You are on page 1of 15

Aquaculture and Fisheries 7 (2022) 525–539

Contents lists available at ScienceDirect

Aquaculture and Fisheries


journal homepage: www.keaipublishing.com/en/journals/aquaculture-and-fisheries

Review article

Sexual plasticity in bony fishes: Analyzing morphological to molecular


changes of sex reversal
Swathi Tenugu, Balasubramanian Senthilkumaran *
Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad, 500046, India

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

Keywords: Sex reversal is one of the characteristic properties of sexual plasticity in bony fishes wherein both natural and
Sex reversal induced sex change happens at various stages of life cycle in different species. Sex determination in gonochoristic
Sequential hermaphrodite species is genetically regulated, wherein the same sex is retained throughout their life span whereas hermaph­
Sex steroids
rodites change their sex during development or adulthood. In sequential hermaphrodites, serial sex change
Gonadal transdifferentiation
occurs at different points of life cycle. Concurrently, synchronous hermaphrodites function as both the sexes
Sexual dimorphism
Teleost during spawning. Other variables like temperature, pH and social factors can trigger sex reversal in teleost. Sex
reversal through gene mutations and chemicals/hormones, including sex steroids, can be induced mostly at early
developmental stages but natural sex reversal can occur at any time. Sex reversal mechanism shows morpho­
logical to molecular changes, which are ideal for identification of sex-specific gene markers. In fact, gonadal
transdifferentiation occurs at the molecular level through differential expression of transcription factors and
steroidogenic enzyme genes vis-a-vis hormones, thereby imparting phenotypic or structural changes. In addition,
brain shows sexual dimorphism which is mostly consequential to gonadal sex development and occasionally
either causative. The major breakthrough in this line is the identification of sex determining genes such as dmy/
dmrt1Yb, gsdfY, sox3 in the Japanese medaka and amhY in Patagonian pejerrey. Incidentally, the induction of
mono-sex population by favouring one sex due to sex-specific differences in growth is an important economic
boom for aquaculture. This review comprehensively highlights key molecular factors involved in natural and
induced sex reversal conditions to illustrate teleostean sexual plasticity and its application perspectives.

regulatory mechanisms leading to endocrine sex differentiation remain


1. Introduction conserved (Kobayashi, Nagahama, & Nakamura, 2013). In this process,
sex reversal becomes an intriguing phenomenon most commonly
One of the fascinating properties exhibited by bony fishes is the ex­ observed in certain groups of teleost. In all mammals, SRY (sex deter­
istence of various reproductive strategies in terms of sexuality. Sex mining region Y in all mammals) is the first sex determining gene
determination in teleost is a crucial event comprising a bipotential discovered that is sufficient for testicular development (Koopman,
gonad to develop either into an ovary or testis, but the underlying Gubbay, Vivian, Goodfellow, & Lovell-Badge, 1991; Koopman, Mün­
mechanisms vary differently among taxa (Devlin & Nagahama, 2002; sterberg, Capel, Vivian, & Lovell-Badge, 1990; Sinclair et al., 1990).
Hayes, 1998). Fishes exhibit gonochorism like most vertebrates, Incidentally, the sry-like genes could not be isolated in fish, yet fishes
whereby the sexual fate of an individual is majorly determined by ge­ express certain master genes which regulate the formation of gonads
netic, environmental and hormonal interactions (Volff, 2002). They that are gender-specific. Among non-mammalian vertebrates, dmy or
develop into either male or female and retain the same sex throughout dmrt1b which is a duplicate copy of doublesex and mab-3 related factor 1
the life. Unlike other vertebrates, few fishes are referred to as her­ (dmrt1) on the Y chromosome, was the first sex determining gene
maphrodites, wherein they function as both male and female during the identified in the Japanese medaka, Oryzias latipes and later in
same spawning period (Helfman, Collette, & Facey, 1997, p. 544). In O. curvinotus, which is associated with testicular differentiation (Mat­
contrast to this, sequential hermaphrodites were found to function as a suda et al., 2002, 2003; Nanda et al., 2002). But in other species of
male and a female at different stages of life (Warner, 1988). Despite of medaka, O. luzonensis gsdfY was the sex determining gene (Myosho et al.,
sexual plasticity and various reproductive patterns, the underlying 2012). In line with this, sex determining genes identified so far in fishes

* Corresponding author. Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad, 500046, India.
E-mail addresses: bsksl@uohyd.ac.in, senthilkumaranbalasubramanian@gmail.com (B. Senthilkumaran).

https://doi.org/10.1016/j.aaf.2022.02.007
Received 29 December 2021; Received in revised form 15 February 2022; Accepted 26 February 2022
Available online 17 March 2022
2468-550X/© 2022 Shanghai Ocean University. 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/).
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

List of abbreviations foxl2 forkhead box protein L2


FSH follicle stimulating hormone
11b-h 11b-hydroxylase Fshr follicle stimulating hormone receptor
11-KT 11-ketotestosterone Gdnf glial cell line-derived neurotrophic factor
5-HT serotonin gfrα-1 glial cell line-derived neurotrophic factor family receptor
5-HTP 5-hydroxy tryptophan GnRH gonadotropin-releasing hormone
Ad adrenosterone Gsdf gonadal soma-derived growth factor
ad4bp/sf-1 adrenal-4-binding protein/steroidogenic factor 1 GTH gonadotropin
AI Aromatase inhibitors hCG human chorionic gonadotropin
amh anti-Müllerian hormone HHG hypothalamo-hypophyseal-gonadal
amhr2 anti-Müllerian hormone receptor type 2 hsd11b2 hydroxysteroid 11β-dehydrogenase 2
amhy Y chromosome-linked anti-Müllerian hormone hsd3b hydroxy-delta-5-steroid dehydrogenase 3 beta
ar androgen receptor LH luteinizing hormone
CA catecholamine Lhr luteinizing hormone receptor
cAMP cyclic adenosine monophosphate Mas masculinization
cyp11a1 cytochrome P450 family 11 subfamily a polypeptide1 MT 17α-methyltestosterone
cyp19a aromatase NE norepinephrine
cyp19a1 cytochrome P450 family 19 subfamily a polypeptide 1 NPY Neuropeptide Y
DA dopamine PGC PGCs progenitor germ cells
DMY Y-specific DM-domain POA-H preoptic area-hypothalamus
DMY/dmrt1bY doublesex and mab-3-related transcription factor 1 ptx pentraxin
dpf days post fertilization sox SRY-box transcription factor
dph days post hatch sox9 SRY-box 9
E2 17β-estradiol T testosterone
EE2 ethynylestradiol th tyrosine hydroxylase
er estrogen receptor tph tryptophan hydroxylase
ET 17α-ethynyltestosterone

tend to vary from species to species, for example, amh-Y in the Pata­ determination/differentiation in most of the teleosts (Fernandino, Hat­
gonian pejerrey, Odontesthes bonariensis, amhr2 in the pufferfish, Taki­ tori, Moreno Acosta, Strüssmann, & Somoza, 2013). Additionally,
fugu rubripes, irf9y in the rainbow trout, Oncorhynchus mykiss and amh in certain endocrine disruptors affect sex determination/differentiation
the Nile tilapia, Oreochromis niloticus (Cáceres et al., 2019; Hattori et al., and reverse differently. This review will aim to highlight different as­
2012; Kamiya et al., 2012; Yano et al., 2012). But, the evolutionary pects including sex reversal induced by various steroid hormones,
conservation of several sex determining genes identified so far is still environmental factors and ultimate use of generating monosex popula­
unclear. These genes specifically regulate some downstream targets, tion through hormonal treatment followed by genetic cross as well as
which are highly conserved among different species. Some of the genes brain sex dimorphism exhibited by few teleost which were categorized.
involved in gonadal development include doublesex/mab3-related
transcription factors (dmrt), Wilms’ tumour suppressor gene (wt1), 2. Sexual development: Morphological changes during normal
anti-Müllerian hormone (amh), SRY-box 9 (sox9), aromatase (cyp19) and gonadal development including sex reversal
steroidogenic factor 1 (Ad4BP/sf-1) were identified in the autosomes of
several species. In most fishes, sex is determined through factors from Sexual development is a conserved process in most of the vertebrates,
somatic as well as sex chromosomes, yet the sexual fate is susceptible to which define particular events that specifically occur during gonadal
hormones as well as environmental factors leading to sex reversal. In a development by allowing the expression of genetic sex into its appro­
holistic view, overriding of genetic factors through hormone or envi­ priate phenotype (Devlin & Nagahama, 2002; Piferrer, 2001; Piferrer &
ronmental influence is likely in teleosts. Blázquez, 2005). The molecular mechanisms leading to females and
Sex reversal property was first demonstrated by the Japanese sci­ males exhibits wide diversity among vertebrates and the cascade of
entist, Yamamoto in medaka through sex reversing the genotypic female events during sex differentiation seems relatively conserved (Nagahama,
to male phenotype and genotypic male to female phenotype (Yama­ 2005). The sex steroids, mainly, androgens and estrogens play a very
moto, 1953; 1958). Later, it was postulated that the sex steroids act as important role in regulating sex differentiation process (Piferrer &
natural female and male sex inducers, which led to a framework in Guiguen, 2008). It commences with the migration of primordial germ
research to find the underlying mechanisms of sex change that have cells, followed by their colonization into genital ridge forming gonadal
been continued till date (Yamamoto, 1969). In addition, aromatase primordium and extends the first process of gametogenesis with the
blockers like fadrozole were often used for sex reversal in a few teleosts release of gametes upon eventful completion. Due to sexual plasticity,
(Afonso, Wassermann, & Terezinha de Oliveira, 2001; Nakamura, some fishes act as functional males and females during their life-time,
Kobayashi, Miura, Alam, & Bhandari, 2005; Zhou & Gui, 2010). Taken referred to as hermaphrodites compared to non-sex-changing species,
together, the sex determination and differentiation, as well as sexual i.e., gonochoristic which retain only one sex throughout their life. Two
plasticity in response to sex steroids and environmental factors, is a terms have been employed for the classification of gonochoristic species
complex process. The sex reversal phenomenon observed during in­ by Yamamoto (1969), referred to as (1) differentiated gonochorists
duction by steroid hormones is perceived only during the critical period where the undifferentiated gonad directly develop into an ovary or a
of gonadal differentiation. More specifically, sex steroids under natural testis during early gonadal development as observed in the European
and induced conditions mediate sex change either to male or female in seabass, Dicentrarchus labrax, the common carp, Cyprinus carpio and the
most of the teleost in a particular time frame. The environmental factors, coho salmon, Onchorynchus kisuch (Blázquez, Zanuy, Carrillo, & Piferrer,
including temperature, pH and social factors show an impact on sex 1998; Komen, De Boer, & Richter, 1992; Piferrer & Donaldson, 1989)

526
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

and (2) undifferentiated gonochorists, in which all the individuals first 3. Sex steroids causing sex reversal
develop gonads containing oocytes which is termed as ovarian differ­
entiation. Later, the oocytes undergo degeneration through apoptosis in Even though the sex determination in fishes is under genetic regu­
half of the population, followed by masculinization of the gonad pro­ lation, the external factors such as temperature, pH, social behavior
ceeding to form an intersexual gonad that eventually develops into induce sex reversal in most of the fishes, where the process of sex
normal testis (Maack & Segner, 2003). This phenomenon is termed as determination, as well as differentiation has already been well estab­
juvenile hermaphroditism and was reported in O. masou and Gambusia lished (Bushmann & Burns, 1994; Lowe & Larkin, 1975; Pandian &
affinis (Koya, Fujita, Niki, Ishihara, & Miyama, 2003; Nakamura, 1984). Koteeswaran, 1998; Strüssmann, Calsina Cota, Phonlor, Higuchi, &
But in a few species, juveniles possessing a bipotential intersex gonad Takashima, 1996). Additionally, in fishes where the sex differentiation is
was found to develop into either ovary or testis as observed in the not yet completed, manipulation of gonadal sex through the adminis­
serranid, Epinephelus striatus, the Japanese eel, Anguilla japonica and the tration of sex steroids via diet, immersion, injection or through im­
European eel, A. anguilla (Beullens et al., 1997; Colombo & Grandi, plantation technique is possible (Devlin & Nagahama, 2002; Frisch,
1995; Sadovy & Colin, 1995). Hermaphroditism is a naturally occurring 2004; Nagahama, 2002; Pandian & Sheela, 1995). Sex reversal through
phenomenon observed in most of the species and categorized as simul­ endocrine mediated techniques is well applied for the production of
taneous or sequential (Shapiro, 1990; Yamamoto, 1969). In simulta­ monosex populations. Steroid hormones are widely used to control and
neous species, the individuals possess both male and female gonadal manipulate the sex of the fish (Baroiller, Guiguen, & Fostier, 1999; Borg,
tissues where both function together. Sequential hermaphrodites are 1994; Lee et al., 2000; Nakamura et al., 1989). This practice was mostly
categorized into protandrous, which initially mature as males and pro­ used in teleosts, among which tilapia and salmonids have received
togynic that matured as females (Ross, 1990). In some protogynous utmost attention. Sex reversal has been limited to only few species such
species such as the bluehead wrasse, Thalassoma bifasciatum, few in­ as the channel catfish, Ictalurus punctatus and the African catfish, Clarias
dividuals differentiate directly into males whereas others first pass gariepinus by giving steroid hormone through dietary supplementation
through the condition of being functionally females (Munday, Buston, & or exposure in water (Galvez, Mazik, Phelps, & Mulvaney, 1995; Goudie,
Warner, 2006; Munday, Wilson White, & Warner, 2006). These species Redner, Simco, & Davis, 1983; Liu, Yao, & Wang, 1996; Raghuveer &
referred to as diandric protogynous which is a rare property observed in Senthilkumaran, 2009). In order to advance the maturation in the cat­
protoandric species (Devlin & Nagahama, 2002). In addition, certain fish, C. gariepinus, human chorionic gonadotropin (hCG) induction was
species like the rubble gobiid fish, Trimma okinawae have the ability to performed by implantation of osmotic pump during off breeding season
undergo serial sex change based on the social colonization behavior. and expression of several steroidogenic enzymes, transcription factors
This has been well demonstrated wherein serial sex change of dominant and hormonal levels were measured (Murugananthkumar, Prathibha,
female to male and vice-versa occurred after the introduction of the Senthilkumaran, Rajakumar, & Kagawa, 2017). However, C. batrachus
dominant male in an artificial culture condition (Sunobe & Nakazono, and C. gariepinus show distinct difference because of the seasonal pattern
1993). These serial sex changing fish have differential expression variance exhibited and hence, these species seem to be the best model to
pattern of gonad-specific genes. This was the first known teleost where study any kind of a seasonal influence or endocrine manipulation
bi-directional sex change was observed which became advantageous (Rajendiran et al., 2021; Senthilkumaran & Kar, 2021). In some studies
wherein a male was forced to become subsidiary, ensuing to take over of the Asian stinging catfish, Heteropneustes fossilis when eggs were
the social unit by a larger male. Other species such as the saddleback treated with 17α-methyltestosterone (MT) and 17α-ethynyltestosterone
wrasse, T. duperrey also undergo protogynous sex change where the (ET) it produced males and upon treatment with 17β-estradiol (E2) and
individuals initially mature as females or males. Later, under social diethylstilbestrol it produced females (Haniffa, Sridhar, & Nagarajan,
conditions initial phase (IP) female or male fish become terminal phase 2004). This sex reversal was amenable as they show sex change at the
(TP) males. But in this species, females housed with IP males undergo time of hatching. This feminization is mainly due to the conversion of
sex change to become TP males which have high circulating levels of androgens to estrogens by the cyp19a enzyme. Similarly, this feminizing
11-KT and spawn individiually with large female successfully (Morrey, effect was also found in Salmo gairdneri and occurred only at higher
Nagahama, & Grau, 2002). Conversely, females housed with TP males, doses of MT treatment (Solar, Donaldson, & Hunter, 1984).
inhibits sex change but helps in the maintenance of ovarian function and Sex-change is majorly triggered by several steroids such as E2, 11-
likelihood of successful pair spawning with immediate proximity fe­ ketotestosterone (11-KT) and Testosterone (T). E2 is one of the most
males. Therefore, under social conditions females change their sex along dominant estrogen amongst protandric species that is associated with
with gonadal phenotype compared to males which are only restricted in sex change in most of the teleosts (Kime, 1993). E2 is a predominant
regulating sex change and maintain ovarian function in females feminizing sex steroid that can induce permanent sex transition in some
(Hourigan, Nakamura, Nagahama, Yamauchi, & Grau, 1991). In such teleosts (Chang & Lin, 1998; Lee et al., 2000) and also increase E2
species, during sex change, the ovaries which possess undetectable production concurrently through progression of natural sex reversal in
testicular tissue in females were fully reorganized to a functional testis several species (Godwin & Thomas, 1993; Guiguen, Jalabert, Benett, &
(Nakamura, Hourigan, Yamauchi, Nagahama, & Grau, 1989). But in the Fostier, 1995; Nakamura et al., 1989). In contrast, 11-KT is the pre­
yellowtail clownfish, Amphiprion clarkii, the dominance of the sex dominant sex steroid which is related to sex change to masculinization
change depends on the size. For example, normally female is a dominant in most of the protogynous species, whereby permanent sex transition
and male is the second-largest member in that population based on size. was observed upon treatment of the correlate in many species (Cardwell
In the absence or death of the female, sex change of male to female & Liley, 1991; Grober, Jackson, & Bass, 1991; Higa, Ogasawara, Saka­
occurs, thus, ultimately leading to female dominance (Fricke & Fricke, guchi, Nagahama, & Nakamura, 2003; Kroon & Liley, 2000). Similar to
1977). The sex steroid hormones such as estrogens and androgens E2, 11-KT levels also showed a progressive increase in synthesis during
induced sex change in several teleost which are categorized as key natural sex change to masculinization in many species (Bhandari,
regulators (Godwin & Thomas, 1993). Thus, due to this sexual plasticity, Komuro, Nakamura, Higa, & Nakamura, 2003; Nakamura et al., 1989).
the underlying mechanisms involved during sex change make it more 11-KT male specific and E2 female specific were found to be the domi­
prominent to identify genes contributing to sex differ­ nant products in the incubates of the gonadal tissues that undergo
entiation/determination. In the first instance, sex steroids seem to play transition (Borg, 1994; Lee et al., 1995; Yeung, Chen, & Chan, 1993).
an essential role in evoking sex reversal. The function of 11-KT in protogynous species is antithetical to that of E2
in protandric species facilitating as switches between female and male
phenotypes (Hunter & Donaldson, 1983; Pandian & Sheela, 1995;
Piferrer, 2001; Piferrer, Baker, & Donaldson, 1993). In transitional

