Next Article in Journal
Colchicine in Athero-Thrombosis: Molecular Mechanisms and Clinical Evidence
Next Article in Special Issue
17β-Estradiol Induced Sex Reversal and Gonadal Transcriptome Analysis in the Oriental River Prawn (Macrobrachium nipponense): Mechanisms, Pathways, and Potential Harm
Previous Article in Journal
Influence of Perinatal Factors on Gene Expression of IAPs Family and Main Factors of Pluripotency: OCT4 and SOX2 in Human Breast Milk Stem Cells—A Preliminary Report
Previous Article in Special Issue
Soy Isoflavones Induce Feminization of Japanese Eel (Anguilla japonica)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Amh/Amhr2 Signaling Causes Masculinization by Inhibiting Estrogen Synthesis during Gonadal Sex Differentiation in Japanese Flounder (Paralichthys olivaceus)

by
Toshiya Yamaguchi
1,* and
Takeshi Kitano
2
1
Nansei Field Station, National Research and Development Agency, Japan Fisheries Research and Education Agency, Minamiise, Mie 516-0193, Japan
2
Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2480; https://doi.org/10.3390/ijms24032480
Submission received: 30 December 2022 / Revised: 24 January 2023 / Accepted: 25 January 2023 / Published: 27 January 2023
(This article belongs to the Special Issue Sex Hormones and Sex Differentiation)

Abstract

:
The anti-Müllerian hormone (Amh) is a protein belonging to the TGF-β superfamily, the function of which has been considered important for male sex differentiation in vertebrates. The Japanese flounder (Paralichthys olivaceus) is a teleost fish that has an XX/XY sex determination system and temperature-dependent sex determination. In this species, amh expression is up-regulated in genetic males and in temperature-induced masculinization during the sex differentiation period. However, to the best of our knowledge, no reports on the Amh receptor (Amhr2) in flounder have been published, and the details of Amh signaling remain unclear. In this study, we produced amhr2-deficient mutants using the CRISPR/Cas9 system and analyzed the gonadal phenotypes and sex-related genes. The results revealed that the gonads of genetically male amhr2 mutants featured typical ovaries, and the sex differentiation-related genes showed a female expression pattern. Thus, the loss of Amhr2 function causes male-to-female sex reversal in Japanese flounder. Moreover, the treatment of genetically male amhr2 mutants with an aromatase inhibitor fadrozole, which inhibits estrogen synthesis, resulted in testicular formation. These results strongly suggest that Amh/Amhr2 signaling causes masculinization by inhibiting estrogen synthesis during gonadal sex differentiation in the flounder.

1. Introduction

Most teleosts exhibit gonochorism, along with the presence of regular males and females such as other vertebrates. Meanwhile, some teleosts are known to have diverse reproductive systems because sex is determined not only by genetic factors, but also by environmental factors including temperature, pH, density, and oxygen concentration [1]. These factors act as master switches that can direct bipotential gonadal primordia to become ovary or testis by activating differentiation pathways. Thus, teleost sex determination can involve genetic sex determination (GSD), environmental sex determination (ESD), or a combination of both of them.
The Japanese flounder (Paralichthys olivaceus), a marine culture fish that is economically important for aquaculture in Asia, is a teleost fish that has an XX/XY sex determination system [2]. However, XX flounder can be completely sex-reversed to become phenotypic males by rearing the larvae at high or low water temperatures early in the sex differentiation period at 50–60 days after hatching (dah) [2,3,4]. Therefore, the Japanese flounder exhibits both GSD and temperature-dependent sex determination (TSD) as a kind of ESD. This makes it an excellent model to study the molecular mechanisms determining the relationship between GSD and TSD [5,6]. In our previous study on TSD, the treatment with an aromatase inhibitor (fadrozole) or an anti-estrogen agent (tamoxifen) and a high temperature induced the masculinization of XX flounder and suppressed the mRNA expression of ovarian aromatase (cyp19a1), a steroidogenic enzyme that converts androgen into estrogen [7,8]. Furthermore, estrogen (17β-estradiol) administration completely inhibited the masculinization due to a high temperature [9]. It is suggested that the suppression of cyp19a1 mRNA expression and the inhibition of estrogen biosynthesis may ultimately trigger the gonadal masculinization of XX flounder induced by a high temperature.
The transcript of the cyp19a1 gene is reported to be expressed in a female-specific manner during sexual differentiation in various teleosts including flounder [6,8]. We previously investigated the molecular mechanism of the regulation of cyp19a1 expression, and we reported that the transcription factor forkhead box protein l2 (Foxl2) and follicle-stimulating hormone (FSH) signal may positively regulate the expression of cyp19a1 [10,11]. The foxl2 mRNA is specifically expressed in females in the gonad during the sex differentiation period, which is similar to cyp19a1, and in vitro experiments have shown that Foxl2 protein directly binds to the transcriptional regulatory promoter present in the 5’ upstream region of the cyp19a1 gene, and it positively regulates the promoter region. FSH is secreted from the pituitary gland, and signals through receptors (Fshr) present in gonadal somatic cells [12,13]. In other teleosts, the transcription factor foxl2 is also expressed in a female-specific manner in the gonads [14]. Thus, it is thought that Foxl2 and FSH signals are present upstream of the cyp19a1 gene regulatory cascade and induce feminization in flounder during sexual differentiation. Meanwhile, we previously reported that cortisol, a stress response hormone, is involved in the molecular mechanism of gonad masculinization during treatments with high temperatures [11]. It was shown that cortisol, the secretion of which increases in response to stress, inhibited the transcriptional regulation of the cyp19a1 gene via the FSH signal in Japanese flounder. Cortisol has also been reported to induce masculinization in medaka (Oryzias latipes) [15] and pejerrey (Odontesthes hatcheri) [16]. Thus, the mechanism of TSD is explained by the expression of the estrogen biosynthetic cyp19a1 gene and the antagonism of this by the stress response hormone cortisol. However, the mechanism by which differentiation into male gonads occurs in flounder with the XX/XY sex determination system has not been elucidated.
In Japanese flounder, the anti-Müllerian hormone (amh) mRNA is reported to be expressed in a male-specific manner, which is in contrast to cyp19a1, during sexual differentiation [17]. Amh, which is also known as Müllerian inhibiting substance (Mis), is a protein belonging to the TGF-β superfamily [18]. In mammals, a distinctive function of Amh is the induction of Müllerian duct regression during early ovarian differentiation [19,20]. The expression of Amh is restricted to the somatic cells in contact with germ cells, namely, Sertoli cells in the testis and granulosa cells in the ovary [21,22]. Amh primarily functions through the anti-Müllerian hormone receptor type 2 (Amhr2) and via a single-transmembrane domain and a serine/threonine kinase domain [23]. The expression of amhr2 is restricted to Sertoli cells in the testis and granulosa cells in the ovary.
Recently, Y-chromosome allele-specific amh (amhy) has been reported to be the master sex-determining gene in flounder [24]. A genomic analysis based on sex-specific SNP information revealed the presence of a Y-allele-specific deletion in the promoter region of the amh gene. A genotype-sex association analysis found a identify the association between phenotypic sex and genotype. Moreover, experiments involving the loss of function of amh produced using CRISPR/Cas9 resulted in the complete sex reversal of XY flounder, thus supporting the role of amhy as a sex-determining gene in flounder. The finding that amhy is the master sex-determining gene has enabled major progress in research on the molecular mechanisms of genetic sex determination in flounder. It has also become possible to determine whether the genetic sex is XX or XY without performing a mating test on Japanese flounder.
In this study, to investigate the molecular cascade of male gonadal differentiation in Japanese flounder, we generated amhr2 mutant flounder using the CRISPR/Cas9 system [25]. The system induced the biallelic mutation of the target gene, resulting in phenotypic knockout in the F0 generation by inducing the wide deletion using two CRISPR RNAs (crRNAs) for the target gene. We also performed a sex-reversal experiment using the aromatase inhibitor fadrozole to confirm the function of estrogen during gonadal differentiation in amh mutants. These experiments showed a functional role of the amhr2 gene in the genetic sex differentiation in Japanese flounder.

