 (10) (2013), e78178.
[57] S.P. Kalra, M. Bagnasco, E.E. Otukonyong, M.G. Dube, P.S. Kalra, Rhythmic, [84] T.M. Chouzouris, E. Dovolou, K. Dafopoulos, P. Georgoulias, N.G. Vasileiou,
reciprocal ghrelin and leptin signaling: new insight in the development of obesity, G.C. Fthenakis, G. Anifandis, G.S. Amiridis, Ghrelin suppresses the GnRH-induced
Regul. Pept. 111 (1) (2003) 1–11. preovulatory gonadotropin surge in dairy heifers, Theriogenology 86 (6) (2016)
[58] H. Volkoff, Cloning, tissue distribution and effects of fasting on mRNA expression 1615–1621.
levels of leptin and ghrelin in red-bellied piranha (Pygocentrus nattereri), Gen. [85] R. Fernandez-Fernandez, M. Tena-Sempere, V.M. Navarro, M.L. Barreiro,
Comp. Endocrinol. 217–218 (2015) 20–27. J.M. Castellano, E. Aguilar, L. Pinilla, Effects of ghrelin upon gonadotropin-
[59] C.-M. Yeung, C.-B. Chan, N.Y.S. Woo, C.H.K. Cheng, Seabream ghrelin: cDNA releasing hormone and gonadotropin secretion in adult female rats: in vivo and in
cloning, genomic organization and promoter studies, J. Endocrinol. 189 (2) vitro studies, Neuroendocrinology 82 (5–6) (2005) 245–255.
(2006) 365–379. [86] M. Kluge, P. Schüssler, D. Schmidt, M. Uhr, A. Steiger, Ghrelin Suppresses
[60] S. Unniappan, X. Lin, L. Cervini, J. Rivier, H. Kaiya, K. Kangawa, R.E. Peter, secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in
Goldfish ghrelin: molecular characterization of the complementary women, J. Clin. Endocrinol. Metab. 97 (3) (2012) E448–E451.
deoxyribonucleic acid, partial gene structure and evidence for its stimulatory role [87] R. Fernandez-Fernandez, M. Tena-Sempere, E. Aguilar, L. Pinilla, Ghrelin effects
in food intake, Endocrinology 143 (10) (2002) 4143–4146. on gonadotropin secretion in male and female rats, Neurosci. Lett. 362 (2) (2004)
[61] G. Terova, S. Rimoldi, G. Bernardini, R. Gornati, M. Saroglia, Sea bass ghrelin: 103–107.
molecular cloning and mRNA quantification during fasting and refeeding, Gen. [88] N.R. Vulliemoz, E. Xiao, L. Xia-Zhang, M. Germond, J. Rivier, M. Ferin, Decrease
Comp. Endocrinol. 155 (2) (2008) 341–351. in luteinizing hormone pulse frequency during a five-hour peripheral ghrelin
[62] N. Amole, S. Unniappan, Fasting induces preproghrelin mRNA expression in the infusion in the ovariectomized rhesus monkey, J. Clin. Endocrinol. Metab. 89 (11)
brain and gut of zebrafish, Danio rerio, Gen. Comp. Endocrinol. 161 (1) (2009) (2004) 5718–5723.
133–137. [89] J. Iqbal, Y. Kurose, B. Canny, I.J. Clarke, Effects of central infusion of ghrelin on
[63] T. Kono, Y. Kitao, K. Sonoda, R. Nomoto, T. Mekata, M. Sakai, Identification and food intake and plasma levels of growth hormone, luteinizing hormone, prolactin,
expression analysis of ghrelin gene in common carp Cyprinus carpio, Fish. Sci. 74 and cortisol secretion in sheep, Endocrinology 147 (1) (2006) 510–519.
(3) (2008) 603–612. [90] S. Unniappan, R.E. Peter, In vitro and in vivo effects of ghrelin on luteinizing
[64] H. Kaiya, B.C. Small, A.L. Bilodeau, B.S. Shepherd, M. Kojima, H. Hosoda, hormone and growth hormone release in goldfish, Am. J. Physiol. Regul. Integr.