527
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

A. melanopus, no apparent E2 levels were evident until after oogonial oocyte development (Sun, Zha, Spear, & Wang, 2007). Ovarian trans­
proliferation indicating E2 is not required for initiating sex change in this differentiation was evident in female zebrafish upon AI treatment
species (Godwin & Thomas, 1993). However, in some other species very wherein first testis-like tissues containing spermatozoa-like cells devel­
low or undetectable levels of 11-KT were observed during transitional oped, which was followed by ovary retraction (Takatsu et al., 2013). Sun
sexual phases that led to a point whether 11-KT involved in sex change et al. (2014) observed the differentiated ovary to undergo trans­
or else mediated through different combination of steroids (Kime, 1993; differentiation and become functional testis upon long-term AI treat­
Kroon, Munday, & Pankhurst, 2003; Yeung & Chan, 1987). In several ment causing functional sex reversal in tilapia. It was noteworthy that
studies of hermaphrodite species, similar to 11-KT, T also play a role the first morphological change of ovarian structure in this species was
during sex change by maintaining its levels independent to the sexual observed at 20 dph (Kobayashi et al., 2008). In medaka, estrogen
stage, indicating T not directly involved during transitions (Cardwell & treatment in XY males resulted in ovarian development while XX fe­
Liley, 1991; Guiguen, Jalabert, Thouard, & Fostier, 1993; Nakamura males upon treatment with androgens induced testis development,
et al., 1989). Interestingly, administration T, MT and testosterone pro­ causing sex reversal (Yamamoto, 1953). Incidentally, fertilized XY eggs
pionate induced sex change in few protogynous species (Glamuzina, of medaka when immersed in water containing exogenous E2 induced
Glavić, Skaramuca, & Kožul, 1998; Hassin et al., 1997; Kuo, Ting, & Yeh, sex reversal of few genotypic males to functional females (Iwamatsu,
1988; Lee et al., 1995; Tan-Fermin, Garcia, & Castillo, 1994; Yeh, Kuo, Kobayashi, Hamaguchi, Sagegami, & Shuo, 2005). However, neither
Ting, & Chang, 2003), whereas in estuary grouper small doses of T has dmy nor the germ cells number was affected during the early gonadal
stimulated ovarian development. In contrast, T or 5α-dihydrotestoster­ development indicating that the Japanese medaka seems not succumb­
one in T. bifasciatum induced ovarian degeneration (Kramer, Koulish, & ing to external E2 treatment during early gonadal sex differentiation
Bertacchi, 1988), suggesting that sex change is not influenced by steroid (Scholz et al., 2003; Suzuki, Nakamoto, Kato, & Shibata, 2005). The
directly but indirectly affects the precursor’s availability for biosyn­ killifish, Kryptolebias marmoratus capable of undergoing self-fertilization
thesis. However, in most of the species, T shows ubiquitous presence showed skewness to male sex development of embryos after treatment
irrespective of the female or male phase. Thus the physiological effect with MT (Kanamori et al., 2006).
upon T administration majorly depends on enzymes, for example,
cyp19a or 11bh which are dominant during the time of treatment. The 4. Factors affecting sex reversal
distinct conversion of the substrates of T into androgens and estrogens
was corroborated in few studies when incubated with testicular and 4.1. Temperature
ovarian tissues (Lee et al., 1995; Yeung et al., 1993). Incidentally, in
C. gariepinus MT treatment did not induce complete sex reversal in entire Temperature-induced sex reversal is one of the properties observed
population which is evident from the occurrence of intersex. On the in several teleost by triggering various candidate genes associated with
other hand, in the same species, E2 treatment induced almost complete it. The first observation of temperature effect on sex reversal was found
sex reversal (Raghuveer & Senthilkumaran, 2009). in the Atlantic silverside, Menidia menidia, wherein warm fluctuating
Apart from classical steroids, non-classical steroids such as adre­ temperature (17–25 ◦ C) treatment during larval development resulted in
nosterone (Ad) are known to induce sex change in certain species such as male population (Conover & Kynard, 1981). Among the poeciliids and
black-eye goby, Coryphopterus nicholsii, but this particular effect is more cichlids families, the blackbelly limia, Poecilia melanogaster and um­
likely due to the presence of 11-KT precursor, as receptors for Ad does brella cichlid, Apistogramma borelli showed a significant effect when
not exist (Kroon & Liley, 2000). Aromatase inhibitors (AI) such as larvae were incubated at a temperature range from 23 to 29 ◦ C, which
fadrozole and exemestane suppress the sex change observed during produced male broodstocks (Römer & Beisenherz, 1996). In medaka,
natural sex reversal in protandric black porgy, Acanthopagrus schlegeli, when embryos were exposed to the high water temperature of 34 ◦ C,
whereas induced sex change in other species such as protogynous female to male sex reversal was evident, and this simple methodology
black-eye goby, C. nicholsii, honeycomb grouper, E. merra, threespot was used to produce XX males (Kobayashi, 2013; Sato, Endo, Yamahira,
wrasse, Halichoeres trimaculatus (Bhandari, Higa, Nakamura, & Naka­ Hamaguchi, & Sakaizumi, 2005). The genotypic females (XX) of Hd-rR
mura, 2004; Kroon & Liley, 2000) indicating E2 dominance. Addition­ strain in medaka exhibited sex reversal into male phenotype upon
ally, E2 when co-administered with 11-KT or AI, also suppressed exposure of fertilized eggs at embryonic stages 5–6 to the temperature
sex-change in H. trimaculatus (Higa et al., 2003). Upon oral adminis­ between 17 and 34 ◦ C until hatching, and most of them showed 100%
tration of MT in carp underwent sex reversal from female to male which sex reversal at 34 ◦ C (Hattori et al., 2007). But this thermal manipulation
was evaluated by using all female gynogenetic carp during sex differ­ for inducing sex reversal was ineffective at stage 36 which is at middle
entiation (Komen, Lodder, Huskens, Richter, & Huisman, 1989; Nagy, embryogenesis stage. But in another study, when Hd-rR and green
Bercsényi, & Csányi, 1981). In another study, masculinization (mas) fluorescent protein transgenic medaka were exposed to 32 ◦ C at em­
mutant which is an autosomal sex-determining gene involved in testes bryonic stage 25 caused sex reversal suggesting that the process of
differentiation, induced sex reversal of male development in XX females gonadal sex differentiation induced by high temperature is limited only
(Komen et al., 1992). In tilapia, MT administration induced masculini­ before the occurrence of morphological sex differentiation stage (Selim,
zation in XX larvae and treatment with AIs caused sex reversal of Shinomiya, Otake, Hamaguchi, & Sakaizumi, 2009). In addition to this,
functional males (Afonso et al., 2001; Kobayashi, Kajiura-Kobayashi, high temperature induced expression of dmrt1 in females after stage 36
Guan, & Nagahama, 2008; Kwon, McAndrew, & Penman, 2002; Naka­ during embryonic development (Hattori et al., 2007), whereas in normal
mura, Kobayashi, Chang, & Nagahama, 1998; Ruksana, Pandit, & males, it is expressed in XY gonads after 10 to 20 dph (Kobayashi et al.,
Nakamura, 2010). However, estrogen treatment did not induce XY sex 2004). Elevated temperature treatment of 32–34 ◦ C in tilapia, produced
reversal in tilapia as easily as androgen treatment in XX sex reversal an increased number of males in XX progeny whereas a decreased
along with AIs whereas 17α-ethynylestradiol treatment from 4 to 6 days number of females in XY and YY progenies (Abucay, Mair, Skibinski, &
post hatch (dph) resulted in complete sex reversal of XY with germ cells Beardmore, 1999; Baroiller, Chourrout, Fostier, & Jalabert, 1995; Kwon
proliferation similar to that of XX gonads (Kobayashi et al., 2008; et al., 2002). The thermosensitivity period in this species ranged from 10
Kobayashi, Kajiura-Kobayashi, & Nagahama, 2003). Similarly, mascu­ to 20 days post fertilization (dpf) or 40 dpf (Baroiller, D’Cotta, & Sail­
linization effect is caused by AI was also observed in the Japanese lant, 2009). Decreased expression of cyp19a1 and foxl2 with elevated
flounder, Paralichthys olivaceus, the Chinook salmon, O. tshawytscha and expression of dmrt1 was evident in XX larvae when exposed to a high
the rainbow trout, O. mykiss (Guiguen et al., 1999; Kitano, Takamune, masculinizing temperature in tilapia (Baroiller, D’Cotta, & Saillant,
Nagahama, & Abe, 2000; Piferrer et al., 1994). 2009; d’Cotta et al., 2001; Poonlaphdecha et al., 2013). Interestingly,
In female breeding medaka, exposure of AI, the letrozole, affected the dmrt1 suppresses the cyp19a1a transcription by repressing the

528
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

activity of Ad4BP/sf1 dependent female pathway during sex reversal stages of development. For example, a male-biased sex ratio was
upon MT treatment in tilapia (Wang et al., 2010). Similarly, decreased observed in zebrafish when reared under hypoxic conditions compared
cyp19a1a expression was observed upon high masculinizing tempera­ to normoxic (Shang & Wu, 2004; Shang, Yu, & Wu, 2006). This is due to
ture exposure in the European sea bass, the Japanese flounder, the the modulation of the expression of steroidogenic enzyme genes, which
Japanese medaka and zebrafish (Kitano, Hayashi, Shiraishi, & Kamei, alters T and E2 balance (Pollock, Dubé, & Schryer, 2010; Wu, Zhou,
2012; Kitano, Takamune, Kobayashi, Nagahama, & Abe, 1999; Nav­ Randall, Woo, & Lam, 2003; Yu et al., 2012). It was reported that the
arro-Martín et al., 2011; Uchida, Yamashita, Kitano, & Iguchi, 2004). African cichlid Egyptian mouth-brooder, Pseudocrenilabrus multicolor
But the underlying molecular mechanism relating temperature with victoriae exhibited higher T levels when exposed to hypoxic conditions
cyp19a1a expression is yet to be deciphered. One of the mechanisms compared to normal oxygenated fish however, E2 levels were not
linking the temperature in sex ratio shift is the epigenetic modification affected (Friesen, Aubin-Horth, & Chapman, 2012). An increase in
observed in the study using the European sea bass model in which of cortisol followed by hypoxic conditions was evident during the early
increased methylation of cyp19a1a promoter in females by temperature stages of development in rainbow trout, suggesting that all are interre­
decreased its expression in masculinized fish during sex reversal (Nav­ lated and majorly active during this particular period (Fuzzen,
arro-Martín et al., 2011). This is due to the prevention of binding of Alderman, Bristow, & Bernier, 2011). In the Atlantic croaker, hypoxia
Ad4BP/sf-1 and foxl2 to the promoter sites and in turn blocking decreased tryptophan hydroxylase (TPH), cyp19a1b expression in the
cyp19a1a transcriptional activation (Wang et al., 2007). The similar hypothalamus. In vivo treatment with AI, 1,4,6-androstatrien-3,17-dione
epigenetic modification was also observed during temperature mediated also decreased TPHs, 5-hydroxy tryptophan (5-HTP), serotonin (5-HT)
sex reversal in zebrafish by methylation and decreased expression of and TPH activity (Rahman & Thomas, 2013). In the same study, E2
cyp19a1a, esr1 and sox9b as well as enrichment of cAMP-responsive treatment partially restored TPH activity, 5-HTP, 5-HT, TPHs contents in
element-binding proteins were observed (Han et al., 2021). In the Jap­ hypoxia-exposed fish, indicating the role of estrogens in controlling
anese flounder, temperature exposure 36 ◦ C induced sex reversal from neuronal responses to hypoxic conditions in aquatic vertebrates (Rah­
XX female to XX males with increased gsdf expression and inhibition of man & Thomas, 2013). Combination of environmental stress of hypoxia
germ cell proliferation leading to testis development (Yang et al., 2019). and xenobiotic exposure of polychlorinated biphenyl mixture (Aroclor
The African catfish, C. gariepinus, upon 36 ◦ C exposure, showed a 1254) in the Atlantic croaker decreased TPH activity that accompanied
masculinization effect when treated for at least 3days between 0 and with fall of 5-HT in hypothalamus and gonadotropin-releasing hormone
23dph and often skewed towards 100% male phenotypes when applied (GnRH1) content in preoptic area-hypothalamus (POA-H) region (Khan
from 6-8dph (Santi et al., 2016). In contrast, full masculinization was & Thomas, 2000). This was further associated with a reduction in LH
observed in common carp when larvae were exposed to 36 ◦ C, partic­ secretion and gonadal development which has been well-reviewed
ularly between 7 and 13 dph (Biswas et al., 2021). (Khan & Thomas, 2001; Rahman, Khan, & Thomas, 2011). 17, 20β,
The elevated temperature leads to stress and increased the level of 21-trihydroxy-4-pregnen-3-one, a progestin hormone that induce
cortisol during sex reversal has also received the most attention on gamete maturation, were significantly decreased under hypoxic condi­
reproduction (Consten et al., 2002; Mommsen, Vijayan, & Moon, 1999; tions in both sexes of the Atlantic croaker leading to impairment in
Schreck, 2010). The plasma levels of cortisol and androgens were gamete maturation (Thomas & Rahman, 2009). Hypoxia is also associ­
observed in few species during the sex determination and differentiation ated with endocrine dysfunction in this species by decreasing endocrine
period. For example, the cortisol levels increased in the pejerrey when indicators of the estrogen signalling pathway, that regulate vitellogenin
larvae reared in warm and male-inducing temperatures compared to production which is a precursor for growing oocytes leading to a
that of low temperature (Hattori et al., 2009). Additionally, when reproductive failure (Thomas, Rahman, Kummer, & Lawson, 2006).
treated with cortisol and dexamethasone, agonist, male-biased sex ratios
were observed when reared under intermediate temperatures. Interest­ 4.3. pH
ingly, very high levels of 11-KT and T were evident (Hattori et al., 2009).
The three possibilities of inducing masculinization may include (i) Other environmental factor affecting on sex ratio include pH, mostly
cortisol inhibiting cyp19a1a expression, (ii) cortisol reducing the num­ studied in poeciliids and chiclids species. Even though the effect of pH is
ber of progenitor germ cells (PGC) and, (iii) cortisol affecting 11-KT less prominent, but in common, acidic or low pH 5–6 exposure Pelvi­
synthesis. The first possibility was evident in the Japanese flounder cachromis subocellatus, P. pulcher, P. taeniatus, A. borelli, A. caucatoides,
and pejerrey where cortisol induced masculinization and cyp19a1a and Xiphophorus helleri skewed towards the appearance of males while a
downregulation were seen (Hattori et al., 2009; Yamaguchi, Yoshinaga, high pH value of 7 skewed towards the occurrence of females (Rubin,
Yazawa, Gen, & Kitano, 2010). The depletion of number of PGCs and 1985; Sullivan & Schultz, 1986). The sex ratio of 1:1 of males and fe­
their proliferation during masculinization was observed in medaka males was found when P. pulcher was exposed to intermediate pH (6.10)
(Tanaka, Saito, Morinaga, & Kurokawa, 2008). Increased incidence of (Rubin, 1985). In contrast, the other species, Poecilia sphenops higher
temperature dependent apoptosis in gonadal primordia during testicular proportion of females was evident when combined with temperature
differentiation was documented in zebrafish, pejerrey and medaka (Ito, (30 ◦ C) and pH 6 or 7 exposure. At the same time male ratio increased
Takahashi, Yamashita, & Strüssmann, 2008; Saito et al., 2007; Saito & when exposed to 22 and 26 ◦ C with pH 8 depicting opposite gender-bias
Tanaka, 2009; Uchida, Yamashita, Kitano, & Iguchi, 2002). An increase effect (Benjamín Barón, Fernando Bückle, & Espina, 2002). Exposure of
in the levels of androgens was also reported in several teleosts with various environmental stress factors and different hormonal treatment
masculinizing effect (Devlin & Nagahama, 2002). This ultimately in­ and expression of genes in depicting gonadal fate in various teleosts are
dicates the inhibition of cyp19a1a expression followed by apoptosis illustrated in Table 1.
might be a consequence for an increase in the production of androgens
to reinsure that cortisol can trigger masculinization in teleost upon high 4.4. Social interactions
temperature exposure.
Population density is an important social factor effecting sex deter­
4.2. Hypoxia mination in few teleosts. This appeared to be more prominent for sex
determination in Anguilliformes (Beullens et al., 1997; Davey & Jelly­
Hypoxia is a condition of the presence of low levels of dissolved man, 2005; Degani & Kushnirov, 1992; Holmgren, 1996; Krueger &
oxygen concentration in water and one of the factors affecting sex dif­ Oliveira, 1999; Tzchori, Degani, Hurvitz, & Moav, 2004). For example,
ferentiation in fish. Hypoxia is more likely perceived as a stressor by in the American eel, A. rostrata, low population density was interrelated
fishes as with other environmental factors, specifically during the early with a high number of females, whereas male-biased sex ratio was