2. Results

2.1. Expression Analysis of amh and amhr2 in Gonads in XY and XX Flounder

We investigated the expression levels of amh and amhr2 transcripts using a quantitative real-time PCR from the sexually indifferent stage (50 dah) to the stage of clear differentiation in the testis or ovary (200 dah) and compared such expression levels between XY and XX flounder. The results showed that amh transcripts were detected in XY fish from 50 to 200 dah, whereas the level of transcripts in genetic females was very low throughout the whole period (Figure 1A). Meanwhile, the amhr2 transcript was detected in both of the genotypes from 50 to 200 dah, and the expression levels showed no difference between XY and XX fish during the same period (Figure 1B). The cellular localizations of amh and amhr2 transcripts were also investigated using in situ hybridization in the XY testis and XX ovary at 200 dah. The gonium cells with characteristic swollen round nuclei were found in both of the genotypes by using hematoxylin-eosin staining (Figure 1C). The localization of the amh transcripts showed positive signals in the testis but not in the ovary; the signals were observed in the presumptive Sertoli cells in contact with the gonium cells in the testis (Figure 1C). The positive signals of amhr2 were detected in both the testis and the ovary (Figure 1C). These signals were localized in presumptive Sertoli cells in testis as well as the amh transcript signals. In the ovary, the expression was detected in the somatic cells in cysts of the gonium cells. Thus, both amh and amhr2 were expressed in the Sertoli cells of XY gonads.

2.2. Generation of amhr2-Deficient Mutant Using CRISPR/Cas9 System in Japanese Flounder

To investigate the function of the amhr2 gene, we produced amhr2-deficient mutant using the CRISPR/Cas9 system. This system can achieve the highly efficient production of the biallelic knockout mutant in the F0 generation by inducing a wide deletion using two crRNAs for the target gene as described previously [25]. The crRNAs were designed in exons seven and nine of the amhr2 gene, containing a conserved serine/threonine kinase domain. This was expected to produce a deletion of approximately 710 bp in the amhr2 gene, disrupting the function of the Amhr2 protein that it encodes (Figure 2A). The two crRNAs were co-injected with tracrRNA and Cas9 proteins into the fertilized eggs, and the mutants were reared to 200 dah to investigate the mutant genotype. To determine the detailed mutant genotype, we carried out a sequencing analysis for the amhr2 mutants using the DNA extracted from the gonad. From the results, representative amplicon fragment sequences showed deletions of 714, 715, or 706 bp between the two crRNA target sequences and a few base mutations in both of the target sequences (Figure 2B). The rates of this wide deletion among the sequenced fragments were 15/16, 13/16, and 15/16 clones in the genetic males and 8/8 and 6/8 clones in the genetic females (Table 1). Mutations were also identified in all of the clones analyzed by sequencing. Thus, wide deletions were induced in amhr2 mutants by using the CRISPR/Cas9 system in Japanese flounder.

2.3. amhr2 Mutants Showed Phenotypic Sex Reversal from Male to Female

We observed the phenotypes of the amhr2 mutants compared with those of the XY and XX flounder using a histological analysis. The wild-type XY gonad was assumed to consist of gonium cells from the morphological features of the cells, while the XX gonad showed an ovarian morphology, including several primary oocytes that were densely stained with eosin and an ovarian cavity (Figure 3A). Thus, the wild-type flounder gonad was differentiated into typical ovaries or testes at 200 dah. We then observed the phenotypes of the amhr2 mutants and compared them with the wild-type gonads. Both XY and XX amhr2 mutants showed primary oocytes and an ovarian cavity that are similar to those in the control group, and no histological differences were found between them (Figure 3A). All the XY amhr2 mutants thus showed a female phenotype with normal ovaries despite them being XY fish (Table 2). This showed that the suppression of the amhr2 gene function in the XY fish induced the phenotypic sex reversal from male to female. To examine the expression pattern of sex-related genes in the gonads of amhr2 mutants, a quantitative real-time PCR analysis was used to confirm the expressions of amh, cyp19a1, foxl2, and fshr in the XY and XX control and amhr2 mutants at 200 dah. amh showed significantly higher expression in the XY control fish than they did in the XX controls, whereas the expression levels were very low in the XY mutant fish, as they were in the XX mutants (Figure 3B). Meanwhile, the expression of cyp19a1 was very low in XY control fish, but it was significantly elevated in XY mutant fish compared with that in the XY controls (Figure 3C). The transcription factor foxl2, which up-regulates cyp19a1, was also expressed at a very low level in the XY control fish, whereas the expression was significantly elevated to be similar to that of cyp19a1 in the XY mutants (Figure 3D). There was no significant difference in fshr expression levels among all of the experimental groups (Figure 3E). Thus, the expression patterns of amh, cyp19a1, and foxl2 in the XX mutants were similar to those in the XX control fish. The expression patterns of the sex differentiation-related genes in the amhr2 mutants were confirmed to be feminized from the results of the histological phenotypic analysis. No significant differences in body length and body weight were observed between the amhr2 mutants and wild-type fish at 200 dah (Supplementary Table S1).