K. Kangawa, Purification, cDNA cloning, and characterization of ghrelin in Comp. Physiol. 286 (6) (2004) R1093–R1101.
channel catfish, Ictalurus punctatus, Gen. Comp. Endocrinol. 143 (3) (2005) [91] M. Sokołowska-Mikołajczyk, M. Socha, P. Szczerbik, P. Epler, The effects of
201–210. ghrelin on the in vitro spontaneous and sGnRH-A stimulated luteinizing hormone
[65] K. Murashita, T. Kurokawa, T.O. Nilsen, I. Rønnestad, Ghrelin, cholecystokinin, (LH) release from the pituitary cells of common carp (Cyprinus carpio L.), Comp.
and peptide YY in Atlantic salmon (Salmo salar): molecular cloning and tissue Biochem. Physiol. Mol. Integr. Physiol. 153 (4) (2009) 386–390.
expression, Gen. Comp. Endocrinol. 160 (3) (2009) 223–235. [92] C.L. Grey, J.P. Chang, Nitric oxide signaling in ghrelin-induced LH release from
[66] K. Feng, G.R. Zhang, K.J. Wei, B.X. Xiong, Molecular cloning, tissue distribution, goldfish pituitary cells, Gen. Comp. Endocrinol. 183 (183) (2013) 7–13.
and ontogenetic expression of ghrelin and regulation of expression by fasting and [93] C.L. Grey, J.P. Chang, Differential involvement of protein kinase c and protein
refeeding in the grass carp (Ctenopharyngodon idellus), J Exp Zool A Ecol Genet kinase a in ghrelin-induced growth hormone and gonadotrophin release from
Physiol 319 (4) (2013) 202–212. goldfish (Carassius auratus) pituitary cells, J. Neuroendocrinol. 23 (12) (2011)
[67] H. Hosoda, M. Kojima, H. Matsuo, K. Kangawa, Ghrelin and des-acyl ghrelin: two 1273–1287.
major forms of rat ghrelin peptide in gastrointestinal tissue, Biochem. Biophys. [94] T. Ishikawa, H. Fujioka, T. Ishimura, A. Takenaka, M. Fujisawa, Ghrelin expression
Res. Commun. 279 (3) (2000) 909–913. in human testis and serum testosterone level, J. Androl. 28 (2) (2007) 320–324.
[68] G. Fernandez, A. Cabral, M.P. Cornejo, P.N. De Francesco, G. Garcia-Romero, [95] M.L. Barreiro, F. Gaytan, J.E. Caminos, L. Pinilla, F.F. Casanueva, E. Aguilar,
M. Reynaldo, M. Perello, Des-acyl ghrelin directly targets the arcuate nucleus in a C. Dieguez, M. Tena-Sempere, Cellular location and hormonal regulation of
ghrelin-receptor independent manner and impairs the orexigenic effect of ghrelin, ghrelin expression in rat Testis1, Biol. Reprod. 67 (6) (2002) 1768–1776.
J. Neuroendocrinol. 28 (2) (2016) 12349. [96] A.V. Sirotkin, R. Grossmann, M.T. María-Peon, J. Roa, M. Tena-Sempere, S. Klein,
[69] P.J. Delhanty, S.J. Neggers, A.J. van der Lely, Des-acyl ghrelin: a metabolically Novel expression and functional role of ghrelin in chicken ovary, Mol. Cell.
active peptide, Endocr. Dev. 25 (2013) 112–121. Endocrinol. 257–258 (2006) 15–25.
[70] P.J. Delhanty, S.J. Neggers, A.J.J.E.j.o.e. van der Lely, Mechanisms in [97] I.S. Parhar, H. Sato, Y. Sakuma, Ghrelin gene in cichlid fish is modulated by sex
endocrinology: ghrelin: the differences between acyl-and des-acyl ghrelin 167 (5) and development, Biochem. Biophys. Res. Commun. 305 (1) (2003) 169–175.
(2012) 601–608. [98] J. Upont, V. Maillard, S. Coyral-Castel, C. Rame, P. Froment, Ghrelin in female and
[71] A.M. Blanco, M. G  Alonso-G
omez-Boronat, A.L. omez, R. Yufa, S. Unniappan, male reproduction, International Journal of Peptides 2010 (2010) 158102.
M.J. Delgado, A.I. Valenciano, Characterization of ghrelin O-acyltransferase [99] J.I. Bertucci, A.M. Blanco, L.F. Canosa, S. Unniappan, Estradiol and testosterone
(GOAT) in goldfish (Carassius auratus), PloS One 12 (2) (2017), e0171874. modulate the tissue-specific expression of ghrelin, ghs-r, goat and nucb2 in
[72] S.-R. Chen, H. Chen, J.-J. Zhou, G. Pradhan, Y. Sun, H.-L. Pan, D.-P. Li, Ghrelin goldfish, Gen. Comp. Endocrinol. 228 (2016) 17–23.
receptors mediate ghrelin-induced excitation of agouti-related protein/ [100] K. Conde, T.A. Roepke, 17β-Estradiol increases arcuate KNDy neuronal sensitivity
neuropeptide Y but not pro-opiomelanocortin neurons, J. Neurochem. 142 (4) to ghrelin inhibition of the M-current in female mice, Neuroendocrinology 110
(2017) 512–520. (7–8) (2020) 582–594.