529
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

Table 1
Exposure of environmental stress factors and different treatment in teleost depicting gonadal fate. A downward arrow pointing indicates decrease in hormone con­
centration, gene expression, enzymatic activity, an upward arrow indicates increase in these parameters, equal symbol depicts normal expression. “NS”: not studied.
Species Environmental Response on Steroids levels/ Physiological Response/ References
Stress factor/treatment transcription factors/steroidogenic Event
enzyme genes related to gonad

Anguilla anguilla Low density high ↑ cyp19a1a Feminization Tzchori et al. (2004)
density, cortisol ↑ cortisol, ↑11-KT Male-skewed population
treatment
Apistogramma sp T, acidic pH NS Male induced population Römer and Beisenherz (1996)
Clarias gariepinus MT = dmrt1, sox9a, foxl2, cyp19a1(Ova- Male-skewed population, Raghuveer and Senthilkumaran (2009)
testis); Intersex (Ova-testis)
= dmrt1 and sox9a (testes); = foxl2
and cyp19a1 (ovary)
Cyprinus carpio MDHT ↑ sox9a/b, ↑ sox30, ↑lhr Monosex induction Anitha & Senthilkumaran, 2020; Anitha, &
E2 ↓sox9a/b, ↓sox30, ↑ sox19, Senthilkumaran, 2022; Gupta & Senthilkumaran,
↑ cyp19a1a, ↑ fshr, ↑ ptx 2020
Danio rerio T (high) ↓ cyp19a1a Female to male sex reversal, Abozaid, Wessels, & Hörstgen-Schwark, 2012;
Hypoxia ↑ T/E2 ratio, ↑ cyp19a1a, apoptosis of PGCs Uchida et al., 2002, 2004
↑ cyp19a1b, ↑cyp17, ↑3β-hsd, Male-skewed population, Baroiller, D’Cotta, & Saillant, 2009; Shang et al.,
↑ cyp11a apoptosis induction of PGCs 2006; Yamamoto, 1969
Dicentrarchus labrax T (high) ↓ cyp19a1a Masculinization. cyp19a1a Blazquez, Navarro-Martín, & Piferrer, 2009;
promoter hypermethylation Navarro-Martín et al., 2011; Pavlidis et al., 2000
Odontesthes bonariensis T (high), cortisol ↑ cortisol,↑11-KT, ↑ T, Male-skewed population, Fernandino et al., 2011; Blázquez & Somoza, 2010;
treatment ↑ amh,↓ cyp19a1a, ↑ dmrt1, apoptosis of germ cells Strüssmann, Kitahara, & Amashita, 2008; Hattori
↑ hsd11b2, ↑ fshr et al., 2009; Shinoda et al., 2010
Oreochromis niloticus T (high) ↓ cyp19a1a Induced masculinization Baroiller et al. (1995)
Oryzias latipes T (high), cortisol ↑ cortisol, ↑ gsdf, ↓ fshr, Inhibition of PGCs Hattori et al., 2007; Hayashi et al., 2010; Selim et al.,
treatment ↓ cyp19a1a proliferation 2009
E2 + cortisol ↓ cortisol, ↑ cyp19a1a, ↓ gsdf, Rescued masculinization Kitano et al. (2012)
E2 + T (high) ↓ cortisol, ↑ fshr and cortisol levels
Metyrapone (cortisol Inhibition of
synthesis inhibitor) masculinization
Paralichthys olivaceus T (high) ↓ foxl2, ↓ cyp19a1a, ↑ cyp26b1 Delayed meiotic initiation in Kitano et al., 1999; Yamaguchi & Kitano, 2012;
Cortisol treatment ↑ cortisol, ↓ foxl2, ↓ cyp19a1a germ cells Yamaguchi et al., 2010
Male induced population
Pseudocrenilabrus Hypoxia ↑ T/E2 ratio Male-skewed population, Friesen et al. (2012)
multicolor victoriae apoptosis of germ cells

induced by high population density in A. japonica (Davey & Jellyman, differentiating males (Vizziano-Cantonnet et al., 2011). Whereas in
2005; Krueger & Oliveira, 1999). Social factors such as size also affects tilapia, upon exposure to 35 ◦ C, the expression of cyp11a1b was
the sex differentiation in Cichlasoma citrinellum whereby larger fishes decreased in genetic females and still lower in genetic males than those
during the juvenile stage mature into males (Francis & Barlow, 1993). found in masculinized females (D’cotta, Fostier, Guiguen, Govoroun, &
The interaction between growth rate as well as density are essential as Baroiller, 2001). In another study on tilapia, XY and YY sex reversal into
both are associated with sex determination (Davey & Jellyman, 2005). females resulted in increased expression of cyp19a1b which was
Representation of outcome of sequencial hermaphrodites, environ­ dependent on ethynylestradiol (EE2) dosage along with increased E2 and
mental factors steroid hormones, as well as monosexpopluation per­ T levels (Gennotte, Mélard, D’Cotta, Baroiller, & Rougeot, 2014).
formed in various species were depicted in Fig. 1. Increased cyp19a1b expression was evident in masculinized XX females
of tilapia along with high estrogen receptor-α (erα) and erβ expressions
5. Brain sex dimorphism upon temperature (27 ◦ C) treatment. Increased expression of cyp19a1b,
erα and erβ was observed in tilapia upon MT treatment in 10 and 20 dph
Fish retain large plasticity on sex reversal that was evident in the (Tsai, Wang, & Fang et al., 2001b). In contrast to this, increased
brain and also exists this reversibility even during adulthood (Godwin, expression of cyp19a1b and erα was evident upon E2 treatment between
2010). Brain sex differentiation in mammals and birds usually occurs 0 and 10 dph (Tsai, Wang, & Fang, 2001). Additionally, high tempera­
during embryonic development which are controlled by gonadal ste­ ture induced testicular differentiation after 10 dph in tilapia whereas
roids as well as genes regulating sex chromosomes, and epigenetic low temperature before 10 dph resulted in ovarian differentiation and
modifications (McCarthy & Arnold, 2011). In teleosts, brain sex within this thermosensitive period both cyp19a1b and erα were down­
dimorphism follows gonadal sex and this may either be a consequential regulated relating brain feminization (Tsai, Chang, Wang, & Chao,
or causative phenomenon. Nevertheless, sexually dimorphic genes sexes 2003). In medaka brain, esr1 mediates male-biased expression, sug­
at the level of brain serve as a gene marker to understand sex reversal. In gesting its role in masculinization whereas esr2β and arβ are associated
non-sex reversal fish like goldfish, gender specific sexual behavior can with feminization (Hiraki et al., 2012). The same study depicted the
be induced through hormonal treatment illustrating the dominance of expression of female-specific estrogen and androgen receptors that are
gonadal sex over brain sex (Kobayashi, Saoshiro, Kawaguchi, Hay­ activated and inhibited by estrogens and androgens, respectively. In the
akawa, & Munakata, 2013). pejerrey, cyp19a1b expression was evident in the brain at male pro­
ducing temperature before the sex differentiation onset (Strobl-Mazzulla
5.1. Brain cyp19a1b and estrogen receptors et al., 2008). Sex reversal was not evident when adult females were
treated with AI in the African cichlid Astatotilapia burtoni, however,
The brain cyp19a1b gene is involved as the feedback loop in regu­ partial masculinization was noticed in steroid level changes, body shape,
lating the steroid production by kisspeptin and hypothal­ appearance, colour, alterations in brain gene expressions that affected
amo–hypophyseal–gonadal (HHG) axis (Hofmann, 2006). The rainbow sexual behavior indicating estrogenic influence in juveniles (Göppert
trout depicted elevated cyp19a1b expression in the brain of et al., 2016).

530
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

Fig. 1. Schematic representation illustrating outcome of sequencial hermaphrodites, steroid hormones, environmental factors as well as monosexpopluation. Note:
References for these with respect to species has been covered in the bibliography of review.

5.2. GnRH why are hormones released on the first place? sex reversal, indicating independence of these two GnRHs in this
mechanism (Breton, DiMaggio, Sower, & Berlinsky, 2015). It is impor­
Hormones released from the HHG axis regulate various reproductive tant to understand the monoaminergic axis and its crucial role in regu­
events which lead to fertility by regulating steroidogenesis through lating the HHG axis to analyse sexual dimorphic GnRH localization, in
feedback at the level of the pituitary and brain. GnRH is known to be a turn GnRH-GTH axis (Goos, Senthilkumaran, & Joy, 1999; Peter, Tru­
hypothalamic decapeptide neurohormone majorly involved in repro­ deau, & Sloley, 1991). In teleosts, catecholamines (CA) and 5-HT are the
duction by stimulating the release of gonadotropins, follicle stimulating monoamines that are the major components of central nervous system
hormone (FSH) and luteinizing hormone (LH) by the pituitary gland, regulating various physiological functions, including reproduction.
that in turn regulates steroidogenesis and gametogenesis (Ando, Hew, & The photic signals known to affect the HHG axis primarily through
Urano, 2001; Ando & Urano, 2005; Gomes, Costa, & Borella, 2013; Kah retina and pineal organ in fishes such as the African cichlid, A. burtoni
et al., 2007; Maruska & Fernald, 2011; Yaron et al., 2003; Zohar, and major carp, Catla catla (Grens, Greenwood, & Fernald, 2005;
Munoz-Cueto, Elizur, & Kah, 2010). GnRH exists in multiple forms Moniruzzaman & Maitra, 2012). The two GnRH receptors, type-I and
which has resulted through genome duplication (Powell et al., 1994). type-II are abundantly expressed in amacrine and ganglion cells
Species specific and other forms of GnRH localized in various brain re­ respectively. The visual information is processed by amacrine cells and
gions were found to play a role in reproduction, neuromodulation and the ganglion cells transmit this processed information to the brain, a
during spawning behavior/migration (Senthilkumaran, Okuzawa, Gen, neuromodulator in order to influence GnRH in this process (Grens et al.,
Ookura, & Kagawa, 1999). The Nile tilapia possessing distinguish XY 2005; Robison et al., 2001). Exposure of major carp, C. catla to constant
and XX populations showed sexually dimorphic localization of seabream darkness and exogenous melatonin treatment resulted in testicular
GnRH (Swapna et al., 2008). It was documented that three distinct forms maturation during preparatory phase of reproductive cycle (Bhatta­
of GnRH in South American cichlid fish were distributed differentially charya, Chattoraj, & Maitra, 2007). It has been reported in the channel
during sex differentiation (Pandolfi et al., 2002). Similarly, it was re­ catfish, I. punctatus that the retinal efferents project towards the supra­
ported that GnRHir-neurons were appeared in the POA-H during chiasmatic nucleus (SCN) and further to other brain areas and later these
gonadal differentiation (Soga, Ogawa, Millar, Sakuma, & Parhar, 2005). SCN neurons projects to the pituitary (Prasada Rao & Sharma, 1982;
These GnRHir-neurons were found to be increased considerably in the Prasada Rao, Job, & Schreibman, 1983). The pineal cells also express
pejerrey during the sex determination period (Somoza, Miranda, Guil­ GnRH receptors in the European sea bass, D. labrax (Servili et al., 2010).
gur, & Strobl-Mazzulla, 2006). Whereas in sea bass, during gonadal Overall the pineal hormone and melatonin affects dopaminergic system,
differentiation, the GnRH levels reached peak in pituitary and brain GnRH, LH, FSH and gonads (Falcón, Migaud, Muñoz-Cueto, & Carrillo,
(Moles, Carrillo, Mañanós, Mylonas, & Zanuy, 2007). Infact, it was 2010).
suggested by Baroiller et al. (1999) that the HHG axis is required for
complete sex differentiation but not its activation. However, stable
GnRH2 and GnRH3 expressions were observed in black sea bass during