2.4. Effect of Fadrozole Treatment on Gonadal Phenotype of amhr2 Mutants

The histological analysis revealed that the amhr2 mutants underwent sex reversal from male to female. To investigate the involvement of estrogens in the feminization of the amhr2 mutants, we administered fadrozole, which inhibits estrogen synthesis, to the amhr2 mutants. The larvae were treated with fadrozole for 30–100 dah, and a histological analysis was performed using the flounder at 200 dah. The results showed that the gonads of the XY amhr2 mutants exhibited normal ovaries, which were similar to those of the females in the control group, whereas the gonads of the fadrozole-treated XY amhr2 mutants exhibited normal testes (Figure 4A; Table 2). All of the fadrozole-treated XX amhr2 mutants also had typical testes in this experiment. Next, the expression of the sex differentiation-related genes was examined using the flounder at 200 dah by a real-time PCR. The results showed that amh was significantly up-regulated in the fadrozole-treated amhr2 mutants (Figure 4B), while cyp19a1 and foxl2 were prominently down-regulated (Figure 4C,D). Meanwhile, the fshr expression levels were not significantly different among all of the experimental groups (Figure 4E). These results indicate that estrogens are indispensable for the feminization of amhr2 mutants in Japanese flounder.

3. Discussion

In the present study, to elucidate the function of amhr2 in the gonadal differentiation in Japanese flounder (Paralichthys olivaceus), we examined the expression pattern of its transcripts and confirmed transcript localization in the somatic cells in contact with gonium cells in both of the sexes. The loss of function of the amhr2 gene produced using the CRISPR/Cas9 system led to a sex reversal from male to female in genetically male fish. The expression pattern of sex differentiation-related genes showed feminization that was similar to that of normal females. Furthermore, the treatment with fadrozole, which inhibits estrogen synthesis, caused the female-to-male sex reversal in the genetically male amhr2 mutants.
Reports on amh in various teleost fish species have been published [17,26,27,28]. The function of this gene has been considered to be important for male sex differentiation in vertebrates. Based on quantitative real-time PCR, the amh expression in XY males was significantly higher than that in XX females from the sexually indifferent stage to gonadal post-differentiation stage (50–200 dah). These results were similar to the expression pattern of amh upon masculinization due to a high temperature in Japanese flounder [8,10]. Meanwhile, the expression levels of the amhr2 transcripts showed no difference in the periods from sex differentiation to post-gonadal differentiation. Indeed, it was previously reported that amhr2 does not exhibit sexually dimorphic expression during the gonadal differentiation period (50–80 dah) in Japanese flounder [24]. However, the expression of amh was shown to be higher in males than it was in females during sex differentiation in various teleost fish [17,26,27]. Based on in situ hybridization data, the amh and amhr2 transcript signals were detected in the somatic cells in contact with gonium cells in Japanese flounder. In other vertebrates (e.g., chickens [29], medaka [30,31,32], and zebrafish [33]), amh and amhr2 transcripts appeared in the Sertoli cells surrounding the male germ cells and in the follicle cells of female germ cells. Although we did not assess the protein expressions of Amh and Amhr2, it is speculated that amh and amhr2 are also expressed in the somatic cells the surrounding germ cells and that Amh/Amhr2 signaling plays an important role in these cells during the sex differentiation period in Japanese flounder.
To investigate the functions of the amhr2 gene in more detail, we produced an amhr2 mutant using the CRISPR/Cas9 system. This system can achieve the highly efficient production of biallelic knockout mutants during F0 generation [25]. In the amhr2 mutant flounder, large deletions were produced within the kinase domain of Amhr2, which is responsible for phosphorylation. Amhr2 is a type II receptor of the TGF-β superfamily, containing a single-transmembrane domain and an intercellular serine/threonine kinase domain [18]. Amh bound to Amhr2 was reported to induce the phosphorylation of type I receptors, transduce signals by phosphorylating Smad proteins, and regulate the transcription of downstream genes in mammals [34]. In medaka fish, a hotei mutant (amhr2-deficient) showed male-to-female sex reversal [35]. The mutant analysis identified a mutation in the amhr2 gene, namely, an A-to-G mutation in exon 9, which is located in the receptor kinase domain. In the tiger pufferfish (Takifugu rubripes), a single-nucleotide polymorphism (SNP) in the amhr2 gene was also reported to be associated with sex determination [36]. This SNP (C to G) in exon 9 changes an amino acid that is located in the receptor kinase domain from His to Asp. The findings showed that the kinase domain is critical for the function of Amhr2, suggesting that amhr2 mutant flounder produced in this study shows a complete loss of function of Amh/Amhr2 signaling.
The Japanese flounder has an XX/XY sex determination system [2]. Recently, Y-chromosome allele-specific amh (amhy) was found to be the master sex-determining gene in flounder [24]. amh-mutant XY flounder showed phenotypic sex reversal from male to female, suggesting that Amh signaling is indispensable for the testicular differentiation in flounder. In mammals, the disruption of Amh signaling in Amh knockout XY mice led to partial hermaphroditism with premature follicle development [37]. Moreover, in Nile tilapia (Oreochromis niloticus), which also has an XX/XY sex determination system, there are two male-specific duplications of amh, which are designated amhy and amhΔ-y [38]. The knockout of amhy, amhΔ-y, and amhr2 using the CRISPR/Cas9 system caused male-to-female sex reversal. Meanwhile, amhr2-deficient XY medaka showed sex reversal at a rate of more than 50% [35]. In this study, the loss of Amhr2 function causes male-to-female sex reversal in Japanese flounder. Thus, Amh/Amhr2 signaling may be responsible for male sex determination and testicular development in all vertebrates.
In vertebrates, estrogen plays an important role in ovarian differentiation and maintenance [13,39]. Foxl2 is a factor involved in female sex determination, which directly activates the expression of cyp19a1 in vertebrates [10,40,41]. In medaka, cyp19a1 expression is up-regulated in amhr2-deficient XY fish [35]. Similar results have reported that the knockdown of amhy in XY Patagonian pejerrey (Odontesthes hatcheri) resulted in the up-regulation of foxl2 and cyp19a1 expression [42]. Consistent with these reports, the knockout of amhr2 in XY flounder resulted in increased cyp19a1 and foxl2 expression, along with normal ovary development. Therefore, to confirm the involvement of estrogens in amhr2 mutants, we administered fadrozole, which inhibits estrogen synthesis. Fadrozole-treated XY amhr2 mutants formed typical testes and exhibited the down-regulation of cyp19a1 and foxl2 transcripts. Previously, a fadrozole treatment caused complete female-to-male sex reversal with the down-regulation of cyp19a1 expression in the flounder [7]. These results indicate that amhr2-deficient mutants display a phenotypic sex reversal from male to female by elevating the level of estrogens, along with increased cyp19a1 and foxl2 expression. Therefore, Amh/Amhr2 signaling appears to cause female-to-male sex reversal by inhibiting estrogen synthesis.
In Japanese flounder, female fish grow faster than the males do, and therefore, females are highly valued in the aquaculture industry [2,3]. In this study, although there was no significant difference in the body length and the body weight among all of the experimental groups at 200 dah (Supplementary Table S1), the sex difference in growth is likely to clarify after they reach 1 year of age [2]. Therefore, future studies will be needed to continuously research the growth of amhr2 mutant flounder.
In conclusion, amhr2 was expressed specifically in the somatic cells surrounding the germ cells during gonadal sex differentiation in both XY and XX flounder, and the loss of Amhr2 function causes male-to-female sex reversal in the flounder, along with increased cyp19a1 and foxl2 expressions. Moreover, fadrozole-treated amhr2 mutants had typical testes and exhibited the down-regulation of cyp19a1 and foxl2 transcripts. These results strongly suggest that Amh/Amhr2 signaling causes masculinization by inhibiting estrogen synthesis during gonadal sex differentiation in the flounder.