[73] S. Luquet, F.A. Perez, T.S. Hnasko, R.D. Palmiter, NPY/AgRP neurons are Essential [101] T.-M. Chouzouris, E. Dovolou, F. Krania, I.S. Pappas, K. Dafopoulos, I.E. Messinis,
for feeding in adult mice but can be ablated in neonates, Science 310 (5748) G. Anifandis, G.S. Amiridis, Effects of ghrelin on activation of Akt1 and ERK1/2
(2005) 683–685. pathways during in vitro maturation of bovine oocytes, Zygote 25 (2) (2017)
[74] C.T. Lim, B. Kola, M. Korbonits, The ghrelin/GOAT/GHS-R system and energy 183–189.
metabolism, Rev. Endocr. Metab. Disord. 12 (3) (2011) 173–186. [102] E. Dovolou, I.E. Messinis, E. Periquesta, K. Dafopoulos, A. Gutierrez-Adan,
[75] R. Barazzoni, A. Bosutti, M. Stebel, M.R. Cattin, E. Roder, L. Visintin, L. Cattin, G.S. Amiridis, Ghrelin accelerates in vitro maturation of bovine oocytes, Reprod.
G. Biolo, M. Zanetti, G. Guarnieri, Ghrelin regulates mitochondrial-lipid Domest. Anim. 49 (4) (2014) 665–672.
metabolism gene expression and tissue fat distribution in liver and skeletal [103] E. Shepperd, C. Peng, S. Unniappan, Ghrelinergic system in fish ovaries and
muscle, Am. J. Physiol. Endocrinol. Metabol. 288 (1) (2005) E228–E235. ghrelin inhibition of germinal vesicle breakdown in zebrafish oocytes, Gen. Comp.
Endocrinol. 176 (3) (2012) 426–431.

134
H. Zhong et al. Reproduction and Breeding 1 (2021) 128–135

[104] H. Mabudi, S. Jamili, N.E. Majd, G. Vosoughi, M.R. Fatemi, S. Rashed, The effects Mice with a Designed Ghrelin O-Acyltransferase Inhibitor, vol. 330, 2010,
of ghrelin on ovary histology in Barbus sharpeyi, J. Anim. Physiol. Anim. Nutr. 95 pp. 1689–1692, 6011.
(5) (2011) 599–602. [109] C.C. Zhu, H. Zhang, J.S. Zhang, Z. Li, J. Zhao, W. Li, Y.Q. Zhang, Inhibition of
[105] Y. Zohar, C.C. Mylonas, Endocrine manipulations of spawning in cultured fish: ghrelin signaling improves the reproductive phenotype of male ob/ob mouse,
from hormones to genes, in: C.-S. Lee, E.M. Donaldson (Eds.), Reproductive Fertil. Steril. 99 (3) (2013) 918–926.
Biotechnology in Finfish Aquaculture, Elsevier, Amsterdam, 2001, pp. 99–136. [110] M. Marchi o, L. Roli, C. Giordano, T. Trenti, A. Guerra, G. Biagini, Decreased
[106] K. Matsuda, T. Miura, H. Kaiya, K. Maruyama, S.-I. Shimakura, M. Uchiyama, ghrelin and des-acyl ghrelin plasma levels in patients affected by
K. Kangawa, S. Shioda, Regulation of food intake by acyl and des-acyl ghrelins in pharmacoresistant epilepsy and maintained on the ketogenic diet, Clin. Nutr. 38
the goldfish, Peptides 27 (9) (2006) 2321–2325. (2) (2019) 954–957.
[107] T. Kitazawa, B. De Smet, K. Verbeke, I. Depoortere, T.L. Peeters, Gastric motor [111] Y. Nishi, H. Hiejima, H. Hosoda, H. Kaiya, K. Mori, Y. Fukue, T. Yanase,
effects of peptide and non-peptide ghrelin agonists in mice in vivo and in vitro, H. Nawata, K. Kangawa, M. Kojima, Ingested medium-chain fatty acids are directly
Gut 54 (8) (2005) 1078–1084. utilized for the acyl modification of ghrelin, Endocrinology 146 (5) (2005)
[108] B.P. Barnett, Y. Hwang, M.S. Taylor, H. Kirchner, P.T. Pfluger, V. Bernard, Y.- 2255–2264.
y. Lin, E.M. Bowers, C. Mukherjee, W.-J. Song, P.A. Longo, D.J. Leahy, [112] F. Lemarie, E. Beauchamp, P. Legrand, V. Rioux, Revisiting the metabolism and
M.A. Hussain, M.H. Tsch€ op, J.D. Boeke, P.A. Cole, Glucose and Weight Control in physiological functions of caprylic acid (C8:0) with special focus on ghrelin
octanoylation, Biochimie 120 (2016) 40–48.

135

You might also like