531
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

5.3. 5-HT hypothalamic and pituitary fragments also upon T and E2 in vivo treat­
ment in goldfish and considered to be one of the mechanisms facilitating
The 5-HT neurons have a stimulatory effect in neuroendocrine the feedback of these two steroids (Peng, Chang, et al., 1993; Peng,
function by innervating through POA-H upon GnRH-GTH release (Sen­ Trudeau, & Peter, 1993). Increased expression of NPY was evident in the
thilkumaran & Joy, 1996; Senthilkumaran, Okuzawa, Gen, & Kagawa, catfish, C. gariepinus brain and testis, and its expression levels were high
2001; Zohar et al., 2010). 5-HT appeared to be a contributing factor for at 100 and 150 dph during brain development in females compared to
sexual dimorphism in brain. This provided clue to authors in order to males. Predominant expression of NPY was found during the
produce a predominant female population with the use of pre-spawning stage of the female brain compared to other reproductive
para-chlorophenyl alanine, a 5-HT synthesis blocker during the crucial stages (Sudhakumari et al., 2017). NPY also stimulates the release of LH
window of sex determination/differentiation (Raghuveer, Sudhakumari, in goldfish through direct action on gonadotroph by stimulating the
et al., 2011; Tsai, Wang, Chang, & Kao, 2001; Tsai & Wang, 1999). Tph GnRH release (Trudeau, 1997). Even in the red seabream, NPY stimu­
is a homotetramer allosteric enzyme that catalyzes the rate-limiting step lated in vitro release of sbGnRH from POA-H slices during immature,
in 5-HT biosynthesis (Raghuveer, Sudhakumari, et al., 2011; Sudhaku­ spawning and recrudescence stages (Senthilkumaran et al., 2001). The
mari et al., 2010). In the Nile tilapia, Tph2 which is a brain form of tph, action of GnRH release by NPY is direct as it was not altered by antag­
showed prominent expression during 5, 10, 15 and 20 dph XY tilapia onists of NA, gamma-aminobutyric acid and 5-HT. Studies revealed that
whereas no expression in 5–20 dph XX tilapia. Additionally, Tph tran­ central injection of NPY caused increased dose-dependent food intake in
scripts localized through in situ hybridization was found only in 11 dph cohosalmon, channel catfish and goldfish (De Pedro et al., 2000;
male brain specifically in the regions of telencephalon-preoptic area, López-Patiño et al., 1999; Narnaware, Peyon, Lin, & Peter, 2000; Sil­
olfactory bulb and olfactory epithelium but not in the female brain of verstein & Plisetskaya, 2000). In another study, pretreatment of T and E2
same age. Tph immunoreactivity positive cells were observed in nucleus in the goldfish, induced 2–3 fold increase of NPY mRNA expression
preopticus-periventricularis region of male brain of 12 dph but not in levels in the forebrain (telencephalon/preoptic area) region. Moreover,
female brain of same age (Sudhakumari et al., 2010). Due to this, the T activated NPY synthetic activity in the preoptic area of brain that was
dimorphic expression of tph may indicate a role in brain sex differenti­ revealed by in situ hybridization (Peng, Gallin, Peter, Blomqvist, &
ation of the Nile tilapia. In the catfish, C. gariepinus, tph expression was Larhammar, 1994).
increased in males in comparison to females, 50 and 75 dph indicating
variance expression in brain during early development (Raghuveer, 5.6. GDNF
Sudhakumari, et al., 2011). Increased expression of tph2 was correlated
with elevated levels of 5-HT and 5-HTP in the MT treated male brain In addition to these factors, glial cell line-derived neurotrophic factor
fishes, and on the other hand, pCPA treatment decreased tph2 expression (GDNF), which is a potent survival factor, is majorly involved in con­
along with 5-HT and 5-HTP levels in the brain of catfish (Raghuveer, trolling several peripheral and central neurons, including dopaminergic
Sudhakumari, et al., 2011). neurons (Airaksinen, Holm, & Hätinen, 2006). GDNF signals bind to the
GDNF family receptors-α through GPI-anchored receptor, which has a
5.4. Catecholamines strong binding affinity over GFRα-1 (Trupp, Raynoschek, Belluardo, &
Ibáñez, 1998). Decreased expression of GFRα-1 was observed upon
Catecholamines, unlike 5-HT, exerts inhibitory action by dopamine treatment with 1-methyl-1,2,3,6-tetrahydropyridine leading to neuro­
(DA) and facilitatory effect through norepinephrine (NE) on GnRH–GTH degeneration with the correlation of L-3,4-dihydroxyphenylalanine,
synthesis and release (Peter et al., 1991; Senthilkumaran & Joy, 1996). catecholamines (CA), DA and NE levels in catfish (Mamta & Senthilku­
So far epinephrine with detectable levels during resting phase and maran, 2018).
increased levels during pre-spawning and spawning phases in the Asian Overall, despite of these sex dimorphic markers identified in the
stinging catfish, H. fossilis was observed, however no role has been brain except 5-HT, which is dominant to shown to induce male popu­
indicated in terms of GnRH–GTH release (Senthilkumaran & Joy, 1995). lation with its 5-HT synthesis blocking drug, pCPA, otherwise all the
In-depth study on the CAergic system during brain sex development of other neurotransmitters are brain factors and all the brain sex changes
both sexes in the catfish, C. batrachus revealed sex-specific changes are based on gonadal sex (Tsai, Wang, & Fang, 2001). In this context,
whereby the female brain depicted elevated tyrosine hydroxylase (th) mismatching sex steroids, i.e., E2 to male and MT to female through
expression levels and high copy number compared to males (Mamta osmotic pump delivery in the catfish, C. gariepinus, produced differential
et al., 2014). Elevated th expression was observed in female brain at 50, effects on certain brain-related genes (Mamta, Sudhakumari, Kagawa,
75 and 100 dph compared to that of the male brain of same age wherein Dutta-Gupta & Senthilkumaran, 2020). This include minimal expression
50 dph is considered to be as a critical period for gonadal differentiation of cyp19a1b, GnRH1, gfrα-1 and th in the brain of MT treated female fish.
in catfish (Mamta et al., 2014; Raghuveer, Senthilkumaran, et al., 2011). On the other hand, increased expression of cyp19a1b, GnRH1, gfrα-1 and
However, the changes in the levels of DA and NE also showed sexual th in the brain of E2 treated male fish (Mamta et al., 2020). However, the
dimorphism. Whereas in saddleback wrasse, CAs plays an important role transcripts of some of these brain related genes were normally overex­
with the commencement as well as completion of sex reversal (Larson, pressed in females compared to males (Senthilkumaran et al., 2015)
Norris, Gordon Grau, & Summers, 2003). Furthermore, EE2 treatment indicating sex steroid based regulation. Thus, sexually dimporphic
ensued increased CA levels and elevated th expression and MT treatment expression of tph was shown for the first time in tilapia wherein exclu­
revealed lower CA levels, and th expression irrespective of sex (Mamta sive expression of tph is evident 5 to 20 dph male brain but not in female
et al., 2014). This indicated 5-HTergic system is prominent in male brain brain. Furthermore, in catfish such gender specific differences were
development, whereas CAergic system dominates in female brain subsequently shown in tph, 5-HT and 5-HTP (Raghuveer, Sudhakumari,
development thus exhibiting brain sex dimorphism. et al., 2011; Sudhakumari et al., 2010). Differential expression of brain
related genes so far studied exhibiting sexual dimorphism play promi­
5.5. NPY nent role at brain level with response to gonadal differentiation, may
causatively or consequentially responds to gonadal sex (Senthilkumaran
Neuropeptide Y (NPY) is considered as the most pivotal orexigenic et al., 2015). Changes in these brain related-genes along with CAs and
neuropeptide well expressed in mammals, and it has been reported that serum 11-KT, E2, T levels indicate brain-gonadal interactions and their
NPY stimulates the food intake in teleost fish as in observed in mammals influence on the GnRH-GTH axis through gonadal recrudescence. Some
(Halford, Cooper, & Dovey, 2004; Kalra et al., 1999; Volkoff et al., of the sex reversal genetic markers so far identified from brain are useful
2005). NPY stimulates the release of GnRH from preoptic-anterior to understand sex differentiation in addition to the endocrine changes in

532
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

teleost under various conditions including endocrine disruption (Kar, in cyprinids, cichlids and salmonids, the field has emerged very quickly
Sangem, Anusha, & Senthilkumaran, 2021; Rajakumar & Senthilku­ towards chromosome manipulations, polyploidy and gynogenesis.
maran, 2020; Tenugu, Pranoty, Mamta, & Senthilkumaran, 2021). In the Nile tilapia, O. niloticus YY male broodstock was produced
genetically on a commercial scale. Initially, fry with mixed population
6. Monosex population and strategy of a genetic cross was treated with E2, to identify XY neofemales progeny, which were
further crossed with XY males to identify YY males. YY males were then
Teleost, most diverse group of vertebrates has increased globally and crossed to XY neomales, and further offspring were treated with estro­
become attractive animal models for extensive investigation related to gen and then progeny were tested for YY neofemales identification. YY
sex reversal strategies. In order to overcome certain difficulties exhibi­ neofemale and YY male were crossed to generate YY male broodstock
ted by fishes, for example, lack of growth after maturation, attaining production (Mair, Abucay, Abella, Beardmore, & Skibinski, 1997).
sexual maturity at different age, flesh quality deterioration which may Monosex XY population was generated finally after crossing YY male
lead to mortality and finally one gender performing better than the with normal XX female. A further selection of different strains within the
other. Considering these, it is worthwhile to produce monosex pop­ broodstock lines were tested through the GMT™ technique (Tuan, Little,
ulations as well as reproductively sterile fish to overcome the hurdles. & Mair, 1998; Tuan, Mair, Little, & Beardmore, 1999). This methodol­
Sex reversal is one of the strategies/approaches used to produce mon­ ogy was first developed in O. mossambicus for small-scale supermale
osex populations for several purposes in aquaculture. Due to sexual production (Pandian & Varadaraj, 1990). Whereas in medaka the
plasticity, fishes can be easily manipulated to induce sex reversal during monosex population was produced by using Hd-rR.YHNI strain males
critical stages by hormonal treatment to understand pubertal and and crossing with the d-rR strain females and further crossing the males
growth related changes and sex dimorphism (Devlin & Nagahama, produced in the offspring once again with females of d-rR strain which
2002). The efficient and standard treatment widely used by farmers is ultimately resulted in d-rR.YHNI population of males (Scholz et al.,
through dietary administration of MT for 3–4 weeks from 10 dpf to 2003). In another study of turbot, Scophthalmus maximus treatment with
produce monosex male populations in the Nile tilapia (Baroiller & androgen and crossing of induced classic females (fZW) with neomales
D’Cotta, 2001; Baroiller, D’Cotta, & Saillant, 2009). In the Japanese (mZW) produced WW female population (Haffray et al., 2009). Female
hirame, P. olivaceus, both low as well as high temperature induced male monosex population through the genetic cross was also performed in
monosex population whereas intermittent temperature produced 1:1 sex carp and salmonoids (Cnaani & Levavi-Sivan, 2009; Piferrer, 2001).
ratio (Yamamoto, 1999). Interestingly other methodologies such as ge­ However, better survival rates of YY males were demonstrated in yellow
netic cross made applicable to produce monosex population, which was catfish, tilapia and rainbow trout compared to low survival rates in other
implied in few species. Initiating with the steroid treatment and com­ species such as medaka and fighting fish (George, Pandian, & Kavum­
bined with hybridization in order to produce sex reversal and sterile fish purath, 2013). This is a good strategy to develop monosex population

Fig. 2. Illustrating the genetic cross and pedigree selection in tilapia.

533
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

across species which should be taken into consideration and make major Afonso, L. O., Wassermann, G. J., & Terezinha de Oliveira, R. (2001). Sex reversal in Nile
tilapia (Oreochromis niloticus) using a nonsteroidal aromatase inhibitor. Journal of
inputs for research in aquaculture. The pedigree for the production of
Experimental Zoology, 290(2), 177–181. https://doi.org/10.1002/jez.1047
monosex population in tilapia through hormonal treatment followed by Airaksinen, M. S., Holm, L., & Hätinen, T. (2006). Evolution of the GDNF family ligands
genetic cross and progeny selection is depicted in Fig. 2. and receptors. Brain, Behavior and Evolution, 68(3), 181–190. https://doi.org/
10.1159/000094087
Ando, H., Hew, C. L., & Urano, A. (2001). Signal transduction pathways and transcription
7. Conclusion and future perspectives factors involved in the gonadotropin-releasing hormone-stimulated gonadotropin
subunit gene expression. Comparative Biochemistry and Physiology Part B: Biochemistry
In both wild and domestic strains of teleost, existence of spontaneous and Molecular Biology, 129(2–3), 525–532. https://doi.org/10.1016/s1096-4959(01)
00356-6
sex reversed and induced sex reversed individuals have been reported. Ando, H., & Urano, A. (2005). Molecular regulation of gonadotropin secretion by
In most of the species, E2 and 11-KT considered as the predominant gonadotropin releasing hormone in salmonid fishes. Zoological Science, 22(4),
steroids which are linked in the regulation of sex change. The estab­ 379–389. https://doi.org/10.2108/zsj.22.379
Anitha, A., & Senthilkumaran, B. (2020). Role of sox30 in regulating testicular
lishment of monosex population in mammals is easy as X is bigger than steroidogenesis of common carp. The Journal of Steroid Biochemistry and Molecular
the Y chromosome whereas in fish no distinguish X and Y chromosomes Biology, 204, 105769. https://doi.org/10.1016/j.jsbmb.2020.105769
are present which is often difficult to distinguish through short gun gene Anitha, A., & Senthilkumaran, B. (2022). Sox19 regulates ovarian steroidogenesis in
common carp. The Journal of Steroid Biochemistry and Molecular Biology, 217, 106044.
approach. Inspite of this, genetic cross technique with XY, ZZ population https://doi.org/10.1016/j.jsbmb.2021.106044
made it easy to work for inducing sex reversal and this is the best way up Baroiller, J. F., Chourrout, D., Fostier, A., & Jalabert, B. (1995). Temperature and sex
to now to obtain monosex population. By combining both natural and chromosomes govern sex ratios of the mouthbrooding cichlid fish Oreochromis
niloticus. Journal of Experimental Zoology, 273(3), 216–223. https://doi.org/
induced steroid and environmental factors sex reversal had improved in
10.1002/jez.1402730306
more advance and used in species such as medaka, tilapia and trout Baroiller, J., & D’Cotta, H. (2001). Environment and sex determination in farmed fish.
which is often difficult. With the consideration of diversity and plasticity Comparative Biochemistry and Physiology - Part C: Toxicology & Pharmacology, 130(4),
different species have varied mechanism to undergo sex-change by 399–409. https://doi.org/10.1016/s1532-0456(01)00267-8
Baroiller, J., D’Cotta, H., Bezault, E., Wessels, S., & Hoerstgen-Schwark, G. (2009).
signifying independence evolvement of sex reversal. As reported the Tilapia sex determination: Where temperature and genetics meet. Comparative
spontaneous and induced sex reversal in teleost understandably Biochemistry and Physiology Part A: Molecular & Integrative Physiology, 153(1), 30–38.
revealed the amazing sexual plasticity exhibited by fishes. The hormonal https://doi.org/10.1016/j.cbpa.2008.11.018
Baroiller, J., D’Cotta, H., & Saillant, E. (2009). Environmental effects on fish sex
treatment warrants to maximize the growth with diverse nutrients also determination and differentiation. Sexual Development, 3(2–3), 118–135. https://doi.
ensuring the meat quality and enhancing the commercial value of the org/10.1159/000223077
edible fish. The technique or by the combination of ploidy induction Baroiller, J., Guiguen, Y., & Fostier, A. (1999). Endocrine and environmental aspects of
sex differentiation in fish. Cellular and Molecular Life Sciences, 55(7), 910. https://
provides a scope for the male and female broodstock development, doi.org/10.1007/s000180050344
which helps us to understand the underlying mechanisms in the sex Benjamín Barón, S., Fernando Bückle, R., & Espina, S. (2002). Environmental factors and
determination and differentiation process. Studies on spontaneous and sexual differentiation in Poecilia sphenops Valenciennes (Pisces: Poeciliidae).
Aquaculture Research, 33(8), 615–619. https://doi.org/10.1046/j.1365-
induced sex reversals in fish provides a clue for better understanding the 2109.2002.00699.x
amazing diversity exhibited by fish as well as vertebrate sex determi­ Beullens, K., Eding, E. H., Gilson, P., Ollevier, F., Komen, J., & Richter, C. J. J. (1997).
nation. However, the steroid hormones, environmental conditions Gonadal differentiation, intersexuality and sex ratios of European eel (Anguilla
anguill L.) maintained in captivity. Aquaculture, 153(1–2), 135–150. https://doi.org/
stimulating specific sex bias and genes have an independent parallel
10.1016/S0044-8486(97)00018-5
differentiating effect which can interrelate to each other but often syn­ Bhandari, R. K., Higa, M., Nakamura, S., & Nakamura, M. (2004). Aromatase inhibitor
ergistically leads to sex differences in the brain. Future work in combi­ induces complete sex change in the protogynous honeycomb grouper (Epinephelus
nation with economical relevance on sex reversal strategy ensure merra). Molecular Reproduction and Development, 67(3), 303–307. https://doi.org/
10.1002/mrd.20027
development of monosex population for aquaculture benefits. Further­ Bhandari, R. K., Komuro, H., Nakamura, S., Higa, M., & Nakamura, M. (2003). Gonadal
more, understanding sex reversal in multiple fish species at the molec­ restructuring and correlative steroid hormone profiles during natural sex change in
ular level may provide new gene markers to distinguish endocrine protogynous honeycomb grouper (Epinephelus merra). Zoological Science, 20(11),
1399–1404. https://doi.org/10.2108/zsj.20.1399
disruption at all levels. Biswas, C., Chakraborty, S., Munilkumar, S., Gireesh-Babu, P., Sawant, P. B.,
Chadha, N. K., et al. (2021). Effect of high temperature during larval and juvenile
Conflict of interest stages on masculinization of common carp (Cyprinus carpio, L). Aquaculture, 530,
735803. https://doi.org/10.1016/j.aquaculture.2020.735803
Blazquez, M., Navarro-Martín, L., & Piferrer, F. (2009). Expression profiles of sex
The authors declare no conflict of interest concerning to the research differentiation-related genes during ontogenesis in the European sea bass acclimated
report in the manuscript. to two different temperatures. Journal of Experimental Zoology Part B: Molecular and
Developmental Evolution, 312(7), 686–700. https://doi.org/10.1002/jez.b.21286
Blázquez, M., & Somoza, G. M. (2010). Fish with thermolabile sex determination (TSD)
Acknowledgements as models to study brain sex differentiation. General and Comparative Endocrinology,
166(3), 470–477. https://doi.org/10.1016/j.ygcen.2009.10.004\\
Blázquez, M., Zanuy, S., Carrillo, M., & Piferrer, F. (1998). Structural and functional
The research work mentioned in this review was supported by grant-
effects of early exposure to estradiol-17β and 17α-ethynylestradiol on the gonads of
in-aid (Ref. No. EMR/2017/000718) from the Science and Engineering the gonochoristic teleost Dicentrarchus labrax. Fish Physiology and Biochemistry, 18(1),
Research Board, India to BS. ST is thankful to Senior Research Fellow­ 37–47. https://doi.org/10.1023/A:1007736110663
ship support by Council of Scientific and Industrial Research (Ref. No. Borg, B. (1994). Androgens in teleost fishes. Comparative Biochemistry and Physiology Part
C: Pharmacology, Toxicology and Endocrinology, 109(3), 219–245. https://doi.org/
09/414(1150)/2017-EMR-I), India. Authors acknowledge BUILDER 10.1016/0742-8413(94)00063-g
Grant from DBT (Ref. No. BUILDER-DBT-BT/INF/22/SP41176/2020), Breton, T. S., DiMaggio, M. A., Sower, S. A., & Berlinsky, D. L. (2015). Brain aromatase
India to School of Life Sciences, University of Hyderabad. (cyp19a1b) and gonadotropin releasing hormone (gnrh2 and gnrh3) expression
during reproductive development and sex change in Black Sea bass (Centropristis
striata). Comparative Biochemistry and Physiology Part A: Molecular & Integrative
References Physiology, 181, 45–53. https://doi.org/10.1016/j.cbpa.2014.11.020
Bushmann, P. J., & Burns, J. R. (1994). Social control of male sexual maturation in the
Abozaid, H., Wessels, S., & Hörstgen-Schwark, G. (2012). Elevated temperature applied swordtail characin, Corynopoma riisei. Journal of Fish Biology, 44(2), 263–272.
during gonadal transformation leads to male bias in Zebrafish (Danio rerio). Sexual https://doi.org/10.1111/j.1095-8649.1994.tb01204.x
Development, 6(4), 201–209. https://doi.org/10.1159/000336297 Cáceres, G., López, M. E., Cádiz, M. I., Yoshida, G. M., Jedlicki, A., Palma-Véjares, R.,
Abucay, J. S., Mair, G. C., Skibinski, D. O., & Beardmore, J. A. (1999). Environmental sex et al. (2019). Fine mapping using whole-genome sequencing confirms anti-müllerian
determination: The effect of temperature and salinity on sex ratio in Oreochromis hormone as a major gene for sex determination in farmed Nile tilapia (Oreochromis
niloticus L. Aquaculture, 173(1–4), 219–234. https://doi.org/10.1016/S0044-8486 niloticus L.). G3 Genes|Genomes|Genetics, 9(10), 3213–3223. https://doi.org/
(98)00489-X 10.1534/g3.119.400297