4. Materials and Methods

4.1. Experimental Animals

The Japanese flounder (Paralichthys olivaceus) was used for this study. The flounder were produced artificially by mating normal males and females, and they were reared at the Nansei Field Station, Japan Fisheries Research and Education Agency. The larvae were reared at a water temperature of 16–18 °C to prevent masculinization due to high water temperatures. All of the fish were sampled after ensuring that they had been completely anesthetized by 2-phenoxyethanol (Fujifilm Wako Chemicals, Osaka, Japan). All of the experimental protocols were approved by the Institutional Animal Care and Use Committee of Nansei Field Station (2021-13).

4.2. Determination of Genetic Sex

The genetic sex of all of the experimental individuals was determined by genomic PCR with reference to Y-allele-specific deletion in the promoter region of amhy [24]. Genomic DNA was extracted from fin or gonad samples using ISOGEN® (Nippon Gene, Toyama, Japan) following the manufacturer’s instructions. A genomic PCR was carried out using AmpliTaq Gold® (Applied Biosystems, Foster City, CA, USA) with the following primers: forward, 5′-GTTCAGTTCAGTTGCACAGC-3′, and reverse, 5′-AATGGGTCCAGCTTCAGAGG-3′.

4.3. Expression Analysis by Real-Time PCR

Total RNA was extracted from the gonads using the RNeasy Mini Kit (Qiagen, Germantown, MD, USA) following the manufacturer’s instructions. cDNA synthesis was carried out using ReverTra Ace® (Toyobo, Osaka, Japan) from 100 ng of total RNA. The first strand cDNAs were diluted to 100 µL for subsequent use. A quantitative real-time PCR was performed using SYBR® Green Real-time PCR Master Mix (Toyobo) and 5 μL of cDNA on an iCycler iQ (Bio-Rad, Hercules, CA, USA) with the following primers: forward for amh, 5′-TGACCCGTACCTACGAGCTG-3′, and reverse, 5′-TCGTCCACGTTCTCGCTCTC-3′; forward for amhr2, 5′-ACTGCTGGTAATGTGAGTGG-3′, and reverse, 5′-GCCCAGAATCTGCTGTAGTT-3′; forward for cyp19a1, 5′-ATCGGATCCCTGCCTGTGAC-3′, and reverse, 5′-TGGCTGATGCTCTGCTGAGG-3′; forward for foxl2, 5′-TCATCAGCAAGTTCCCCTTC-3′, and reverse, 5′-TGCCGTTGTAAGAGTTCACC-3′; forward for fshr, 5′-TCCAAACTGACAGTTCCTCG-3′, and reverse, 5′-AGAAGGCTAGGATGTTGAGG-3′; forward for elongation factor 1 alpha (ef1α), 5′-AGTTCGAGAAGGAAGCTGCC-3′, and reverse, 5′-ATCCAGAGCATCCAGCAGTG-3′; forward for α-tubulin, 5′-TGGTACGTAGGAGAGGGCAT-3′, and reverse, 5′-CCCTCTTCGTCTTCCTCGAACG-3′; forward for β-actin, 5′- ATTGCCCCACCTGAGCGTA-3′, and reverse, 5′- CATTTGCGGTGGACGATGGA-3′; forward for glyceraldehyde-3-phosphate dehydrogenase (gapdh), 5′- CGCTTCAAGGGTGAGGTCAA-3′, and reverse, 5′- TCAACCACATAGTGGGCACC-3′. The PCR conditions were as follows: initial denaturation at 95 °C for 60 s, followed by 40 cycles of 95 °C for 15 s, 60 °C for 15 s, and 72 °C for 45 s. The transcript abundance was quantified using the standard curve method with four dilution points. RefFinder [43], which integrates four specific algorithms (the comparative delta-Ct method [44], BestKeeper [45], NormFinder [46], and GeNorm [47]), was used for the assessment and screening of candidate reference genes (ef1α, α-tubulin, β-actin, and gapdh). The most stably expressed gene ef1α was used as a reference gene in this study (Supplementary Table S2).

4.4. Histological Analysis of amhr2 Transcripts Using In Situ Hybridization

The gonads were fixed in 4% paraformaldehyde (PFA) solution at 4 °C overnight, dehydrated in graded ethanol, embedded in paraffin, and sectioned serially at 5 μm thickness. In situ hybridization was performed using a digoxigenin (DIG)-labeled amh or amhr2 RNA probe using the sections as described previously [17]. The probes for DIG-labeled RNA single strands were synthesized in vitro using DIG RNA labeling kit (Roche Diagnostics GmbH, Mannheim, Germany).