534
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

Cardwell, J., & Liley, N. (1991). Hormonal control of sex and color change in the Goos, H. J., Senthilkumaran, B., & Joy, K. P. (1999). Neuroendocrine integrative
stoplight parrotfish, Sparisoma viride. General and Comparative Endocrinology, 81(1), mechanisms in the control of gonadotropin secretion in teleosts. Comparative
7–20. https://doi.org/10.1016/0016-6480(91)90120-u endocrinology and reproduction, 114–137.
Chang, C., & Lin, B. (1998). Estradiol-17β stimulates aromatase activity and reversible Göppert, C., Harris, R. M., Theis, A., Boila, A., Hohl, S., Rüegg, A., et al. (2016).
sex change in protandrous Black porgy, Acanthopagrus schlegeli. Journal of Inhibition of Aromatase induces partial sex change in a cichlid fish: Distinct
Experimental Zoology, 280(2), 165–173. https://doi.org/10.1002/(sici)1097-010x functions for sex steroids in brains and gonads. Sexual Development, 10(2), 97–110.
(19980201)280:2<165::aid-jez7>3.0.co;2-m https://doi.org/10.1159/000445463
Cnaani, A., & Levavi-Sivan, B. (2009). Sexual development in fish, practical applications Goudie, C. A., Redner, B. D., Simco, B. A., & Davis, K. B. (1983). Feminization of channel
for aquaculture. Sexual Development, 3(2–3), 164–175. https://doi.org/10.1159/ catfish by oral administration of steroid sex hormones. Transactions of the American
000223080 Fisheries Society, 112(5), 670–672. https://doi.org/10.1577/1548-8659(1983)
Colombo, G., & Grandi, G. (1995). Sex differentiation in the European eel: Histological 112<670:foccbo>2.0.co;2
analysis of the effects of sex steroids on the gonad. Journal of Fish Biology, 47(3), Grober, M. S., Jackson, I. M., & Bass, A. H. (1991). Gonadal steroids affect LHRH preoptic
394–413. https://doi.org/10.1111/j.1095-8649.1995.tb01909.x cell number in a sex/role changing fish. Journal of Neurobiology, 22(7), 734–741.
Conover, D. O., & Kynard, B. E. (1981). Environmental sex determination: Interaction of https://doi.org/10.1002/neu.480220708
temperature and genotype in a fish. Science, 213(4507), 577–579. https://doi.org/ Guiguen, Y., Baroiller, J., Ricordel, M., Iseki, K., Mcmeel, O., Martin, S., et al. (1999).
10.1126/science.213.4507.577 Involvement of estrogens in the process of sex differentiation in two fish species: The
Consten, D., Keuning, E., Bogerd, J., Zandbergen, M., Lambert, J., Komen, J., et al. rainbow trout (Oncorhynchus mykiss) and a tilapia (Oreochromis niloticus). Molecular
(2002). Sex steroids and their involvement in the cortisol-induced inhibition of Reproduction and Development, 54(2), 154–162. https://doi.org/10.1002/(sici)1098-
pubertal development in male common carp, Cyprinus carpio L.1. Biology of 2795(199910)54:2<154::aid-mrd7>3.0.co;2-5
Reproduction, 67(2), 465–472. https://doi.org/10.1095/biolreprod67.2.465 Guiguen, Y., Jalabert, B., Benett, A., & Fostier, A. (1995). Gonadal in vitro
D’cotta, H., Fostier, A., Guiguen, Y., Govoroun, M., & Baroiller, J. (2001). Aromatase Androstenedione metabolism and changes in some plasma and gonadal steroid
plays a key role during normal and temperature-induced sex differentiation of tilapia hormones during sex inversion of the protandrous sea bass, Lates calcarifer. General
Oreochromis niloticus. Molecular Reproduction and Development, 59(3), 265–276. and Comparative Endocrinology, 100(1), 106–118. https://doi.org/10.1006/
https://doi.org/10.1002/mrd.1031 gcen.1995.1139
Davey, A. J., & Jellyman, D. J. (2005). Sex determination in freshwater eels and Guiguen, Y., Jalabert, B., Thouard, E., & Fostier, A. (1993). Changes in plasma and
management options for manipulation of sex. Reviews in Fish Biology and Fisheries, 15 gonadal steroid hormones in relation to the reproductive cycle and the sex inversion
(1–2), 37–52. https://doi.org/10.1007/s11160-005-7431-x process in the protandrous Seabass, Lates calcarifer. General and Comparative
De Pedro, N., López-Patiño, M. A., Guijarro, A. I., Pinillos, M. L., Delgado, M. J., & Endocrinology, 92(3), 327–338. https://doi.org/10.1006/gcen.1993.1170
Alonso-Bedate, M. (2000). NPY receptors and opioidergic system are involved in Gupta, Y. R., & Senthilkumaran, B. (2020). Common carp pentraxin gene: Evidence for its
NPY-induced feeding in goldfish. Peptides, 21(10), 1495–1502. https://doi.org/ role in ovarian differentiation and growth. General and Comparative Endocrinology,
10.1016/s0196-9781(00)00303-x 290, 113398. https://doi.org/10.1016/j.ygcen.2020.113398
Degani, G., & Kushnirov, D. (1992). Effects of 17β-Estradiol and grouping on sex Haffray, P., Lebègue, E., Jeu, S., Guennoc, M., Guiguen, Y., Baroiller, J. F., et al. (2009).
determination of European eels. The Progressive Fish-Culturist, 54(2), 88–91. https:// Genetic determination and temperature effects on turbot Scophthalmus maximus sex
doi.org/10.1577/1548-8640(1992)054<0088:eoeago>2.3.co;2 differentiation: An investigation using steroid sex-inverted males and females.
Devlin, R. H., & Nagahama, Y. (2002). Sex determination and sex differentiation in fish: Aquaculture, 294(1–2), 30–36. https://doi.org/10.1016/j.aquaculture.2009.05.004
An overview of genetic, physiological, and environmental influences. Aquaculture, Halford, J., Cooper, G., & Dovey, T. (2004). The pharmacology of human appetite
208(3–4), 191–364. https://doi.org/10.1016/s0044-8486(02)00057-1 expression. Current Drug Targets, 5(3), 221–240. https://doi.org/10.2174/
Fernandino, J. I., Hattori, R. S., Moreno Acosta, O. D., Strüssmann, C. A., & 1389450043490541
Somoza, G. M. (2013). Environmental stress-induced testis differentiation: Androgen Han, J., Hu, Y., Qi, Y., Yuan, C., Naeem, S., & Huang, D. (2021). High temperature
as a by-product of cortisol inactivation. General and Comparative Endocrinology, 192, induced masculinization of zebrafish by down-regulation of sox9b and esr1 via DNA
36–44. https://doi.org/10.1016/j.ygcen.2013.05.024 methylation. Journal of Environmental Sciences, 107, 160–170. https://doi.org/
Fernandino, J., Popesku, J., Paul-Prasanth, B., Xiong, H., Hattori, R., Oura, M., et al. 10.1016/j.jes.2021.01.032
(2011). Analysis of sexually dimorphic expression of genes at early Gonadogenesis of Haniffa, M. A., Sridhar, S., & Nagarajan, M. (2004). Hormonal manipulation of sex in
Pejerrey Odontesthes bonariensis using a heterologous Microarray. Sexual stinging catfish Heteropneustes fossilis (Bloch). Current Science, 1012–1017. https://
Development, 5(2), 89–101. https://doi.org/10.1159/000324423 www.jstor.org/stable/24109289.
Francis, R. C., & Barlow, G. W. (1993). Social control of primary sex differentiation in the Hassin, S., De Monbrison, D., Hanin, Y., Elizur, A., Zohar, Y., & Popper, D. (1997).
Midas cichlid. Proceedings of the National Academy of Sciences, 90(22), 10673–10675. Domestication of the white grouper, Epinephelus aeneus 1. Growth and reproduction.
https://doi.org/10.1073/pnas.90.22.10673 Aquaculture, 156(3–4), 305–316. https://doi.org/10.1016/s0044-8486(97)00136-1
Fricke, H., & Fricke, S. (1977). Monogamy and sex change by aggressive dominance in Hattori, R. S., Fernandino, J. I., Kishii, A., Kimura, H., Kinno, T., Oura, M., et al. (2009).
coral reef fish. Nature, 266(5605), 830–832. https://doi.org/10.1038/266830a0 Cortisol-induced Masculinization: Does thermal stress affect gonadal fate in Pejerrey,
Friesen, C. N., Aubin-Horth, N., & Chapman, L. J. (2012). The effect of hypoxia on sex a teleost fish with temperature-dependent sex determination? PLoS One, 4(8), e6548.
hormones in an African cichlid Pseudocrenilabrus multicolor victoriae. Comparative https://doi.org/10.1371/journal.pone.0006548
Biochemistry and Physiology Part A: Molecular & Integrative Physiology, 162(1), 22–30. Hattori, R., Gould, R., Fujioka, T., Saito, T., Kurita, J., Strüssmann, C., et al. (2007).
https://doi.org/10.1016/j.cbpa.2012.01.019 Temperature-dependent sex determination in Hd-RR medaka Oryzias latipes: Gender
Frisch, A. (2004). Sex-change and gonadal steroids in sequentially-hermaphroditic sensitivity, thermal threshold, critical period, and dmrt1 expression profile. Sexual
teleost fish. Reviews in Fish Biology and Fisheries, 14(4), 481–499. https://doi.org/ Development, 1(2), 138–146. https://doi.org/10.1159/000100035
10.1007/s11160-005-3586-8 Hattori, R. S., Murai, Y., Oura, M., Masuda, S., Majhi, S. K., Sakamoto, T., et al. (2012).
Fuzzen, M. L., Alderman, S. L., Bristow, E. N., & Bernier, N. J. (2011). Ontogeny of the A Y-linked anti-mullerian hormone duplication takes over a critical role in sex
corticotropin-releasing factor system in rainbow trout and differential effects of determination. Proceedings of the National Academy of Sciences, 109(8), 2955–2959.
hypoxia on the endocrine and cellular stress responses during development. General https://doi.org/10.1073/pnas.1018392109
and Comparative Endocrinology, 170(3), 604–612. https://doi.org/10.1016/j. Hayashi, Y., Kobira, H., Yamaguchi, T., Shiraishi, E., Yazawa, T., Hirai, T., et al. (2010).
ygcen.2010.11.022 High temperature causes masculinization of genetically female medaka by elevation
Galvez, J. I., Mazik, P. M., Phelps, R. P., & Mulvaney, D. R. (1995). Masculinization of of cortisol. Molecular Reproduction and Development, 77(8), 679–686. https://doi.org/
channel catfish Ictalurus punctatus by oral administration of Trenbolone acetate. 10.1002/mrd.21203
Journal of the World Aquaculture Society, 26(4), 378–383. https://doi.org/10.1111/ Hayes, T. B. (1998). Sex determination and primary sex differentiation in amphibians:
j.1749-7345.1995.tb00832.x Genetic and developmental mechanisms. Journal of Experimental Zoology, 281(5),
Gennotte, V., Mélard, C., D’Cotta, H., Baroiller, J., & Rougeot, C. (2014). The sensitive 373–399. https://doi.org/10.1002/(sici)1097-010x(19980801)281:5<373::aid-
period for male-to-female sex reversal begins at the embryonic stage in the Nile jez4>3.0.co;2-l
tilapia and is associated with the sexual genotype. Molecular Reproduction and Helfman, G. S., Collette, B. B., & Facey, D. E. (1997). The diversity of fishes. Malden, MA:
Development, 81(12), 1146–1158. https://doi.org/10.1002/mrd.22436 Blackwell Science.
George, T., Pandian, T. J., & Kavumpurath, S. (2013). Inviability of yy zygotes of the Higa, M., Ogasawara, K., Sakaguchi, A., Nagahama, Y., & Nakamura, M. (2003). Role of
fighting fish, betta-splendens. Israeli Journal of Aquaculture-Bamidgeh, 46(1), 3–8. steroid hormones in sex change of protogynous wrasse. Fish Physiology and
http://ir.mkuniversity.ac.in/id/eprint/1489. Biochemistry, 28(1–4), 149–150. https://doi.org/10.1023/b:
Glamuzina, B., Glavić, N., Skaramuca, B., & Kožul, V. (1998). Induced sex reversal of fish.0000030505.28138.d1
dusky grouper, Epinephelus marginatus(Lowe). Aquaculture Research, 29(8), 563–567. Hiraki, T., Takeuchi, A., Tsumaki, T., Zempo, B., Kanda, S., Oka, Y., et al. (2012). Female-
https://doi.org/10.1046/j.1365-2109.1998.00238.x specific target sites for both oestrogen and androgen in the teleost brain. Proceedings
Godwin, J. (2010). Neuroendocrinology of sexual plasticity in teleost fishes. Frontiers in of the Royal Society B: Biological Sciences, 279(1749), 5014–5023. https://doi.org/
Neuroendocrinology, 31(2), 203–216. https://doi.org/10.1016/j.yfrne.2010.02.002 10.1098/rspb.2012.2011
Godwin, J. R., & Thomas, P. (1993). Sex change and steroid profiles in the protandrous Hofmann, H. A. (2006). Gonadotropin-releasing hormone signaling in behavioral
Anemonefish Amphiprion melanopus (Pomacentridae, teleostei). General and plasticity. Current Opinion in Neurobiology, 16(3), 343–350. https://doi.org/10.1016/
Comparative Endocrinology, 91(2), 144–157. https://doi.org/10.1006/ j.conb.2006.05.005
gcen.1993.1114 Holmgren, K. (1996). Effect of water temperature and growth variation on the sex ratio
Gomes, C., Costa, F., & Borella, M. (2013). Distribution of GnRH in the brain of the of experimentally reared eels. Ecology of Freshwater Fish, 5(4), 203–212. https://doi.
freshwater teleost Astyanax altiparanae. Micron, 52–53, 33–38. https://doi.org/ org/10.1111/j.1600-0633.1996.tb00134.x
10.1016/j.micron.2013.07.008 Hourigan, T. F., Nakamura, M., Nagahama, Y., Yamauchi, K., & Grau, E. (1991).
Histology, ultrastructure, and in vitro steroidogenesis of the testes of two male

535
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

phenotypes of the protogynous fish, Thalassoma duperrey (Labridae). General and Koya, Y., Fujita, A., Niki, F., Ishihara, E., & Miyama, H. (2003). Sex differentiation and
Comparative Endocrinology, 83(2), 193–217. https://doi:10.1016/0016-6480(91) pubertal development of gonads in the viviparous mosquitofish. Gambusia affinis
90023-y. Zoological Science, 20(10), 1231–1242. https://doi.org/10.2108/zsj.20.1231
Hunter, G. A., & Donaldson, E. M. (1983). 5 Hormonal sex control and its application to Kramer, C. R., Koulish, S., & Bertacchi, P. L. (1988). The effects of testosterone implants
fish culture. Fish Physiology, 9, 223–303. https://doi.org/10.1016/S1546-5098(08) on ovarian morphology in the bluehead wrasse, Thalassoma bifasciatum (Bloch)
60305-2 (Teleostei: Labridae). Journal of Fish Biology, 32(3), 397–407. https://doi.org/
Ito, L. S., Takahashi, C., Yamashita, M., & Strüssmann, C. A. (2008). Warm water induces 10.1111/j.1095-8649.1988.tb05376.x
apoptosis, gonadal degeneration, and germ cell loss in subadult pejerrey Odontesthes Kroon, F. J., & Liley, N. (2000). The role of steroid hormones in protogynous sex change
bonariensis (Pisces, Atheriniformes). Physiological and Biochemical Zoology, 81(6), in the Blackeye goby, Coryphopterus nicholsii (Teleostei: Gobiidae). General and
762–774. https://doi.org/10.1086/590219 Comparative Endocrinology, 118(2), 273–283. https://doi.org/10.1006/
Iwamatsu, T., Kobayashi, H., Hamaguchi, S., Sagegami, R., & Shuo, T. (2005). Estradiol- gcen.2000.7459
17β content in developing eggs and induced sex reversal of the medaka (Oryzias Kroon, F. J., Munday, P. L., & Pankhurst, N. W. (2003). Steroid hormone levels and Bi-
latipes). Journal of Experimental Zoology Part A: Comparative Experimental Biology, directional sex change in Gobiodon histrio. Journal of Fish Biology, 62(1), 153–167.
303A(2), 161–167. https://doi.org/10.1002/jez.a.120 https://doi.org/10.1046/j.1095-8649.2003.00017.x
Kah, O., Lethimonier, C., Somoza, G., Guilgur, L., Vaillant, C., & Lareyre, J. (2007). GnRH Krueger, W. H., & Oliveira, K. (1999). Evidence for environmental sex determination in
and GnRH receptors in metazoa: A historical, comparative, and evolutive the American eel, Anguilla rostrata. Environmental Biology of Fishes, 55(4), 381–389.
perspective. General and Comparative Endocrinology, 153(1–3), 346–364. https://doi. https://doi.org/10.1023/a:1007575600789
org/10.1016/j.ygcen.2007.01.030 Kuo, C., Ting, Y., & Yeh, S. (1988). Induced sex reversal and spawning of blue-spotted
Kalra, S. P., Dube, M. G., Pu, S., Xu, B., Horvath, T. L., & Kalra, P. S. (1999). Interacting grouper. Epinephelus fario. Aquaculture, 74(1–2), 113–126. https://doi.org/10.1016/
appetite-regulating pathways in the hypothalamic regulation of body weight. 0044-8486(88)90091-9
Endocrine Reviews, 20(1), 68–100. https://doi.org/10.1210/edrv.20.1.0357 Kwon, J. Y., McAndrew, B. J., & Penman, D. J. (2002). Treatment with an aromatase
Kamiya, T., Kai, W., Tasumi, S., Oka, A., Matsunaga, T., Mizuno, N., et al. (2012). inhibitor suppresses high-temperature feminization of genetic male (YY) Nile tilapia.
A trans-species Missense SNP in Amhr2 is associated with sex determination in the Journal of Fish Biology, 60(3), 625–636. https://doi.org/10.1111/j.1095-8649.2002.
tiger Pufferfish, Takifugu rubripes (Fugu). PLoS Genetics, 8(7), Article e1002798. tb01689.x
https://doi.org/10.1371/journal.pgen.1002798 Larson, E. T., Norris, D. O., Gordon Grau, E., & Summers, C. H. (2003). Monoamines
Kanamori, A., Yamamura, A., Koshiba, S., Lee, J., Orlando, E. F., & Hori, H. (2006). stimulate sex reversal in the saddleback wrasse. General and Comparative
Methyltestosterone efficiently induces male development in the self-fertilizing Endocrinology, 130(3), 289–298. https://doi.org/10.1016/s0016-6480(02)00622-6
hermaphrodite fish, Kryptolebias marmoratus. Genesis, 44(10), 495–503. https://doi. Lee, S., Kime, D., Chao, T., Lim, H., Chou, R., Lam, T., et al. (1995). In vitro metabolism
org/10.1002/dvg.20240 of testosterone by gonads of the grouper (Epinephelus tauvina) before and after sex
Kar, S., Sangem, P., Anusha, N., & Senthilkumaran, B. (2021). Endocrine disruptors in inversion with 17α-Methyltestosterone. General and Comparative Endocrinology, 99
teleosts: Evaluating environmental risks and biomarkers. Aquaculture and Fisheries, 6 (1), 41–49. https://doi.org/10.1006/gcen.1995.1083
(1), 1–26. https://doi.org/10.1016/j.aaf.2020.07.013 Lee, Y., Lee, F., Yueh, W., Tacon, P., Du, J., Chang, C., et al. (2000). Profiles of gonadal
Khan, I., & Thomas, P. (2000). Lead and Aroclor 1254 disrupt reproductive development, sex steroids, Aromatase activity, and gonadotropin II in the controlled
neuroendocrine function in Atlantic croaker. Marine Environmental Research, 50 sex change of protandrous Black porgy, Acanthopagrus schlegeli Bleeker. General and
(1–5), 119–123. https://doi.org/10.1016/s0141-1136(00)00108-2 Comparative Endocrinology, 119(1), 111–120. https://doi.org/10.1006/
Khan, I. A., & Thomas, P. (2001). Disruption of neuroendocrine control of luteinizing gcen.2000.7499
hormone secretion by Aroclor 1254 involves inhibition of hypothalamic tryptophan Liu, S., Yao, Z., & Wang, Y. (1996). Sex hormone induction of sex reversal in the teleost
Hydroxylase Activity1. Biology of Reproduction, 64(3), 955–964. https://doi.org/ Clarias lazera and evidence for female homogamety and male heterogamety. Journal
10.1095/biolreprod64.3.955 of Experimental Zoology, 276(6), 432–438. https://doi.org/10.1002/(sici)1097-010x
Kime, D. E. (1993). ‘Classical’ and ‘non-classical’ reproductive steroids in fish. Reviews in (19961215)276:6<432::aid-jez7>3.0.co;2-o
Fish Biology and Fisheries, 3(2), 160–180. https://doi.org/10.1007/bf00045230 López-Patiño, M. A., Guijarro, A. I., Isorna, E., Delgado, M. J., Alonso-Bedate, M., & De
Kitano, T., Hayashi, Y., Shiraishi, E., & Kamei, Y. (2012). Estrogen rescues Pedro, N. (1999). Neuropeptide Y has a stimulatory action on feeding behavior in
masculinization of genetically female medaka by exposure to cortisol or high goldfish (Carassius auratus). European Journal of Pharmacology, 377(2–3), 147–153.
temperature. Molecular Reproduction and Development, 79(10), 719–726. https://doi. https://doi.org/10.1016/s0014-2999(99)00408-2
org/10.1002/mrd.22080 Lowe, T. P., & Larkin, J. R. (1975). Sex reversal inBetta splendens Regan with emphasis
Kitano, T., Takamune, K., Kobayashi, T., Nagahama, Y., & Abe, S. (1999). Suppression of on the problem of sex determination. Journal of Experimental Zoology, 191(1), 25–31.
P450 aromatase gene expression in sex-reversed males produced by rearing https://doi.org/10.1002/jez.1401910104
genetically female larvae at a high water temperature during a period of sex Maack, G., & Segner, H. (2003). Morphological development of the gonads in zebrafish.
differentiation in the Japanese flounder (Paralichthys olivaceus). Journal of Molecular Journal of Fish Biology, 62(4), 895–906. https://doi.org/10.1046/j.1095-
Endocrinology, 23(2), 167–176. https://doi.org/10.1677/jme.0.0230167 8649.2003.00074.x
Kitano, T., Takamune, K., Nagahama, Y., & Abe, S. (2000). Aromatase inhibitor and 17?- Mair, G. C., Abucay, J. S., Abella, T. A., Beardmore, J. A., & Skibinski, D. (1997). Genetic
methyltestosterone cause sex-reversal from genetical females to phenotypic males manipulation of sex ratio for the large-scale production of all-male tilapia
and suppression of P450 aromatase gene expression in Japanese flounder Oreochromis niloticus. Canadian Journal of Fisheries and Aquatic Sciences, 54(2),
(Paralichthys olivaceus). Molecular Reproduction and Development, 56(1), 1–5. https:// 396–404. https://doi.org/10.1139/f96-282
doi.org/10.1002/(sici)1098-2795(200005)56:1<1::aid-mrd1>3.0.co;2-3 Mamta, S. K., Raghuveer, K., Sudhakumari, C., Rajakumar, A., Basavaraju, Y., &
Kobayashi, T. (2013). Molecular mechanisms of gonadal sex differentiation and sex- Senthilkumaran, B. (2014). Cloning and expression analysis of tyrosine hydroxylase
reversal in fish: Tilapia and medaka. In B. Senthilkumaran (Ed.), Sexual plasticity and and changes in catecholamine levels in brain during ontogeny and after sex steroid
gametogenesis in fishes (pp. 203–220). USA: Nova Biomedical (Chapter 13). analogues exposure in the catfish, Clarias batrachus. General and Comparative
Kobayashi, T., Kajiura-Kobayashi, H., Guan, G., & Nagahama, Y. (2008). Sexual Endocrinology, 197, 18–25. https://doi.org/10.1016/j.ygcen.2013.11.022
dimorphic expression of DMRT1 and Sox9a during gonadal differentiation and Mamta, S., & Senthilkumaran, B. (2018). GDNF family receptor α-1 in catfish: Possible
hormone-induced sex reversal in the teleost fish Nile tilapia (Oreochromis niloticus). implication to brain dopaminergic activity. Brain Research Bulletin, 140, 270–280.
Developmental Dynamics, 237(1), 297–306. https://doi.org/10.1002/dvdy.21409 https://doi.org/10.1016/j.brainresbull.2018.05.010
Kobayashi, T., Kajiura-Kobayashi, H., & Nagahama, Y. (2003). Induction of XY sex Mamta, S., Sudhakumari, C., Kagawa, H., Dutta-Gupta, A., & Senthilkumaran, B. (2020).
reversal by estrogen involves altered gene expression in a teleost, tilapia. Cytogenetic Controlled release of sex steroids through osmotic pump alters brain GnRH1 and
and Genome Research, 101(3–4), 289–294. https://doi.org/10.1159/000074351 catecholaminergic system dimorphically in the catfish, Clarias gariepinus. Brain
Kobayashi, T., Matsuda, M., Kajiura-Kobayashi, H., Suzuki, A., Saito, N., Nakamoto, M., Research Bulletin, 164, 325–333. https://doi.org/10.1016/j.
et al. (2004). Two DM domain genes,DMY andDMRT1, involved in testicular brainresbull.2020.08.022
differentiation and development in the medaka, Oryzias latipes. Developmental Maruska, K. P., & Fernald, R. D. (2011). Social regulation of gene expression in the
Dynamics, 231(3), 518–526. https://doi.org/10.1002/dvdy.20158 hypothalamic-pituitary-gonadal axis. Physiology, 26(6), 412–423. https://doi.org/
Kobayashi, Y., Nagahama, Y., & Nakamura, M. (2013). Diversity and plasticity of sex 10.1152/physiol.00032.2011
determination and differentiation in fishes. Sexual Development, 7(1–3), 115–125. Matsuda, M., Nagahama, Y., Shinomiya, A., Sato, T., Matsuda, C., Kobayashi, T., et al.
https://doi.org/10.1159/000342009 (2002). DMY is a Y-specific DM-domain gene required for male development in the
Kobayashi, M., Saoshiro, S., Kawaguchi, Y., Hayakawa, Y., & Munakata, A. (2013). medaka fish. Nature, 417(6888), 559–563. https://doi.org/10.1038/nature751
Sexual plasticity of behavior in goldfish. In B. Senthilkumaran (Ed.), Sexual plasticity Matsuda, M., Sato, T., Toyazaki, Y., Nagahama, Y., Hamaguchi, S., & Sakaizumi, M.
and gametogenesis in fishes (pp. 183–201). USA: Nova Biomedical (Chapter 12). (2003). Oryzias curvinotus has DMY, a gene that is required for male development in
Komen, J., De Boer, P., & Richter, C. J. (1992). Male sex reversal in Gynogenetic XX the medaka, O. latipes. Zoological Science, 20(2), 159–161. https://doi.org/10.2108/
females of common carp (Cyprinus carpio L.) by a recessive mutation in a sex- zsj.20.159
determining gene. Journal of Heredity, 83(6), 431–434. https://doi.org/10.1093/ McCarthy, M. M., & Arnold, A. P. (2011). Reframing sexual differentiation of the brain.
oxfordjournals.jhered.a111246 Nature Neuroscience, 14(6), 677–683. https://doi.org/10.1038/nn.2834
Komen, J., Lodder, P., Huskens, F., Richter, C., & Huisman, E. (1989). Effects of oral Moles, G., Carrillo, M., Mañanós, E., Mylonas, C., & Zanuy, S. (2007). Temporal profile of
administration of 17α-methyltestosterone and 17β-estradiol on gonadal development brain and pituitary GnRHs, GnRH-R and gonadotropin mRNA expression and
in common carp, Cyprinus carpio L. Aquaculture, 78(3–4), 349–363. https://doi.org/ content during early development in European sea bass (Dicentrarchus labrax L.).
10.1016/0044-8486(89)90111-7 General and Comparative Endocrinology, 150(1), 75–86. https://doi.org/10.1016/j.
Koopman, P., Münsterberg, A., Capel, B., Vivian, N., & Lovell-Badge, R. (1990). ygcen.2006.07.012
Expression of a candidate sex-determining gene during mouse testis differentiation.
Nature, 348(6300), 450–452. https://doi.org/10.1038/348450a0