4.5. Generation of amhr2 Mutant Using CRISPR/Cas9 System

Synthetic crRNA and trans-activating crRNA (tracrRNA) were obtained from Fasmac Co., Ltd. (Kanagawa, Japan). The sequences of the two crRNAs for amhr2 exons were as follows: Target 1, CAGUGACAGCAUCCAGCUGGguuuuagagcuaugcuguuuug; Target 2, UGCUUUGGGACUGCUGCUGUguuuuagagcuaugcuguuuug. These sequences were designed as described previously [25]. The two crRNAs (250 ng/µL), tracrRNA (500 ng/µL), and Cas9 protein (750 ng/µL; Nippon Gene) were mixed and injected into fertilized eggs immediately after fertilization using a microinjector (IM-9B; Narishige, Tokyo, Japan) as described previously [25]. The two crRNAs and Cas9 protein without tracrRNA were injected for the experimental control group. The artificial fertilization, microinjection, and rearing of the experimental fish were performed at Nansei Field Station, Japan Fisheries Research and Education Agency. Eggs and sperm were produced by 3-year-old females and males.

4.6. Analysis of Genotype in amhr2 Mutants

For genotyping, genomic DNA was extracted from the gonads using ISOGEN® (Nippon Gene) following the manufacturer’s instructions. Genomic PCR was carried out using AmpliTaq Gold® (Applied Biosystems) with the following primers specific for amhr2 (forward, 5′-TCTCTTCATTCCTTTCTGTG-3′, and reverse, 5′-TAAATCTGAGCAGCGCATC-3′). The PCR conditions were as follows: preheating at 95 °C for 2 min; 35 cycles of 95 °C for 15 s, 59 °C for 30 s, and 72 °C for 2 min; then, a final extension at 72 °C for 5 min. The amplified PCR fragments were subcloned using TA PCR Cloning Kit (BioDynamics Laboratory, Tokyo, Japan) following the manufacturer’s instructions. The sequencing of subcloned amplicon PCR fragments was outsourced to a sequencing service (Fasmac Co., Ltd.).

4.7. Histological Observations

At 200 dah, the gonads were immediately fixed with Bouin’s solution at 4 °C overnight. The fixed gonads were dehydrated in a series of ethanol solutions and cleared using Lemosol (Fujifilm Wako Chemicals). The gonadal tissue was embedded in paraffin, sectioned serially at 4 µm thickness, stained using hematoxylin-eosin, and observed under a microscope (BZ-X700; Keyence, Osaka, Japan).

4.8. Experimental Treatment with Fadrozole

Fadrozole was orally administered with an artificial diet (Love Larva; Maruha Co., Yamaguchi, Japan) mixed with a 100 μg/g diet of fadrozole (Sigma-Aldrich, St. Louis, MO, USA) at 18 °C from 30 to 100 dah as described previously [7]. Phenotypic sex was determined at 200 dah by histological observation.

4.9. Statistical Analysis

The significance of the differences in the experimental results in the relative mRNA expressions among the various experimental groups was tested by one-way ANOVA based on normalized data, followed by Tukey’s test or Student’s t-test. All of the statistical analyses were performed using SPSS version 22 (IBM Corp., Armonk, NY, USA), and all of the experimental data are presented as mean ± SEM.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24032480/s1.

Author Contributions

T.Y. and T.K. obtained funding and designed the study. T.Y. and T.K. performed the experiments and collected the data. T.Y. and T.K. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the JSPS KAKENHI Grant Number 21K05740 from the Ministry of Education, Culture, Sports, Science and Technology of Japan.

Institutional Review Board Statement

This study was conducted according to the guidelines provided by the Institutional Animal Care and Use Committee of Nansei Field Station (2021-13).