536
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

Mommsen, T. P., Vijayan, M. M., & Moon, T. W. (1999). Cortisol in teleosts: Dynamics, Piferrer, F. (2001). Endocrine sex control strategies for the feminization of teleost fish.
mechanisms of action, and metabolic regulation. Reviews in Fish Biology and Fisheries, Reproductive Biotechnology in Finfish Aquaculture, 229–281. https://doi.org/10.1016/
9(3), 211–268. https://doi.org/10.1023/a:1008924418720 b978-0-444-50913-0.50014-x
Morrey, C. E., Nagahama, Y., & Grau, E. G. (2002). Terminal phase males stimulate Piferrer, F., Baker, I. J., & Donaldson, E. M. (1993). Effects of natural, synthetic,
ovarian function and inhibit sex change in the protogynous wrasse Thalassoma Aromatizable, and Nonaromatizable androgens in inducing male sex differentiation
duperrey. Zoological Science, 19(1), 103–109. https://doi.org/10.2108/zsj.19.103 in genotypic female Chinook salmon (Oncorhynchus tshawytscha). General and
Munday, P., Buston, P., & Warner, R. (2006). Diversity and flexibility of sex-change Comparative Endocrinology, 91(1), 59–65. https://doi.org/10.1006/gcen.1993.1104
strategies in animals. Trends in Ecology & Evolution, 21(2), 89–95. https://doi.org/ Piferrer, F., & Blázquez, M. (2005). Aromatase distribution and regulation in fish. Fish
10.1016/j.tree.2005.10.020 Physiology and Biochemistry, 31(2–3), 215–226. https://doi.org/10.1007/s10695-
Munday, P. L., Wilson White, J., & Warner, R. R. (2006). A social basis for the 006-0027-0
development of primary males in a sex-changing fish. Proceedings of the Royal Society Piferrer, F., & Donaldson, E. M. (1989). Gonadal differentiation in coho salmon,
B: Biological Sciences, 273(1603), 2845–2851. https://doi.org/10.1098/ Oncorhynchus kisutch, after a single treatment with androgen or estrogen at different
rspb.2006.3666 stages during ontogenesis. Aquaculture, 77(2–3), 251–262. https://doi.org/10.1016/
Murugananthkumar, R., Prathibha, Y., Senthilkumaran, B., Rajakumar, A., & Kagawa, H. 0044-8486(89)90207-x
(2017). In vivo induction of human chorionic gonadotropin by osmotic pump Piferrer, F., & Guiguen, Y. (2008). Fish Gonadogenesis. Part II: Molecular biology and
advances sexual maturation during pre-spawning phase in adult catfish. General and genomics of sex differentiation. Reviews in Fisheries Science, 16(sup1), 35–55. https://
Comparative Endocrinology, 251, 74–84. https://doi.org/10.1016/j. doi.org/10.1080/10641260802324644
ygcen.2016.09.015 Piferrer, F., Zanuy, S., Carrillo, M., Solar, I. I., Devlin, R. H., & Donaldson, E. M. (1994).
Myosho, T., Otake, H., Masuyama, H., Matsuda, M., Kuroki, Y., Fujiyama, A., et al. Brief treatment with an aromatase inhibitor during sex differentiation causes
(2012). Tracing the emergence of a novel sex-determining gene in medaka, Oryzias chromosomally female salmon to develop as normal, functional males. Journal of
luzonensis. Genetics, 191(1), 163–170. https://doi.org/10.1534/genetics.111.137497 Experimental Zoology, 270(3), 255–262. https://doi.org/10.1002/jez.1402700304
Nagahama, Y. (2002). Endocrine regulation of gametogenesis in fish. International Pollock, M. S., Dubé, M. G., & Schryer, R. (2010). Investigating the link between pulp
Journal of Developmental Biology, 38(2), 217–229. mill effluent and endocrine disruption: Attempts to explain the presence of intersex
Nagahama, Y. (2005). Molecular mechanisms of sex determination and gonadal sex fish in the Wabigoon river, Ontario, Canada. Environmental Toxicology & Chemistry,
differentiation in fish. Fish Physiology and Biochemistry, 31(2–3), 105–109. https:// 29(4), 952–965. https://doi.org/10.1002/etc.118
doi.org/10.1007/s10695-006-7590-2 Poonlaphdecha, S., Pepey, E., Canonne, M., De Verdal, H., Baroiller, J. F., & D’cotta, H.
Nagy, A., Bercsényi, M., & Csányi, V. (1981). Sex reversal in carp (Cyprinus carpio) by (2013). Temperature induced-masculinisation in the Nile tilapia causes rapid up-
oral administration of Methyltestosterone. Canadian Journal of Fisheries and Aquatic regulation of both dmrt1 and amh expressions. General and Comparative
Sciences, 38(6), 725–728. https://doi.org/10.1139/f81-096 Endocrinology, 193, 234–242. https://doi.org/10.1016/j.ygcen.2013.06.007
Nakamura, M. (1984). Effects of estradiol-17β on gonadal sex differentiation in two Powell, J. F., Zohar, Y., Elizur, A., Park, M., Fischer, W. H., Craig, A. G., et al. (1994).
species of salmonids, the masu salmon, Oncorhynchus masou, and the chum salmon. Three forms of gonadotropin-releasing hormone characterized from brains of one
O. keta. Aquaculture, 43(1–3), 83–90. https://doi.org/10.1016/0044-8486(84) species. Proceedings of the National Academy of Sciences, 91(25), 12081–12085.
90012-7 https://doi.org/10.1073/pnas.91.25.12081
Nakamura, M., Hourigan, T. F., Yamauchi, K., Nagahama, Y., & Grau, E. G. (1989). Raghuveer, K., & Senthilkumaran, B. (2009). Identification of multiple dmrt1s in catfish:
Histological and ultrastructural evidence for the role of gonadal steroid hormones in Localization, dimorphic expression pattern, changes during testicular cycle and after
sex change in the protogynous wrasse Thalassoma duperrey. Environmental Biology of methyltestosterone treatment. Journal of Molecular Endocrinology, 42(5), 437–448.
Fishes, 24(2), 117–136. https://doi.org/10.1007/bf00001282 https://doi.org/10.1677/jme-09-0011
Nakamura, M., Kobayashi, T., Chang, X., & Nagahama, Y. (1998). Gonadal sex Raghuveer, K., Senthilkumaran, B., Sudhakumari, C., Sridevi, P., Rajakumar, A.,
differentiation in teleost fish. Journal of Experimental Zoology, 281(5), 362–372. Singh, R., et al. (2011). Dimorphic expression of various transcription factor and
https://doi.org/10.1002/(sici)1097-010x(19980801)281:5<362::aid-jez3>3.0.co; Steroidogenic enzyme genes during gonadal ontogeny in the air-breathing catfish,
2-m Clarias gariepinus. Sexual Development, 5(4), 213–223. https://doi.org/10.1159/
Nakamura, M., Kobayashi, Y., Miura, S., Alam, M. A., & Bhandari, R. K. (2005). Sex 000328823
change in coral reef fish. Fish Physiology and Biochemistry, 31(2–3), 117–122. https:// Raghuveer, K., Sudhakumari, C., Senthilkumaran, B., Kagawa, H., Dutta-Gupta, A., &
doi.org/10.1007/s10695-006-7595-x Nagahama, Y. (2011). Gender differences in tryptophan hydroxylase-2 mRNA,
Nanda, I., Kondo, M., Hornung, U., Asakawa, S., Winkler, C., Shimizu, A., et al. (2002). serotonin, and 5-hydroxytryptophan levels in the brain of catfish, Clarias gariepinus,
A duplicated copy of DMRT1 in the sex-determining region of the Y chromosome of during sex differentiation. General and Comparative Endocrinology, 171(3), 94–104.
the medaka, Oryzias latipes. Proceedings of the National Academy of Sciences, 99(18), https://doi.org/10.1016/j.ygcen.2011.02.020
11778–11783. https://doi.org/10.1073/pnas.182314699 Rahman, M. S., Khan, I. A., & Thomas, P. (2011). Tryptophan hydroxylase: A target for
Narnaware, Y. K., Peyon, P. P., Lin, X., & Peter, R. E. (2000). Regulation of food intake by neuroendocrine disruption. Journal of Toxicology and Environmental Health, Part B, 14
neuropeptide Y in goldfish. American Journal of Physiology - Regulatory, Integrative (5–7), 473–494. https://doi.org/10.1080/10937404.2011.578563
and Comparative Physiology, 279(3), R1025–R1034. https://doi.org/10.1152/ Rahman, M. S., & Thomas, P. (2013). Interactive effects of hypoxia with estradiol-17β on
ajpregu.2000.279.3.r1025 tryptophan hydroxylase activity and serotonin levels in the Atlantic croaker
Navarro-Martín, L., Viñas, J., Ribas, L., Díaz, N., Gutiérrez, A., Di Croce, L., et al. (2011). hypothalamus. General and Comparative Endocrinology, 192, 71–76. https://doi.org/
DNA methylation of the gonadal aromatase (cyp19a) promoter is involved in 10.1016/j.ygcen.2013.03.001
temperature-dependent sex ratio shifts in the European sea bass. PLoS Genetics, 7 Rajakumar, A., & Senthilkumaran, B. (2020). Steroidogenesis and its regulation in
(12), Article e1002447. https://doi.org/10.1371/journal.pgen.1002447 teleost-a review. Fish Physiology and Biochemistry, 46(3), 803–818. https://doi.org/
Pandian, T. A., & Koteeswaran, R. (1998). Ploidy induction and sex control in fish. 10.1007/s10695-019-00752-0
Hydrobiologia, 384(1), 167–243. https://doi.org/10.1023/A:1003332526659 Rajendiran, P., Jaafar, F., Kar, S., Sudhakumari, C., Senthilkumaran, B., & Parhar, I. S.
Pandian, T. J., & Sheela, S. G. (1995). Hormonal induction of sex reversal in fish. (2021). Sex determination and differentiation in teleost: Roles of genetics,
Aquaculture, 138(1–4), 1–22. https://doi.org/10.1016/0044-8486(95)01075-0 environment, and brain. Biology, 10(10), 973. https://doi.org/10.3390/
Pandian, T. J., & Varadaraj, K. (1990). Development of monosex female Oreochromis biology10100973
mossambicus broodstock by integrating gynogenetic technique with endocrine sex Römer, U., & Beisenherz, W. (1996). Environmental determination of sex in
reversal. Journal of Experimental Zoology, 255(1), 88–96. https://doi.org/10.1002/ Apistogrammai (Cichlidae) and two other freshwater fishes (Teleostei). Journal of
jez.1402550112 Fish Biology, 48(4), 714–725. https://doi.org/10.1006/jfbi.1996.0072
Pandolfi, M., Parhar, I., Ravaglia, M., Meijide, F., Maggese, C., & Paz, D. (2002). Ross, R. M. (1990). The evolution of sex-change mechanisms in fishes. Environmental
Ontogeny and distribution of gonadotropin-releasing hormone (GnRH) neuronal Biology of Fishes, 29(2), 81–93. https://doi.org/10.1007/bf00005025
systems in the brain of the cichlid fish Cichlasoma dimerus. Anatomy and Embryology, Rubin, D. A. (1985). Effect of pH on sex ratio in cichlids and a Poecilliid (Teleostei).
205(4), 271–281. https://doi.org/10.1007/s00429-002-0253-x Copeia, 1985(1), 233. https://doi.org/10.2307/1444818
Pavlidis, M., Koumoundouros, G., Sterioti, A., Somarakis, S., Divanach, P., & Kentouri, M. Ruksana, S., Pandit, N. P., & Nakamura, M. (2010). Efficacy of exemestane, a new
(2000). Evidence of temperature-dependent sex determination in the European sea generation of aromatase inhibitor, on sex differentiation in a gonochoristic fish.
bass (Dicentrarchus labrax L.). Journal of Experimental Zoology, 287(3), 225–232. Comparative Biochemistry and Physiology - Part C: Toxicology & Pharmacology, 152(1),
https://doi.org/10.1002/1097-010x(20000801)287:3<225::aid-jez4>3.0.co;2-d 69–74. https://doi.org/10.1016/j.cbpc.2010.02.014
Peng, C., Chang, J. P., Yu, K. L., Wong, A. O., Van Goor, F., Peter, R. E., et al. (1993). Sadovy, Y., & Colin, P. L. (1995). Sexual development and sexuality in the Nassau
Neuropeptide-Y stimulates growth hormone and gonadotropin-II secretion in the grouper. Journal of Fish Biology, 46(6), 961–976. https://doi.org/10.1111/j.1095-
goldfish pituitary: Involvement of both presynaptic and pituitary cell actions. 8649.1995.tb01401.x
Endocrinology, 132(4), 1820–1829. https://doi.org/10.1210/endo.132.4.8462479 Saito, D., Morinaga, C., Aoki, Y., Nakamura, S., Mitani, H., Furutani-Seiki, M., et al.
Peng, C., Gallin, W., Peter, R. E., Blomqvist, A. G., & Larhammar, D. (1994). (2007). Proliferation of germ cells during gonadal sex differentiation in medaka:
Neuropeptide-Y gene expression in the goldfish brain: Distribution and regulation by Insights from germ cell-depleted mutant zenzai. Developmental Biology, 310(2),
ovarian steroids. Endocrinology, 134(3), 1095–1103. https://doi.org/10.1210/ 280–290. https://doi.org/10.1016/j.ydbio.2007.07.039
endo.134.3.8119148 Saito, D., & Tanaka, M. (2009). Comparative aspects of gonadal sex differentiation in
Peng, C., Trudeau, V. L., & Peter, R. E. (1993). Seasonal variation of neuropeptide Y medaka: A conserved role of developing oocytes in sexual canalization. Sexual
actions on growth hormone and gonadotropin-ll secretion in the goldfish: Effects of Development, 3(2–3), 99–107. https://doi.org/10.1159/000223075
sex steroids. Journal of Neuroendocrinology, 5(3), 273–280. https://doi.org/10.1111/ Santi, S., Gennotte, V., Toguyeni, A., Mélard, C., Antoine, N., & Rougeot, C. (2016).
j.1365-2826.1993.tb00483.x Thermosensitivity of the sex differentiation process in the African catfish, Clarias
Peter, R. E., Trudeau, V. L., & Sloley, B. D. (1991). Brain regulation of reproduction in gariepinus: Determination of the thermosensitive period. Aquaculture, 455, 73–80.
teleosts. Bulletin of the Institute of Zoology Academia Sinica, 16, 89–118. https://doi.org/10.1016/j.aquaculture.2016.01.009