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are available within this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Baroiller, J.F.; D’Cotta, H. The Reversible Sex of Gonochoristic Fish: Insights and Consequences. Sex Dev. 2016, 10, 242–266. [Google Scholar] [CrossRef] [PubMed]
  2. Yamamoto, E. Studies on sex-manipulation and production of cloned populations in hirame, Paralichthys olivaceus (Temminck et Schlegel). Aquaculture 1999, 173, 235–246. [Google Scholar] [CrossRef]
  3. Tabata, K. Reduction of female proportion in lower growing fish separated from normal and feminized seedlings of hirame Paralichthys olivaceus. Fish Sci. 1995, 61, 199–201. [Google Scholar] [CrossRef] [Green Version]
  4. Kitano, T.; Takamune, K.; Nagahama, Y.; Abe, S. Role of P450 aromatase in gonadal sex differentiation in Japanese flounder (Paralichthys olivaceus). Environ. Sci. 2001, 8, 1–11. [Google Scholar]
  5. Ospina-Alvarez, N.; Piferrer, F. Temperature-dependent sex determination in fish revisited: Prevalence, a single sex ratio response pattern, and possible effects of climate change. PLoS ONE 2008, 3, e2837. [Google Scholar] [CrossRef] [Green Version]
  6. Rajendiran, P.; Jaafar, F.; Kar, S.; Sudhakumari, C.; Senthilkumaran, B.; Parhar, I.S. Sex Determination and Differentiation in Teleost: Roles of Genetics, Environment, and Brain. Biology 2021, 10, 973. [Google Scholar] [CrossRef]
  7. Kitano, T.; Takamune, K.; Nagahama, Y.; Abe, S.I. Aromatase inhibitor and 17alpha-methyltestosterone cause sex-reversal from genetical females to phenotypic males and suppression of P450 aromatase gene expression in Japanese flounder (Paralichthys olivaceus). Mol. Reprod. Dev. 2000, 56, 1–5. [Google Scholar] [CrossRef]
  8. Kitano, T.; Yoshinaga, N.; Shiraishi, E.; Koyanagi, T.; Abe, S. Tamoxifen induces masculinization of genetic females and regulates P450 aromatase and Müllerian inhibiting substance mRNA expression in Japanese flounder (Paralichthys olivaceus). Mol. Reprod. Dev. 2007, 74, 1171–1177. [Google Scholar] [CrossRef]
  9. Kitano, T.; Hayashi, Y.; Shiraishi, E.; Kamei, Y. Estrogen rescues masculinization of genetically female medaka by exposure to cortisol or high temperature. Mol. Reprod. Dev. 2012, 79, 719–726. [Google Scholar] [CrossRef]
  10. Yamaguchi, T.; Yamaguchi, S.; Hirai, T.; Kitano, T. Follicle-stimulating hormone signaling and Foxl2 are involved in transcriptional regulation of aromatase gene during gonadal sex differentiation in Japanese flounder, Paralichthys olivaceus. Biochem. Biophys. Res. Commun. 2007, 359, 935–940. [Google Scholar] [CrossRef]
  11. Yamaguchi, T.; Yoshinaga, N.; Yazawa, T.; Gen, K.; Kitano, T. Cortisol is involved in temperature-dependent sex determination in the Japanese flounder. Endocrinology 2010, 151, 3900–3908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Fitzpatrick, S.L.; Richards, J.S. Identification of a cyclic adenosine 3’,5’-monophosphate-response element in the rat aromatase promoter that is required for transcriptional activation in rat granulosa cells and R2C Leydig cells. Mol. Endocrinol. 1994, 8, 1309–1319. [Google Scholar] [PubMed] [Green Version]
  13. Guiguen, Y.; Fostier, A.; Piferrer, F.; Chang, C.F. Ovarian aromatase and estrogens: A pivotal role for gonadal sex differentiation and sex change in fish. Gen. Comp. Endocrinol. 2010, 165, 352–366. [Google Scholar] [CrossRef]
  14. Huang, S.; Ye, L.; Chen, H. Sex determination and maintenance: The role of DMRT1 and FOXL2. Asian J. Androl. 2017, 19, 619–624. [Google Scholar] [PubMed]
  15. Hayashi, Y.; Kobira, H.; Yamaguchi, T.; Shiraishi, E.; Yazawa, T.; Hirai, T.; Kamei, Y.; Kitano, T. High temperature causes masculinization of genetically female medaka by elevation of cortisol. Mol. Reprod. Dev. 2010, 77, 679–686. [Google Scholar] [CrossRef] [PubMed]
  16. Hattori, R.S.; Fernandino, J.I.; Kishii, A.; Kimura, H.; Kinno, T.; Oura, M.; Somoza, G.M.; Yokota, M.; Strüssmann, C.A.; Watanabe, S. Cortisol-induced masculinization: Does thermal stress affect gonadal fate in pejerrey, a teleost fish with temperature-dependent sex determination? PLoS ONE 2009, 4, e6548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Yoshinaga, N.; Shiraishi, E.; Yamamoto, T.; Iguchi, T.; Abe, S.; Kitano, T. Sexually dimorphic expression of a teleost homologue of Müllerian inhibiting substance during gonadal sex differentiation in Japanese flounder, Paralichthys olivaceus. Biochem. Biophys. Res. Commun. 2004, 322, 508–513. [Google Scholar] [CrossRef]
  18. Josso, N. In vitro synthesis of müllerian-inhibiting hormone by seminiferous tubules isolated from the calf fetal testis. Endocrinology 1973, 93, 829–834. [Google Scholar] [CrossRef]
  19. Tilmann, C.; Capel, B. Cellular and molecular pathways regulating mammalian sex determination. Recent Prog. Horm. Res. 2002, 57, 1–18. [Google Scholar] [CrossRef] [Green Version]
  20. Park, S.Y.; Jameson, J.L. Minireview: Transcriptional regulation of gonadal development and differentiation. Endocrinology 2005, 146, 1035–1042. [Google Scholar] [CrossRef] [Green Version]
  21. Josso, N.; Racine, C.; di Clemente, N.; Rey, R.; Xavier, F. The role of anti-Müllerian hormone in gonadal development. Mol. Cell. Endocrinol. 1998, 145, 3–7. [Google Scholar] [CrossRef] [PubMed]
  22. Behringer, R.R.; Cate, R.L.; Froelick, G.J.; Palmiter, R.D.; Brinster, R.L. Abnormal sexual development in transgenic mice chronically expressing müllerian inhibiting substance. Nature 1990, 345, 167–170. [Google Scholar] [CrossRef] [PubMed]
  23. Mishina, Y.; Whitworth, D.J.; Racine, C.; Behringer, R.R. High specificity of Müllerian-inhibiting substance signaling in vivo. Endocrinology 1999, 140, 2084–2088. [Google Scholar] [CrossRef] [PubMed]
  24. Hattori, R.S.; Kumazawa, K.; Nakamoto, M.; Nakano, Y.; Yamaguchi, T.; Kitano, T.; Yamamoto, E.; Fuji, K.; Sakamoto, T. Y-specific amh allele, amhy, is the master sex-determining gene in Japanese flounder Paralichthys olivaceus. Front. Genet. 2022, 13, 1007548. [Google Scholar] [CrossRef] [PubMed]