537
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

Sato, T., Endo, T., Yamahira, K., Hamaguchi, S., & Sakaizumi, M. (2005). Induction of Neuropeptide-Y during early brain development and gonadal recrudescence in the
female-to-male sex reversal by high temperature treatment in medaka, Oryzias catfish, Clarias gariepinus. General and Comparative Endocrinology, 251, 54–65.
latipes. Zoological Science, 22(9), 985–988. https://doi.org/10.2108/zsj.22.985 https://doi.org/10.1016/j.ygcen.2017.03.006
Scholz, S., Rösler, S., Schäffer, M., Hornung, U., Schartl, M., & Gutzeit, H. O. (2003). Sudhakumari, C., Senthilkumaran, B., Raghuveer, K., Wang, D., Kobayashi, T.,
Hormonal induction and stability of Monosex populations in the medaka (Oryzias Kagawa, H., et al. (2010). Dimorphic expression of tryptophan hydroxylase in the
latipes): Expression of sex-specific marker genes. Biology of Reproduction, 69(2), brain of XX and XY Nile tilapia during early development. General and Comparative
673–678. https://doi.org/10.1095/biolreprod.103.016873 Endocrinology, 166(2), 320–329. https://doi.org/10.1016/j.ygcen.2009.11.009
Schreck, C. B. (2010). Stress and fish reproduction: The roles of allostasis and hormesis. Sullivan, J. A., & Schultz, R. J. (1986). Genetic and environmental basis of variable sex
General and Comparative Endocrinology, 165(3), 549–556. https://doi.org/10.1016/j. ratios in laboratory strains of Poeciliopsis lucida. Evolution, 40(1), 152. https://doi.
ygcen.2009.07.004 org/10.2307/2408612
Selim, K. M., Shinomiya, A., Otake, H., Hamaguchi, S., & Sakaizumi, M. (2009). Effects of Sun, L., Jiang, X., Xie, Q., Yuan, J., Huang, B., Tao, W., et al. (2014). Transdifferentiation
high temperature on sex differentiation and germ cell population in medaka. Oryzias of differentiated ovary into functional testis by long-term treatment of Aromatase
latipes. Aquaculture, 289(3–4), 340–349. https://doi.org/10.1016/j. inhibitor in Nile tilapia. Endocrinology, 155(4), 1476–1488. https://doi.org/
aquaculture.2008.12.019 10.1210/en.2013-1959
Senthilkumaran, O., Gen, O., & Kagawa. (1999). Distribution and seasonal variations in Sunobe, T., & Nakazono, A. (1993). Sex change in both directions by alteration of social
levels of three native GnRHs in the brain and pituitary of perciform fish. Journal of dominance in Trimma okinawae (Pisces: Gobiidae). Ethology, 94(4), 339–345.
Neuroendocrinology, 11(3), 181–186. https://doi.org/10.1046/j.1365- https://doi.org/10.1111/j.1439-0310.1993.tb00450.x
2826.1999.00304.x Sun, L., Zha, J., Spear, P. A., & Wang, Z. (2007). Toxicity of the aromatase inhibitor
Senthilkumaran, B., & Joy, K. (1995). Changes in hypothalamic catecholamines, letrozole to Japanese medaka (Oryzias latipes) eggs, larvae and breeding adults.
dopamine-β-hydroxylase, and Phenylethanolamine-N-methyltransferase in the Comparative Biochemistry and Physiology - Part C: Toxicology & Pharmacology, 145(4),
catfish Heteropneustes fossilis in relation to season, raised photoperiod and 533–541. https://doi.org/10.1016/j.cbpc.2007.01.017
temperature, ovariectomy, and estradiol-17β replacement. General and Comparative Suzuki, A., Nakamoto, M., Kato, Y., & Shibata, N. (2005). Effects of estradiol-17β on germ
Endocrinology, 97(1), 121–134. https://doi.org/10.1006/gcen.1995.1012 cell proliferation and DMY Expression during early sexual differentiation of the
Senthilkumaran, B., & Joy, K. (1996). Effects of administration of some monoamine- medaka Oryzias latipes. Zoological Science, 22(7), 791–796. https://doi.org/10.2108/
synthesis blockers and precursors on ovariectomy-induced rise in plasma zsj.22.791
gonadotropin II in the Catfish Heteropneustes fossilis. General and Comparative Swapna, I., Sudhakumari, C. C., Sakai, F., Sreenivasulu, G., Kobayashi, T., Kagawa, H.,
Endocrinology, 101(2), 220–226. https://doi.org/10.1006/gcen.1996.0024 et al. (2008). Seabream GnRH immunoreactivity in brain and pituitary of XX and XY
Senthilkumaran, B., & Kar, S. (2021). Advances in reproductive endocrinology and Nile tilapia, Oreochromis niloticus during early development. Journal of Experimental
neuroendocrine research using catfish models. Cells, 10(11), 2807. https://doi.org/ Zoology Part A: Ecological Genetics and Physiology, 309A(7), 419–426. https://doi.
10.3390/cells10112807 org/10.1002/jez.467
Senthilkumaran, B., Okuzawa, K., Gen, K., & Kagawa, H. (2001). Effects of serotonin, Takatsu, K., Miyaoku, K., Roy, S. R., Murono, Y., Sago, T., Itagaki, H., et al. (2013).
GABA and neuropeptide Y on Seabream gonadotropin releasing hormone release in Induction of female-to-male sex change in adult zebrafish by aromatase inhibitor
vitro from preoptic-anterior hypothalamus and pituitary of red Seabream, Pagrus treatment. Scientific Reports, 3(1). https://doi.org/10.1038/srep03400
major. Journal of Neuroendocrinology, 13(5), 395–400. https://doi.org/10.1046/ Tan-Fermin, J. D., Garcia, L. M. B., & Castillo, A. R. (1994). Induction of sex inversion in
j.1365-2826.2001.00645.x juvenile grouper, Epinephelus suillus, (Valenciennes) by injections of 17α-
Senthilkumaran, B., Sudhakumari, C., Mamta, S., Raghuveer, K., Swapna, I., & methyltestosterone. Japanese Journal of Ichthyology, 40(4), 413–420. https://doi.org/
Murugananthkumar, R. (2015). Brain sex differentiation” in teleosts: Emerging 10.11369/jji1950.40.413
concepts with potential biomarkers. General and Comparative Endocrinology, 220, Tanaka, M., Saito, D., Morinaga, C., & Kurokawa, H. (2008). Cross talk between germ
33–40. https://doi.org/10.1016/j.ygcen.2015.06.003 cells and gonadal somatic cells is critical for sex differentiation of the gonads in the
Shang, E. H., & Wu, R. S. (2004). Aquatic hypoxia is a teratogen and affects fish teleost fish, medaka (Oryzias latipes). Development Growth and Differentiation, 50(4),
embryonic development. Environmental Science & Technology, 38(18), 4763–4767. 273–278. https://doi.org/10.1111/j.1440-169x.2008.01015.x
https://doi.org/10.1021/es0496423 Tenugu, S., Pranoty, A., Mamta, S., & Senthilkumaran, B. (2021). Development and
Shang, E. H., Yu, R. M., & Wu, R. S. (2006). Hypoxia affects sex differentiation and organisation of gonadal steroidogenesis in BONY fishes - a review. Aquaculture and
development, leading to a male-dominated population in zebrafish (Danio rerio). Fisheries, 6(3), 223–246. https://doi.org/10.1016/j.aaf.2020.09.004
Environmental Science & Technology, 40(9), 3118–3122. https://doi.org/10.1021/ Thomas, P., & Rahman, M. S. (2009). Chronic hypoxia impairs gamete maturation in
es0522579 Atlantic croaker induced by progestins through nongenomic mechanisms resulting in
Shapiro, D. Y. (1990). Sex-changing fish as a manipulable system for the study of the reduced reproductive success. Environmental science & technology, 43(11),
determination, differentiation, and stability of sex in vertebrates. Journal of 4175–4180. https://doi.org/10.1021/es9000399
Experimental Zoology, 256(S4), 132–136. https://doi.org/10.1002/jez.1402560423 Thomas, P., Rahman, M. S., Kummer, J. A., & Lawson, S. (2006). Reproductive endocrine
Shinoda, T., Miranda, L. A., Okuma, K., Hattori, R. S., Fernandino, J. I., Yoshizaki, G., dysfunction in Atlantic croaker exposed to hypoxia. Marine Environmental Research,
et al. (2010). Molecular cloning and expression analysis of Fshr and Lhr in relation to 62, S249–S252. https://doi.org/10.1016/j.marenvres.2006.04.031
Fshβ and Lhβ subunits during the period of temperature-dependent sex Trudeau, V. (1997). Neuroendocrine regulation of gonadotrophin II release and gonadal
determination in pejerrey Odontesthes bonariensis. Molecular Reproduction and growth in the goldfish, Carassius auratus. Reviews of Reproduction, 2(1), 55–68.
Development, 77(6), 521–532. https://doi.org/10.1002/mrd.21179 https://doi.org/10.1530/ror.0.0020055
Silverstein, J. T., & Plisetskaya, E. M. (2000). The effects of NPY and insulin on food Trupp, M., Raynoschek, C., Belluardo, N., & Ibáñez, C. F. (1998). Multiple GPI-anchored
intake regulation in fish. American Zoologist, 40(2), 296–308. https://doi.org/ receptors control GDNF-dependent and independent activation of the C-ret receptor
10.1093/icb/40.2.296 tyrosine kinase. Molecular and Cellular Neuroscience, 11(1–2), 47–63. https://doi.
Sinclair, A. H., Berta, P., Palmer, M. S., Hawkins, J. R., Griffiths, B. L., Smith, M. J., et al. org/10.1006/mcne.1998.0667
(1990). A gene from the human sex-determining region encodes a protein with Tsai, C., Chang, S., Wang, L., & Chao, T. (2003). Temperature influences the ontogenetic
homology to a conserved DNA-binding motif. Nature, 346(6281), 240–244. https:// expression of Aromatase and oestrogen receptor mRNA in the developing tilapia
doi.org/10.1038/346240a0 (Oreochromis mossambicus) brain. Journal of Neuroendocrinology, 15(1), 97–102.
Soga, T., Ogawa, S., Millar, R. P., Sakuma, Y., & Parhar, I. S. (2005). Localization of the https://doi.org/10.1046/j.1365-2826.2003.00950.x
three GnRH types and GnRH receptors in the brain of a cichlid fish: Insights into Tsai, C., & Wang, L. (1999). Effects of gonadal steroids on the serotonin synthesis and
their neuroendocrine and neuromodulator functions. The Journal of Comparative metabolism in the early developing tilapia brain. Neuroscience Letters, 264(1–3),
Neurology, 487(1), 28–41. https://doi.org/10.1002/cne.20519 45–48. https://doi.org/10.1016/s0304-3940(99)00160-3
Solar, I. I., Donaldson, E. M., & Hunter, G. A. (1984). Optimization of treatment regimes Tsai, C., Wang, L., Chang, C., & Kao, C. (2001). Effects of gonadal steroids on brain
for controlled sex differentiation and sterilization in wild rainbow trout (Salmo Serotonergic and Aromatase activity during the critical period of sexual
gairdneri Richardson) by oral administration of 17α-methyltestosterone. Aquaculture, differentiation in tilapia, Oreochromis mossambicus. Journal of Neuroendocrinology, 12
42(2), 129–139. https://doi.org/10.1016/0044-8486(84)90360-0 (9), 894–898. https://doi.org/10.1046/j.1365-2826.2000.00536.x
Somoza, G. M., Miranda, L. A., Guilgur, L. G., & Strobl-Mazzulla, P. H. (2006). Tsai, C., Wang, L., & Fang, L. (2001). Estradiol and para-chlorophenylalanine
Characterization of the brain-pituitary axis in pejerrey Odontesthes bonariensis. Downregulate the expression of brain Aromatase and estrogen receptor-α mRNA
Biocell, 30(1), 89–95. during the critical period of feminization in tilapia (Oreochromis mossambicus).
Strobl-Mazzulla, P. H., Lethimonier, C., Gueguen, M. M., Karube, M., Fernandino, J. I., Neuroendocrinology, 74(5), 325–334. https://doi.org/10.1159/000054699
Yoshizaki, G., et al. (2008). Brain aromatase (Cyp19A2) and estrogen receptors, in Tuan, P. A., Little, D. C., & Mair, G. C. (1998). Genotypic effects on comparative growth
larvae and adult pejerrey fish Odontesthes bonariensis: Neuroanatomical and performance of all-male tilapia Oreochromis niloticus (L.). Aquaculture, 159(3–4),
functional relations. General and Comparative Endocrinology, 158(2), 191–201. 293–302. https://doi.org/10.1016/s0044-8486(97)00189-0
https://doi.org/10.1016/j.ygcen.2008.07.006 Tuan, P. A., Mair, G. C., Little, D. C., & Beardmore, J. A. (1999). Sex determination and
Strüssmann, C. A., Calsina Cota, J. C., Phonlor, G., Higuchi, H., & Takashima, F. (1996). the feasibility of genetically male tilapia production in the Thai-chitralada strain of
Temperature effects on sex differentiation of two south American atherinids, Oreochromis niloticus (L.). Aquaculture, 173(1–4), 257–269. https://doi.org/10.1016/
Odontesthes argentinensis and Patagonina hatcheri. Environmental Biology of Fishes, 47 s0044-8486(98)00450-5
(2), 143–154. https://doi.org/10.1007/bf00005037 Tzchori, I., Degani, G., Hurvitz, A., & Moav, B. (2004). Cloning and developmental
Strüssmann, C. A., Kitahara, A., & Amashita, M. (2008). Role of apoptosis in temperature- expression of the cytochrome P450 aromatase gene (CYP19) in the European eel
dependent sex determination of pejerrey Odontesthes bonariensis. Cybium, 32(2), (Anguilla anguilla). General and Comparative Endocrinology, 138(3), 271–280. https://
77–79. doi.org/10.1016/j.ygcen.2004.06.007
Sudhakumari, C., Anitha, A., Murugananthkumar, R., Tiwari, D. K., Bhasker, D., Uchida, D., Yamashita, M., Kitano, T., & Iguchi, T. (2002). Oocyte apoptosis during the
Senthilkumaran, B., et al. (2017). Cloning, localization and differential expression of transition from ovary-like tissue to testes during sex differentiation of juvenile

538
S. Tenugu and B. Senthilkumaran Aquaculture and Fisheries 7 (2022) 525–539

zebrafish. Journal of Experimental Biology, 205(6), 711–718. https://doi.org/ Yamamoto, T. (1958). Artificial induction of functional sex-reversal in genotypic females
10.1242/jeb.205.6.711 of the medaka (Oryzias latipes). Journal of Experimental Zoology, 137(2), 227–263.
Uchida, D., Yamashita, M., Kitano, T., & Iguchi, T. (2004). An aromatase inhibitor or https://doi.org/10.1002/jez.1401370203
high water temperature induce oocyte apoptosis and depletion of P450 aromatase Yamamoto, T. (1969). 3 sex differentiation. Fish Physiology, 117–175. https://doi.org/
activity in the gonads of genetic female zebrafish during sex-reversal. Comparative 10.1016/s1546-5098(08)60113-2
Biochemistry and Physiology Part A: Molecular & Integrative Physiology, 137(1), 11–20. Yamamoto, E. (1999). Studies on sex-manipulation and production of cloned populations
https://doi.org/10.1016/s1095-6433(03)00178-8 in hirame, Paralichthys olivaceus (Temminck et schlegel). Aquaculture, 173(1–4),
Vizziano-Cantonnet, D., Anglade, I., Pellegrini, E., Gueguen, M., Fostier, A., Guiguen, Y., 235–246. https://doi.org/10.1016/s0044-8486(98)00448-7
et al. (2011). Sexual dimorphism in the brain aromatase expression and activity, and Yang, Y., Liu, Q., Xiao, Y., Xu, S., Wang, X., Yang, J., et al. (2019). High temperature
in the central expression of other steroidogenic enzymes during the period of sex increases the gsdf expression in masculinization of genetically female Japanese
differentiation in monosex rainbow trout populations. General and Comparative flounder (Paralichthys olivaceus). General and Comparative Endocrinology, 274, 17–25.
Endocrinology, 170(2), 346–355. https://doi.org/10.1016/j.ygcen.2010.10.009 https://doi.org/10.1016/j.ygcen.2018.12.012
Volff, J. N. (2002). Sex determination in fish. Genome Biology, 3(9), 1–4. https://doi.org/ Yano, A., Guyomard, R., Nicol, B., Jouanno, E., Quillet, E., Klopp, C., et al. (2012). An
10.1186/gb-2002-3-9-reports0052 immune-related gene evolved into the master sex-determining gene in rainbow trout,
Volkoff, H., Canosa, L., Unniappan, S., Cerdá-Reverter, J., Bernier, N., Kelly, S., et al. Oncorhynchus mykiss. Current Biology, 22(15), 1423–1428. https://doi.org/10.1016/
(2005). Neuropeptides and the control of food intake in fish. General and Comparative j.cub.2012.05.045
Endocrinology, 142(1–2), 3–19. https://doi.org/10.1016/j.ygcen.2004.11.001 Yaron, Z., Gur, G., Melamed, P., Rosenfeld, H., Elizur, A., & Levavi-Sivan, B. (2003).
Wang, D., Kobayashi, T., Zhou, L., Paul-Prasanth, B., Ijiri, S., Sakai, F., et al. (2007). Regulation of fish gonadotropins. International Review of Cytology, 131–185. https://
Foxl2 up-regulates Aromatase gene transcription in a female-specific Manner by doi.org/10.1016/s0074-7696(05)25004-0
binding to the promoter as well as interacting with Ad4 binding protein/ Yeh, S., Kuo, C., Ting, Y., & Chang, C. (2003). The effects of exogenous androgens on
Steroidogenic factor 1. Molecular Endocrinology, 21(3), 712–725. https://doi.org/ ovarian development and sex change in female orange-spotted protogynous grouper.
10.1210/me.2006-0248 Epinephelus coioides. Aquaculture, 218(1–4), 729–739. https://doi.org/10.1016/
Wang, D., Zhou, L., Kobayashi, T., Matsuda, M., Shibata, Y., Sakai, F., et al. (2010). s0044-8486(02)00565-3
Doublesex- and mab-3-Related transcription factor-1 repression of aromatase Yeung, W., & Chan, S. (1987). The plasma sex steroid profiles in the freshwater, sex-
transcription, a possible mechanism favoring the male pathway in tilapia. reversing teleost fish, Monopterus albus (Zuiew). General and Comparative
Endocrinology, 151(3), 1331–1340. https://doi.org/10.1210/en.2009-0999 Endocrinology, 65(2), 233–242. https://doi.org/10.1016/0016-6480(87)90171-7
Warner, R. R. (1988). Sex change in fishes: Hypotheses, evidence, and objections. Yeung, W., Chen, H., & Chan, S. (1993). The in vitro metabolism of radioactive
Environmental Biology of Fishes, 22(2), 81–90. https://doi.org/10.1007/bf00001539 androstenedione and testosterone by the gonads of the protogynous Monopterus albus
Wu, R. S., Zhou, B. S., Randall, D. J., Woo, N. Y., & Lam, P. K. (2003). Aquatic hypoxia is at different sexual phases: A time-course and seasonal study. General and Comparative
an endocrine disruptor and impairs fish reproduction. Environmental Science & Endocrinology, 89(3), 313–322. https://doi.org/10.1006/gcen.1993.1039
Technology, 37(6), 1137–1141. https://doi.org/10.1021/es0258327 Yu, R. M., Chu, D. L., Tan, T., Li, V. W., Chan, A. K., Giesy, J. P., et al. (2012). Leptin-
Yamaguchi, T., & Kitano, T. (2012). High temperature induces cyp26b1 mRNA mediated modulation of Steroidogenic gene expression in hypoxic Zebrafish
expression and delays meiotic initiation of germ cells by increasing cortisol levels embryos: Implications for the disruption of sex steroids. Environmental Science &
during gonadal sex differentiation in Japanese flounder. Biochemical and Biophysical Technology, 46(16), 9112–9119. https://doi.org/10.1021/es301758c
Research Communications, 419(2), 287–292. https://doi.org/10.1016/j. Zhou, L., & Gui, J. (2010). Molecular mechanisms underlying sex change in
bbrc.2012.02.012 hermaphroditic groupers. Fish Physiology and Biochemistry, 36(2), 181–193. https://
Yamaguchi, T., Yoshinaga, N., Yazawa, T., Gen, K., & Kitano, T. (2010). Cortisol is doi.org/10.1007/s10695-008-9219-0
involved in temperature-dependent sex determination in the Japanese flounder. Zohar, Y., Munoz-Cueto, J. A., Elizur, A., & Kah, O. (2010). Neuroendocrinology of
Endocrinology, 151(8), 3900–3908. https://doi.org/10.1210/en.2010-0228 reproduction in teleost fish. General and Comparative Endocrinology, 165(3),
Yamamoto, T. (1953). Artificially induced sex-reversal in genotypic males of the medaka 438–455. https://doi.org/10.1016/j.ygcen.2009.04.017
(Oryzias latipes). Journal of Experimental Zoology, 123(3), 571–594. https://doi.org/
10.1002/jez.1401230309

539

You might also like