  25. Sawamura, R.; Osafune, N.; Murakami, T.; Furukawa, F.; Kitano, T. Generation of biallelic F0 mutants in medaka using the CRISPR/Cas9 system. Genes Cells 2017, 22, 756–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Rodríguez-Marí, A.; Yan, Y.L.; Bremiller, R.A.; Wilson, C.; Cañestro, C.; Postlethwait, J.H. Characterization and expression pattern of zebrafish Anti-Müllerian hormone (Amh) relative to sox9a, sox9b, and cyp19a1a, during gonad development. Gene Expr. Patterns 2005, 5, 655–667. [Google Scholar] [CrossRef]
  27. Ijiri, S.; Kaneko, H.; Kobayashi, T.; Wang, D.S.; Sakai, F.; Paul-Prasanth, B.; Nakamura, M.; Nagahama, Y. Sexual dimorphic expression of genes in gonads during early differentiation of a teleost fish, the Nile tilapia Oreochromis niloticus. Biol. Reprod. 2008, 78, 333–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Mullen, R.D.; Ontiveros, A.E.; Moses, M.M.; Behringer, R.R. AMH and AMHR2 mutations: A spectrum of reproductive phenotypes across vertebrate species. Dev. Biol. 2019, 455, 1–9. [Google Scholar] [CrossRef]
  29. Oréal, E.; Mazaud, S.; Picard, J.Y.; Magre, S.; Carré-Eusèbe, D. Different patterns of anti-Müllerian hormone expression, as related to DMRT1, SF-1, WT1, GATA-4, Wnt-4, and Lhx9 expression, in the chick differentiating gonads. Dev. Dyn. 2002, 225, 221–232. [Google Scholar] [CrossRef]
  30. Klüver, N.; Pfennig, F.; Pala, I.; Storch, K.; Schlieder, M.; Froschauer, A.; Gutzeit, H.O.; Schartl, M. Differential expression of anti-Müllerian hormone (amh) and anti-Müllerian hormone receptor type II (amhrII) in the teleost medaka. Dev. Dyn. 2007, 236, 271–281. [Google Scholar] [CrossRef]
  31. Shiraishi, E.; Yoshinaga, N.; Miura, T.; Yokoi, H.; Wakamatsu, Y.; Abe, S.; Kitano, T. Mullerian inhibiting substance is required for germ cell proliferation during early gonadal differentiation in medaka (Oryzias latipes). Endocrinology 2008, 149, 1813–1819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kawabe, T.; Kariya, H.; Hara, S.; Shirozu, T.; Shiraishi, E.; Mukai, K.; Yazawa, T.; Inoue, S.; Kitano, T. Transcriptional regulation of Müllerian inhibiting substance (MIS) and establishment of a gonadal somatic cell line using mis-GFP transgenic medaka (Oryzias latipes). Front. Endocrinol. 2020, 11, 578885. [Google Scholar] [CrossRef] [PubMed]
  33. von Hofsten, J.; Larsson, A.; Olsson, P.E. Novel steroidogenic factor-1 homolog (ff1d) is coexpressed with anti-Mullerian hormone (AMH) in zebrafish. Dev. Dyn. 2005, 233, 595–604. [Google Scholar] [CrossRef] [PubMed]
  34. Belville, C.; Maréchal, J.D.; Pennetier, S.; Carmillo, P.; Masgrau, L.; Messika-Zeitoun, L.; Galey, J.; Machado, G.; Treton, D.; Gonzalès, J.; et al. Natural mutations of the anti-Mullerian hormone type II receptor found in persistent Mullerian duct syndrome affect ligand binding, signal transduction and cellular transport. Hum. Mol. Genet. 2009, 18, 3002–3013. [Google Scholar] [CrossRef] [Green Version]
  35. Morinaga, C.; Saito, D.; Nakamura, S.; Sasaki, T.; Asakawa, S.; Shimizu, N.; Mitani, H.; Furutani-Seiki, M.; Tanaka, M.; Kondoh, H. The hotei mutation of medaka in the anti-Mullerian hormone receptor causes the dysregulation of germ cell and sexual development. Proc. Natl. Acad. Sci. USA 2007, 104, 9691–9696. [Google Scholar] [CrossRef] [Green Version]
  36. Kamiya, T.; Kai, W.; Tasumi, S.; Oka, A.; Matsunaga, T.; Mizuno, N.; Fujita, M.; Suetake, H.; Suzuki, S.; Hosoya, S.; et al. A trans-species missense SNP in Amhr2 is associated with sex determination in the tiger pufferfish, Takifugu rubripes (fugu). PLoS Genet. 2012, 8, e1002798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Josso, N.; di Clemente, N.; Gouédard, L. Anti-Müllerian hormone and its receptors. Mol. Cell. Endocrinol. 2001, 179, 25–32. [Google Scholar] [CrossRef] [PubMed]
  38. Li, M.; Sun, Y.; Zhao, J.; Shi, H.; Zeng, S.; Ye, K.; Jiang, D.; Zhou, L.; Sun, L.; Tao, W.; et al. A Tandem Duplicate of Anti-Müllerian Hormone with a Missense SNP on the Y Chromosome Is Essential for Male Sex Determination in Nile Tilapia, Oreochromis niloticus. PLoS Genet. 2015, 11, e1005678. [Google Scholar] [CrossRef] [Green Version]
  39. Wang, D.S.; Kobayashi, T.; Zhou, L.Y.; Paul-Prasanth, B.; Ijiri, S.; Sakai, F.; Okubo, K.; Morohashi, K.; Nagahama, Y. Foxl2 up-regulates aromatase gene transcription in a female-specific manner by binding to the promoter as well as interacting with ad4 binding protein/steroidogenic factor 1. Mol. Endocrinol. 2007, 21, 712–725. [Google Scholar] [CrossRef] [Green Version]
  40. Okada, E.; Yoshimoto, S.; Ikeda, N.; Kanda, H.; Tamura, K.; Shiba, T.; Takamatsu, N.; Ito, M. Xenopus W-linked DM-W induces Foxl2 and Cyp19 expression during ovary formation. Sex Dev. 2009, 3, 38–42. [Google Scholar] [CrossRef] [PubMed]
  41. Uhlenhaut, N.H.; Jakob, S.; Anlag, K.; Eisenberger, T.; Sekido, R.; Kress, J.; Treier, A.C.; Klugmann, C.; Klasen, C.; Holter, N.I.; et al. Somatic sex reprogramming of adult ovaries to testes by FOXL2 ablation. Cell 2009, 139, 1130–1142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hattori, R.S.; Oura, M.; Sakamoto, T.; Yokota, M.; Watanabe, S.; Strüssmann, C.A. Establishment of a strain inheriting a sex-linked SNP marker in Patagonian pejerrey (Odontesthes hatcheri), a species with both genetic and temperature-dependent sex determination. Anim. Genet. 2010, 41, 81–84. [Google Scholar] [CrossRef] [PubMed]
  43. Xie, F.; Xiao, P.; Chen, D.; Xu, L.; Zhang, B. miRDeepFinder: A miRNA analysis tool for deep sequencing of plant small RNAs. Plant Mol. Biol. 2012, 80, 75–84. [Google Scholar] [CrossRef] [PubMed]
  44. Silver, N.; Best, S.; Jiang, J.; Thein, S.L. Selection of housekeeping genes for gene expression studies in human reticulocytes using real-time PCR. BMC Mol. Biol. 2006, 7, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Pfaffl, M.W.; Tichopad, A.; Prgomet, C.; Neuvians, T.P. Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper--Excel-based tool using pair-wise correlations. Biotechnol. Lett. 2004, 26, 509–515. [Google Scholar] [CrossRef]
  46. Andersen, C.L.; Jensen, J.L.; Ørntoft, T.F. Normalization of real-time quantitative reverse transcription-PCR data: A model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res. 2004, 64, 5245–5250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Vandesompele, J.; De Preter, K.; Pattyn, F.; Poppe, B.; Van Roy, N.; De Paepe, A.; Speleman, F. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002, 3, 1–12. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Expression analysis of amh and amhr2 in XY and XX flounder. Real-time PCR analysis of amh (A) and amhr2 (B) expression in XY and XX flounder from the sexually indifferent stage (50 dah) to the stage of clear differentiation in testis or ovary (200 dah). * p < 0.05, ** p < 0.01. (C) Cellular localizations of amh and amhr2 in the flounder gonads (200 dah) by in situ hybridization. Arrowheads indicate signals positive for amh or amhr2 transcripts, while arrows indicate gonium cells. Oo: oocyte. Scale bar: 20 μm.
Figure 1. Expression analysis of amh and amhr2 in XY and XX flounder. Real-time PCR analysis of amh (A) and amhr2 (B) expression in XY and XX flounder from the sexually indifferent stage (50 dah) to the stage of clear differentiation in testis or ovary (200 dah). * p < 0.05, ** p < 0.01. (C) Cellular localizations of amh and amhr2 in the flounder gonads (200 dah) by in situ hybridization. Arrowheads indicate signals positive for amh or amhr2 transcripts, while arrows indicate gonium cells. Oo: oocyte. Scale bar: 20 μm.
Ijms 24 02480 g001
Figure 2. Construction of amhr2-mutant Japanese flounder using the CRISPR/Cas9 system. Schematic of positions targeted by crRNAs on the amhr2 locus, showing exons (white boxes), introns (horizontal lines), and target positions (arrows) of the crRNAs with the CRISPR/Cas9 system (A). The target sequences of wild-type and amhr2 mutants (B). The horizontal lines and dashed lines indicate wild-type sequences and deletions, respectively. Slashes indicate omission of long sequences. The numbers beside the target sequences indicate the approximate size of deletion (−) or insertion (+).
Figure 2. Construction of amhr2-mutant Japanese flounder using the CRISPR/Cas9 system. Schematic of positions targeted by crRNAs on the amhr2 locus, showing exons (white boxes), introns (horizontal lines), and target positions (arrows) of the crRNAs with the CRISPR/Cas9 system (A). The target sequences of wild-type and amhr2 mutants (B). The horizontal lines and dashed lines indicate wild-type sequences and deletions, respectively. Slashes indicate omission of long sequences. The numbers beside the target sequences indicate the approximate size of deletion (−) or insertion (+).
Ijms 24 02480 g002
Figure 3. Phenotypic analysis of amhr2 knockout mutants. Histological images of gonads from controls and amhr2 mutants at 200 dah (A). Scale bar, 100 µm. Real-time PCR analyses of amh (B), cyp19a1 (C), foxl2 (D), and fshr (E) expressions were carried out using the gonads of the flounder at 200 dah. Different letters denote significant differences at p < 0.01.
Figure 3. Phenotypic analysis of amhr2 knockout mutants. Histological images of gonads from controls and amhr2 mutants at 200 dah (A). Scale bar, 100 µm. Real-time PCR analyses of amh (B), cyp19a1 (C), foxl2 (D), and fshr (E) expressions were carried out using the gonads of the flounder at 200 dah. Different letters denote significant differences at p < 0.01.
Ijms 24 02480 g003
Figure 4. Phenotypic analysis of amhr2 knockout mutants treated with the aromatase inhibitor fadrozole. Histological images of gonads from control fish, amhr2 mutants, and fadrozole-treated amhr2 mutants at 200 dah (A). Scale bar, 100 µm. Real-time PCR analyses of amh (B), cyp19a1 (C), foxl2 (D), and fshr (E) expressions were carried out using the gonads of the flounder at 200 dah. Different letters denote significant differences at p < 0.01.
Figure 4. Phenotypic analysis of amhr2 knockout mutants treated with the aromatase inhibitor fadrozole. Histological images of gonads from control fish, amhr2 mutants, and fadrozole-treated amhr2 mutants at 200 dah (A). Scale bar, 100 µm. Real-time PCR analyses of amh (B), cyp19a1 (C), foxl2 (D), and fshr (E) expressions were carried out using the gonads of the flounder at 200 dah. Different letters denote significant differences at p < 0.01.
Ijms 24 02480 g004
Table 1. The types of mutations in amhr2 knockout mutants.
Table 1. The types of mutations in amhr2 knockout mutants.
Types of Mutations
Genetic SexPhenotypic Sex
(Gonad)
Number of Clones SequencedWide Deletion Short DeletionNo Mutation
wild-typeXY♂ (Testis)4004
XX♀ (Ovary)4004
amhr2 mutantXY♀ (Ovary)161510
XY♀ (Ovary)161330
XY♀ (Ovary)161510
XX♀ (Ovary)8800
XX♀ (Ovary)8620
Table 2. Sex ratios in wild-type and amhr2 mutant flounder fed an fadrozole-treated diet or untreated diet (control).
Table 2. Sex ratios in wild-type and amhr2 mutant flounder fed an fadrozole-treated diet or untreated diet (control).
TreatmentGenotypeGenetic Sex♂ (Testis)♀ (Ovary)
Controlwild-typeXY90
Controlwild-typeXX011
Controlamhr2 mutantXY06
Controlamhr2 mutantXX04
Fadrozoleamhr2 mutantXY50
Fadrozoleamhr2 mutantXX50
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yamaguchi, T.; Kitano, T. Amh/Amhr2 Signaling Causes Masculinization by Inhibiting Estrogen Synthesis during Gonadal Sex Differentiation in Japanese Flounder (Paralichthys olivaceus). Int. J. Mol. Sci. 2023, 24, 2480. https://doi.org/10.3390/ijms24032480

AMA Style

Yamaguchi T, Kitano T. Amh/Amhr2 Signaling Causes Masculinization by Inhibiting Estrogen Synthesis during Gonadal Sex Differentiation in Japanese Flounder (Paralichthys olivaceus). International Journal of Molecular Sciences. 2023; 24(3):2480. https://doi.org/10.3390/ijms24032480

Chicago/Turabian Style

Yamaguchi, Toshiya, and Takeshi Kitano. 2023. "Amh/Amhr2 Signaling Causes Masculinization by Inhibiting Estrogen Synthesis during Gonadal Sex Differentiation in Japanese Flounder (Paralichthys olivaceus)" International Journal of Molecular Sciences 24, no. 3: 2480. https://doi.org/10.3390/ijms24032480

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop