Skip to main content

The ontogenic gonadal transcriptomes provide insights into sex change in the ricefield eel Monopterus albus

Abstract

Background

The ricefield eel is a freshwater protogynous hermaphrodite fish and has become an important aquaculture species in China. The sex change of ricefield eel is impeding its aquaculture practice, particularly the large-scale artificial breeding. Many studies including transcriptomes of mixed gonadal samples from different individuals have been aimed to elucidate mechanisms underlying the sex change. However, the key physiological factors involved in the initiation of sex change remain to be identified. 

Results

The present study performed transcriptomic analysis on gonadal samples of different sexual stages obtained through biopsy from the same fish undergoing sex change. A total of 539,764,816 high-quality reads were generated from twelve cDNA libraries of gonadal tissues at female (F), early intersexual (EI), mid-intersexual (MI), and late intersexual (LI) stages of three individual sex-changing fish. Pairwise comparisons between EI and F, MI and EI, and LI and MI identified 886, 319, and 10,767 differentially expressed genes (DEGs), respectively. Realtime quantitative PCR analysis of 12 representative DEGs showed similar expression profiles to those inferred from transcriptome data, suggesting the reliability of RNA-seq data for gene expression analysis. The expression of apoeb, csl2, and enpp2 was dramatically increased and peaked at EI while that of cyp19a1a, wnt4a, fgf16, and foxl2a significantly downregulated from F to EI and remained at very low levels during subsequent development until LI, which suggests that apoeb, csl2, enpp2, cyp19a1a, wnt4a, fgf16, and foxl2a may be closely associated with the initiation of sex change of ricefield eels.

Conclusions

Collectively, results of the present study confirmed that the down-regulation of female-related genes, such as cyp19a1a, wnt4a, fgf16, and foxl2a, is important for the sex change of ricefield eels. More importantly, some novel genes, including apoeb, csl2, and enpp2, were shown to be expressed with peak values at EI, which are potentially involved in the initiation of sex change. The present transcriptomic data may provide an important research resource for further unraveling the mechanisms underlying the sex change and testicular development in ricefield eels as well as other teleosts.

Peer Review reports

Background

Most vertebrates are gonochoristic, in which individual organisms contain only male or female sex organs throughout their lifetime [1, 2]. In contrast, hermaphrodites in which individual organisms contain both male and female sex organs, are observed in about 2% of all extant teleost species scattering across more than 20 taxonomic families in 9 orders, which can be categorized into sequential and synchronous modes [3]. The sequential hermaphroditism can change their primary sexual status and takes on three forms including protogynous (female-to-male), protandrous (male-to-female), and serial bidirectional sex change [4]. How natural sex change may initiate and progress remains to be elucidated [5]. Natural sex change involves coordinated transformations across multiple biological systems, including behavioural, anatomical, neuroendocrine, and molecular axes, but the molecular basis underlying this amazing transformation remains poorly understood [6].

Ricefield eel Monopterus albus, a member of the order Synbranchiformes, is a protogynous hermaphrodite [7] and widely distributes throughout Asia [8]. It is one of the favorite food fish and emerges as an important freshwater cultured species in China with an annual production of more than 300,000 metric tons. The ricefield eel fries for aquaculture are mainly obtained through catching from wild environments and half artificial reproduction [9]. The natural sex change severely affects the sex ratio and thus decreases the productivity of ricefield eel broodstock. The elucidation of the mechanisms underlying the sex change will aid in developing techniques for controlling sex change of ricefield eels.

Previous studies have shown that the treatment with gonadotropin-releasing hormone analogue of Salmon (sGnRH-A) induced the precocious sex change of ricefield eels from females to males and promote the maturation of males [10]. Recently, many sex-related genes have been revealed to be associated with sex change of ricefield eels, including cyp19a1a [11], dmrt1 [12], foxl2 [13], gsdf [14], 17β-hsd12, dmc1, foxn5, gal3, maats1, spag6, spef2, and zpsbp3 [15]. In addition, epigenetic modifications of cyp19a1a gene [16] and gonadal miRNAs [17] have also been shown to be involved in the sex change of ricefield eels. However, the mechanisms triggering the sex change remain unclear, and the effective ways to manipulate sex change of ricefield eels remain to be developed.

The process of sex change of ricefield eels from females to males could be divided into two phases, namely the initiation of sex change and the subsequent gonadal development towards the testis. The mechanisms that initiate sex change should be activated at the very early stage of the sex transformation process. Thus, obtaining gonadal samples at the early stages of sex change is key to explore the mechanisms underlying the gonadal sex change in ricefield eels. Because the initiation of gonadal sex change was asynchronous and shown to differ in the age, bodyweight, and body length among individual ricefield eels [18], the pooled gonadal sample from different individuals could be a mixture of gonads at different developmental states during sex change and this may hinder the identification of key genes responsible for the process of gonadal transformation. By following the biopsy technique developed previously [19], our present study traced the gonadal transformation process of female ricefield eels, and obtained the gonadal samples at the female, early intersexual, mid-intersexual, and late intersexual stages from the same individual ricefield eel. Ontogenic transcriptomic analysis of these gonadal samples revealed the shutdown expression of cyp19a1a (encoding the gonadal aromatase in teleosts) and decreased expression of foxl2a in gonads at the early intersexual stage. Moreover, some novel genes such as apoeb and enpp2 were shown to be expressed with peak values at the early intersexual stage, which may be potentially involved in the initiation of gonadal sex change. Results of present study may aid in further understanding the underlying mechanisms of the gonadal sex change in ricefield eels as well as other protogynous sex changing teleosts.

Results

Ontogenic gonadal histology of sex changing ricefield eels

Among the thirty-six female ricefield eels biopsied, three (designated as S5, S7, and R10, respectively) were shown to undergo sexual changes with similar developmental pace from female to late intersexual stages during the experiment. Gonadal samples at female (F), early intersexual (EI), mid-intersexual (MI), and late intersexual (LI) stages were obtained from each of the three ricefield eels, respectively. The female gonad contained thin gonadal lamellae with oocytes at the vitellogenic stage (Fig. 1 A and A1). The early intersexual gonad contained thickening gonadal lamellae with oocytes at early growth stages and a few spermatogenic cysts containing SPG or SPC (Fig. 1 B and B1). The mid-intersexual gonad contained a mass of male germ cells including spermatogonia and spermatocytes, and a few oocytes at early growth stages or under degradation (Fig. 1C). The late intersexual gonad contained numerous spermatogenic tubules and a very few degrading oocytes (Fig. 1D). The quality of total RNA isolated from gonadal samples was shown to be suitable for transcriptome sequencing by agarose gel electrophoresis (Additional file 1: Figure S1) and an Agilent 2100 Bioanalyzer (Agilent 2100, Agilent Technologies, USA; the minimum value of RIN after RNA extraction is 7.7).

Fig. 1
figure 1

The histology of gonadal samples obtained from a representative sex changing ricefield eel (S7) through biopsy. A, the gonad at female stage. B, the gonad at the early intersexual stage. C, the gonad at the mid-intersexual stage. D, the gonad at the late intersexual stage. A1 and B1 are higher magnification of the black-line boxed areas in A and B, respectively. The insets within C and D are higher magnification of the corresponding boxed areas. F, female; EI, early intersex; MI, mid-intersex; LI, late intersex; GL, gonadal lamellae; PG, primary growth stage oocyte; EVO, early vitellogenic oocyte; VO, vitellogenic oocyte; DO, degraded oocyte; SPG, spermatogonia; SPC, spermatocyte; ST, spermatid. Scale bar = 100 µm

Quality control parameters of RNA-Seq data

Twelve cDNA libraries were constructed with the total RNA extracted from gonadal tissues of ricefield eels at stages of female (n = 3), early intersex (n = 3), mid-intersex (n = 3), and late intersex (n = 3), respectively. The cDNA libraries were sequenced on an Illumina Hiseq platform and generated a total of 541,302,380 raw reads, which generated 539,764,816 high-quality reads and 80.4 Gb high-quality bases after the removal of adapter sequences and low-quality reads, with more than 37,250,062 high-quality reads and 5.5 Gb high-quality bases from each cDNA library. The percentage of bases with a Phred value of at least 20 (Q20) was higher than 96.78%, and the percentage of bases with a Phred value of at least 30 (Q30) was higher than 91.53% (Supplementary Dataset File: Table S1). Guanine Cytosine (GC) contents were around 51% for all the libraries (Supplementary Dataset File: Table S1). The percentages of high-quality reads that were mapped to the ribosome database ranged from 0.14% to 2.92% (Supplementary Dataset File: Table S2), which were removed. The left high-quality reads were annotated against the ricefield eel (Monopterus albus) reference genome (https://www.ncbi.nlm.nih.gov/genome/24053?genome_assembly_id=302095). An average percentage of exon mapped reads higher than 84.6% was obtained for all the libraires (Supplementary Dataset File: Table S3), which suggests that the reference genome was well-annotated. A total of 23,558 genes were identified in the transcriptome data, of which 20,941 were refer genes and 2,617 were novel genes (Supplementary Dataset File: Table S4 and Table S5).

Differentially expressed genes (DEGs) and enrichment analysis in gonads during sexual change

Analysis of RNA-seq data identified 886, 319 and 10,676 DEGs from pairwise comparisons between F and EI (EI vs. F), EI and MI (MI vs. EI), MI and LI (LI vs. MI), respectively (Fig. 2). Up-regulated and down-regulated genes were 603 and 283 for EI vs. F, 218 and 101 for MI vs. EI, and 8,350 and 2, 326 for LI vs. MI, respectively (Fig. 2A). There were 9,734 DEGs only identified between MI and LI (Fig. 2B), which represents about 89.87% of total DEGs and suggests that dramatic developmental changes occurred in gonads from stage MI to LI.

Fig. 2
figure 2

The histogram and venn diagram of the number of DEGs in transcriptome data of gonads of different sexual stages. A, the histogram shows the number of up-regulated and down-regulated DEGs for the different pairwise comparisons. Y-axis shows the number of DEGs, and X-axis shows the comparison groups. up: up-regulated genes; down: down-regulated genes. B, the venn diagram showing the number of DEGs specific or common to the comparisons. F: female; EI: early intersex; MI: mid-intersex; LI: late intersex

Gene ontology analysis was performed on DEGs for EI vs. F, MI vs. EI, and LI vs. MI, respectively, which showed similar enrichments for all the comparisons (Additional file 1: Figure S2; Supplementary Dataset File: Table S6, S7 and S8). KEGG pathway enrichment analysis showed that PI3K-Akt signaling pathway (ko04151), protein digestion and absorption (ko04974), ECM-receptor interaction (ko04512), cholesterol metabolism (ko04979), and steroid hormone biosynthesis (ko00140) were frequently observed during gonadal transition from the ovary to the mid-intersexual ovotestis. DEGs identified during gonadal transition from the mid- to late intersexual ovotestes were enriched in cytokine-cytokine receptor interaction (ko04060), ribosome (ko03010), hematopoietic cell lineage(ko04640), ECM-receptor interaction (ko04512), and cell adhesion molecules (ko04514) (Fig. 3; Supplementary Dataset File: Table S9, S10 and S11).

Fig. 3
figure 3

The top 20 of KEGG pathways of enrichment in comparisons between gonads at different sexual stages. The gene ratio is the number of DEGs as a percentage of the total number of genes in a KEGG pathway [20]. The color of the plot represents the q-value (negative log10 transformed) of the enriched pathway, and the twenty pathways with minimal q-values were defined as the top 20 pathways. F: female; EI: early intersex; MI: mid-intersex; LI: late intersex

The expression of DEGs for EI vs. F during sex change from female to late intersexual stages

In order to search for genes potentially involved in the initiation of sex change, the expression DEGs for EI vs. F during sex change was further examined. The representative down-regulated DEGs include cyp19a1a (cytochrome P450 aromatase), wnt4a (wingless-type MMTV integration site family, member 4a), fgf16 (fibroblast growth factor 16), foxl2a (forkhead box L2a), pgr (progesterone receptor), and ghrb (growth hormone receptor b). The expression of cyp19a1a mRNA was decreased by 100% at EI, and remained at very low levels from MI to LI with TPM values less than 0.1. The expression of wnt4a mRNA was decreased by 91% at EI, and then remained at similarly low levels thereafter with TPM values less than 0.4. The expression of fgf16 mRNA was decreased by 65% at EI, and then remained at similarly low levels thereafter with TPM values less than 1.3. The expression of foxl2a mRNA was decreased from F to MI by about 88%, and then slightly increased at LI with a TPM value less than 2.5. The expression of pgr and ghrb mRNA was decreased at EI by 96% and 73%, respectively, and then increased from MI to LI (Fig. 4; Supplementary Dataset File: Table S12).

Fig. 4
figure 4

The expression of representative DEGs for EI vs. F in gonads of ricefield eels during sexual change from female to late intersexual stages. Y-axis shows TPM values of genes inferred from the transcriptome data. X-axis shows symbols of genes, and Entrez (NCBI) IDs are 109963333 (cyp19a1a), 109,971,989 (wnt4a), 109,971,151 (fgf16), 109,951,921 (foxl2a), 109,968,475 (pgr), 109,973,650 (ghrb), 109,954,649 (apoeb), 109,967,173 (csl2), 109,973,198 (enpp2), 109,955,915 (mafbb), 109,962,336 (clu), 109,968,774 (igfbp5a), 109,956,374 (sgk1), 109,964,672 (mafba), 109,963,795 (inhbb), 109,951,032 (slc16a1a), 109,959,091 (emb), 109,951,177 (dzip1), 109,956,679 (fsta), and 109,973,748 (flot2). F: female; EI: early intersex; MI: mid-intersex; LI: late intersex. *up-regulated significantly as compared to that at the previous stage; #down-regulated significantly as compared to that at the previous stage

The 603 up-regulated DEGs for EI vs. F (Supplementary Dataset File: Table S13) showed different expression patterns during gonadal sex change. Some DEGs, including apoeb (apolipoprotein Eb), csl2 (L-rhamnose-binding lectin SML-like), and enpp2 (ectonucleotide pyrophosphatase/phosphodiesterase 2), were upregulated at EI by about 114-, 30-, and 14.5-fold respectively as compared to that at F, and then their expression decreased significantly thereafter. Some DEGs, including mafbb (v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog Bb), clu (clusterin), igfbp5a (insulin-like growth factor binding protein 5a), sgk1(serum/glucocorticoid regulated kinase 1), and mafba (v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog Ba), were upregulated at EI by about 28-, 15-,13-, 8-, and 7-fold respectively as compared to that at F, then their expression decreased significantly at MI but increased again at LI to a similar level as that at EI. Other DEGs, including inhbb (inhibin subunit beta B), slc16a1a (monocarboxylate transporter 1), emb (embigin), dzip1(zinc finger protein Dzip1-like), fsta (follistatin a), and flot2 (flotillin-2), were increased at EI by about 31-,18-, 15-, 13-, 12-, and 4-fold, respectively as compared to that at F, then their expression remained at a similar level at MI but increased significantly at LI (Fig. 4).

The expression of genes encoding receptors for gonadotropin-releasing hormones, gonadotropins, and sex steroids in gonads during sex change

The expression of gnrhr2 (gonadotropin-releasing hormone II receptor-like) seemed to be higher than those of gnrhr1 (gonadotropin-releasing hormone II receptor-like) in gonads at all stages examined, but neither of them showed significant variation during sex change from female to late intersexual stages (Fig. 5). Both gonadotropin receptor genes lhr (luteinizing hormone/choriogonadotropin receptor) and fshr (follicle stimulating hormone receptor) showed similar expression profiles during sex change, with slightly decreased expression levels at EI and MI as compared to those at F but up-regulated significantly at LI. The expression of two esr2 genes (esr2a for estrogen receptor 2a and esr2b for estrogen receptor 2b) seemed to be higher than those of esr1 (estrogen receptor 1) gene in gonads at all stages examined. The expression of esr1 was up-regulated significantly at LI while the expression of both esr2a and esr2b was only slightly increased at LI during sex change. The expression of ar (androgen receptor) was slightly decreased at EI and MI but then up-regulated significantly at LI.

Fig. 5
figure 5

The expression of genes encoding receptors for gonadotropin-releasing hormones, gonadotropins, and sex steroids in gonads of ricefield eels during sex change. Y-axis shows TPM values of genes inferred from the transcriptome data. X-axis shows symbols of genes, and Entrez (NCBI) IDs are 109,971,570 (gnrhr1), 109,970,856 (gnrhr2), 109,961,972 (lhr), 109,962,940 (fshr), 109,962,316 (esr2a), 109,966,606 (esr2b), 109,952,736 (esr1), and 109,959,413 (ar). F: female; EI: early intersexual; MI: mid-intersexual; LI: late intersexual. *up-regulated significantly as compared to that at the previous stage

Quantitative real-time PCR (qPCR) validation of gene expression inferred from the transcriptome data

qPCR was performed to validate the expression patterns of 12 transcripts inferred from the transcriptome data, which are presumably associated with sex differentiation (Fig. 6). The female-related genes, including cyp19a1a, wnt4a, and foxl2a, were highly expressed at F, but down-regulated during sex change from MI to LI stages. The male-related genes, including sox9, dmrt1a, amh, and spef2 (sperm flagellar protein 2), showed low expression levels from F to MI stages but were all highly expressed at LI. Some other genes also showed significant variations during sex change. The expression of foxl2l was low from F to MI stages, but increased significantly at LI. The expression of apoeb and g0s2 (G0/G1 switch protein 2) was high at EI but low at the other three stages. igfbp5a and mafbb were expressed with high abundances at EI and LI but with low abundances at F and MI. The expression profiles of the 12 transcripts as revealed by qPCR analysis were basically similar to those inferred from the transcriptome data (Fig. 6), implying that the analysis of gene expression through RNA-seq data in the present study is reliable.

Fig. 6
figure 6

Quantitative real-time PCR (qPCR) validation of the expression of DEGs inferred from the transcriptome data. The left Y-axis and solid bars show mRNA levels by qPCR, and the right Y-axis and empty bars show TPM values inferred from the transcriptome data. The solid bar represents mean ± SEM (n = 3). Different letters above the bars indicate significant differences (p < 0.05). One-way analysis of variance, followed by Duncan multiple comparison test, was used to estimate significant differences in gene expression across stages except apoeb and amh, for which Dunnett T3 was used for the multiple comparison test due to the heterogeneity of data. F, female; EI, early intersex; MI, mid-intersex; LI, late intersex

Discussion

The ricefield eel, a protogynous hermaphrodite teleost [7, 21], is considered as a model system for vertebrate sexual development [22]. Although many studies have been aimed to identify factors contributing to the sex changing phenomenon of ricefield eels, the underlying mechanisms remain elusive [23]. In the present study, transcriptomic analysis was applied to gonadal samples encompassing the sex changing process from female to late intersexual stages obtained from the same individual ricefield eel through biopsy, which, we believe, is more informative for exploring genes and regulatory pathways related to the sexual change because the genetic backgrounds of the samples are more uniform. The gonadal sexual stages of collected gonadal tissues from individual ricefield eels were carefully verified by histological analysis, which are designated as F (female stage), EI (early intersexual stage), MI (mid-intersexual stage), and LI (late intersexual stage). The assembly of transcriptome data from gonadal cDNA libraries of ricefield eels in the present study resulted in a total of 23,558 genes, of which 20,941 genes were mapped to Monopterus albus genome database and 2,617 genes are novel ones. The gene number were comparable to the genome database of Monopterus albus updated recently (24,116 genes; https://www.ncbi.nlm.nih.gov/gene/?term=monopterus%20albus), which suggests that the quality of the present transcriptome data is pretty good.

The differential expression of genes along with the gonadal transitional process was analyzed in the following three pairwise comparisons, namely EI vs. F, MI vs. EI, and LI vs. MI. The expression profiles of the selected representative genes across stages inferred from transcriptomic data were similar to those obtained with qPCR, suggesting that transcriptome-based gene expression analysis is essentially reliable. There were about 1.17% of DEGs only identified in the comparisons of EI vs F and MI vs EI. In contrast, about 89.87% of DEGs were identified only in the comparison of LI vs MI, which implies that dramatic developmental changes occurred in gonads from stage MI to LI, and these changes are presumably related to testicular development in ricefield eels.

In protogynous hermaphroditic fish, the decreases of E2 and cyp19a1a expression were suggested to play essential roles in the sex change from female to male [24,25,26,27,28]. In ricefield eels, similarly, the expression of cyp19a1a in gonads was down-regulated from female to male [11, 29]. In support of the above reports, data of the present study indicated that the expression of cyp19a1a in gonads was totally blocked at the initial stage of sex change, and remained at very low levels thereafter with TPM below 0.1. These lines of evidence suggest an important role for the down-regulation of cyp19a1a in the initiation of sex change in ricefield eels. It has been shown that foxl2 up-regulates the transcription of cyp19a1a of ricefield eels [30] and other teleosts, and plays important roles in the ovarian differentiation [26]. The present transcriptomic data showed that the expression of foxl2a was decreased from the female to the mid-intersexual stage. Taken together, these data suggests that the decrease in foxl2a expression during sex change may lead to the down-regulation of cyp19a1a and contribute to the initiation of sex change in ricefield eels. Dmrt1, a conserved factor essential to testicular development in vertebrates [31], has been demonstrated to inhibit the Ad4BP/SF-1 (Nr5a1a)-activated cyp19a1a transcription [32]. Similarly, Dmrt1a was also shown to inhibit Foxl2a-induced activation of ricefield eel cyp19a1a promoter (unpublished results). However, whether Dmrt1a contributes to the initiation of sex change through inhibition of cyp19a1a or not remains to be elucidated because the expression of dmrt1a was increased slightly at the early intersexual stage, and the cellular co-localization of this increased dmrt1a mRNA with cyp19a1a mRNA needs to be confirmed.

In contrast to foxl2a, the expression of the duplicated paralogue foxl2l was very low at the female stage, but then increased slightly at the early intersexual stage and significantly at the late intersexual stage. Previously, ricefield eel foxl2l was named as foxl3, and its expression was shown to be male-biased [33] and barely detectable in ovarian follicles [34]. These lines of evidence support a major role for foxl2l in testicular development of ricefield eels [33]. In the bluehead wrasse, a protogynous teleost, the duplicates of foxl2 also showed contrasting sex-specific expression patterns [35]. Thus Foxl2l of ricefield eel may acquire a novel function after duplication possibly through neofunctionalization during evolution [36].

The Wnt/β-catenin pathway is regarded as a major player in ovarian differentiation in mammals [37], and also possibly involved in the differentiation of the ovary in rainbow trout [38]. The present transcriptomic data showed higher expression of wnt4a in the ovary than the gonads of other sexual stages in ricefield eels, which is in line with the major roles of wnt4 in the ovarian differentiation and maintenance. Fibroblast growth factors (FGF) also play important roles in sex determination and sex differentiation in vertebrates. In mammals, Fgf9 has been shown to downregulate Wnt4 to establish the testis pathway in the XY gonad [39]. In teleosts, Fgf homologues other than Fgf9 were identified [40, 41]. In tilapia, fgf16 and fgf20b are highly expressed both in normal XX (genetically female) and sexually changed XY (genetically male) ovaries induced by E2, but are low in normal XY testis and AI-induced XX testis, suggesting that fgf16 and fgf20b may play roles in the differentiation of ovaries [42]. Similarly in our present study, fgf16 was expressed at a higher level at the female stage and down-regulated at EI through LI, implying a major role for fgf16 in ovarian development of ricefield eels. These lines of evidence suggest that roles of Fgf homologues in gonadal differentiation may be different between mammals and teleosts. Together with the down-regulation of cyp19a1a, our present study implied that the inhibition of female pathways is likely a pre-requisite for sexual change in ricefield eels.

In addition to the down-regulated DEGs, the present transcriptomic data identified 603 DEGs that were up-regulated at EI as compared to F. These up-regulated DEGs are presumably closely associated with sex change, and their different expression profiles are suggestive of differential roles in the gonadal transformation process.

Among the 603 upregulated DEGs, the expression of apoeb, csl2, and enpp2 was dramatically increased and peaked at EI, but then decreased significantly thereafter. The gene apoeb encodes an apolipoprotein E-like protein. It has been shown that in rats, the greatest apolipoprotein E mRNA was expressed in theca cells of atretic follicles, and exogenous apolipoprotein E induced apoptosis of cultured ovarian theca and interstitial cells [43]. The gene csl2 encodes a L-rhamnose-binding lectin SML-like protein. Lectins are a group of sugar-binding proteins that can stimulate cell proliferation [44]. The gene enpp2 encodes the ectonucleotide pyrophosphatase/phosphodiesterase 2, which catalyzes the synthesis of lysophosphatidic acid and affects proliferation, cell survival, motility, morphological rearrangements and differentiation [45]. These lines of evidence imply that the upregulation of apoeb, csl2, and enpp2 at EI may be predominantly associated with the initiation of sex change in ricefield eels, possibly through apoptosis of ovarian follicular cells, and proliferation and/or differentiation of testicular somatic and germ cells. Their physiological roles and action mechanisms in sex change of ricefield eels are worth further study.

The expression of mafba, clu, mafbb, igfbp5a, and sgk1 showed similar biphasic patterns during sex change of ricefield eels, with two peaks observed at EI and LI, respectively. Interestingly, Mafb is a putative marker for fetal Leydig cell progenitors, and expressed in the testis at the earliest stages of interstitial development in mice [46]. Clusterin (CLU) is a glycoprotein with ubiquitous expression in various tissues and body fluids that acts as an ATP-independent extracellular molecular chaperone. CLU overexpression effectively enhanced spermatogenic cell viability and suppressed high glucose-induced apoptosis [47]. The insulin-like growth factor-binding protein-5 (IGFBP-5) is one of the 6 members of the IGFBP family, and is involved in the regulation of cell growth, apoptosis, and other IGF-stimulated signaling pathways [48]. SGK1 (serum and glucocorticoid-inducible kinase 1) is a growth factor-responsive kinase, which has been shown to promote vascular smooth muscle cell proliferation [49]. These lines of evidence indicated that in ricefield eels, mafba, clu, mafbb, igfbp5a, and sgk1 may be associated with the initiation of sexual change and subsequent gonadal development towards testis, possibly through the regulation of follicular atresia and the differentiation and proliferation of male germ and somatic cells including the Leydig cell. Their physiological roles and mechanisms need further study.

The expression of dzip1, flot2, emb, fsta, inhbb, and mot1 was continuously increased during sex change, with peaks at LI. The human DAZ (Deleted in Azoospermia) genes have been shown to be essential to spermatogenesis [50]. The gene DZIP1 encodes a DAZ-interacting protein, which is expressed in male germ cells in human, mouse, and rat [51, 52]. Flot2 gene encodes the protein flotillin 2, which has been shown to be expressed at high levels in male germ cells during mouse spermatogenesis, and be involved in sperm acrosome biogenesis [53]. It is possible that in ricefield eels, both Dzip1 and Flot2 may also play roles in male germ cell development during sex change from female to male.

Follistatin, encoded by fst, is an antagonist of activin and can affect the reproductive ability by inhibiting the action of activin [54]. Studies suggested that a reduction in the level of activin signaling may be required for gonocytes to differentiate into spermatogonia in rat testes [55]. Whether the up-regulation of fsta during sex change from female to male in ricefield eels is also be associated with the differentiation of spermatogonia is worth further study.

Inhibin, a member of TGF-β superfamily, can act on gonads indirectly through pituitary or directly as a paracrine factor [56]. Inhibin consists of two covalently linked subunits, a common alpha subunit and a beta subunit, the latter of which exists in two forms, A and B (denoted beta A and beta B, respectively). In the male, inhibin B is produced principally by the Sertoli cells in the testis [57]. The expression pattern of inhbb in gonads of ricefield eels during sex change probably reflects the differentiation and proliferation of Sertoli cells for the testicular development.

It has been established that developing male germ cells receive an adequate level of energy substrates, preferentially lactate which is produced by the Sertoli cells and transported to germ cells via monocarboxylate transporters (MCTs) including MCT1 [58]. Conditional knockout of Mct1 (now renamed as Slc16a1a) resulted in total absence of spermatozoa and morphological alterations of Sertoli cells [59]. The correct expression, localization, and activity of MCTs depend on interactions with chaperon proteins such as embigin [60]. The parallel increases in the expression of slc16a1a and emb during sex change from female to the late intersexual stage suggest that both monocarboxylate transporter 1 and embigin may interact to support the spermatogenesis in ricefield eels, possibly through the transfer of lactate from Sertoli cells to germ cells.

Hypothalamic and pituitary hormonal signals were also implicated in the regulation of gonadal sex change in hermaphrodite teleosts, including bluehead wrasse [61, 62], honeycomb grouper [63], and the orange-spotted grouper [64]. In ricefield eels, the precocious sexual change could be induced by the administration of gonadotropin-releasing hormone analogue (sGnRH-A) [10, 65] and ovine-luteinizing hormone (oLH) [66]. Due to the presence of GnRH receptors in the pituitary [67] as well as in the gonad [68], the exogenous GnRH analogue could induce precocious sex change either through indirect actions on the pituitary or direct actions on the gonad. The transcriptomic data of the present study showed that the abundance of either gnrhr1 or gnrhr2 did not vary significantly across stages during sex change. In contrast, the abundances of both lhr and fshr were significantly increased at the late intersexual stage. These lines of evidence favor that GnRH signals most likely induce sex change of ricefield eels indirectly through acting on the pituitary by increasing the synthesis and release of Lh and/or Fsh. As the increases in lhr and fshr mRNA were significant only at the late intersexual stage rather than the early intersexual stage, these signals presumably mainly contribute to the progression of sex change towards testis from ovotestis but probably not the initiation of sex change.

In contrast to cyp19a1a, transcriptomic data showed increased expression of genes encoding estrogen nuclear receptors including esr1, esr2a, and esr2b during natural sex change from female to the late intersexual stage, particularly the esr1. Similarly, significant increases in the expression of gonadal estrogen receptors (esr1, esr2a, and esr2b) were observed in the testis of sex-changed juvenile ricefield eels induced by AI treatment [69]. In channel catfish (Ictalurus punctatus), esr1 and esr2 genes were co-expressed in the mature testis and seemed to be developmentally regulated in spermatocytes [70]. In goldfish (Carassius auratus), both ovary and testis had high transcript levels of esr2a [71]. Interestingly, exogenous estradiol-17b (E2) induced spermatogonial stem cell division in cultured testicular tissue of Japanese eel (Anguilla japonica) [72]. These lines of evidence suggest that estrogen and/or estrogen receptors may also play significant roles in the development of testis in ricefield eels as in other teleosts.

Conclusions

Collectively, data of the present study suggest that initiation of sex change in ricefield eels may require the inhibition of female-related genes, particularly the complete shutdown of cyp19a1a, and concurrent activation of some genes like apoeb and enpp2, etc. Subsequently, progress of sex change from intersexual towards male stages requires up-regulation of male-related genes like dmrt1 and amh, etc. Further studies on the mechanisms of cyp19a1a downregulation and the physiological roles and action mechanisms of Apoeb and Enpp2 may help to elucidate the molecular mechanisms underlying the natural sex change of ricefield eels.

Materials and methods

Experimental fish and gonadal samples for ontogenic transcriptome analysis

Thirty-six female ricefield eels (body length 29.2 ± 3.4 cm and body weight 20.8 ± 6.7 g) were raised in four indoor concrete ponds (100 × 100 × 90 cm) under natural photoperiods and temperatures from March to April of 2020 in Guangzhou, China, with 9 fish each pond. The ricefield eels were fed with live Tenebrio molitor at the daily ration of 5% bodyweight. Serial gonadal samples were obtained through biopsy from each individual fish to trace its gonadal development and sexual change. The biopsy operation was performed by following a previously reported method [19] with minor modifications. Briefly, the fish were first anesthetized with MS-222 (500 mg/L), and then a small incision of about 0.5-1 cm length was made on the right side of ventral body wall ahead of the cloacal orifice of fish. The gonad was identified and separated from the mesentery and the dorsal-intestinal artery with great care to avoid any major injury. A small portion of gonadal tissue was dissected out and cut into two parts. One part was snap frozen in liquid nitrogen for the subsequent extraction of tissue RNA, and the other part was fixed in Bouin solution for histological examination of gonadal stages. After cutting out the gonadal tissue, a unique electronic tag (HT950; Hongteng Barcode Technology Co., Ltd., Guangzhou, China) was implanted into the abdomen for identifying each individual fish. The incision was then sutured and erythromycin ointment was applied to the wound to prevent potential inflammation. The biopsied fish were put in aerated fresh water to recover before returning to the pond. A total of 4 biopsies were made on each individual fish at an interval of about 4 weeks. All procedures and investigations followed the ARRIVE guidelines [73], and were reviewed and approved by the Center for Laboratory Animals of Sun Yat-sen University.

The Bouin-fixed gonadal samples were dehydrated and embedded in paraffin through conventional histological processing. Serial cross sections were cut at 4–5 μm and stained with haematoxylin/eosin. The gonadal sections were examined and photographed with a Nikon DS-Ri2 digital camera on a Nikon E800 microscope (Nikon, Japan). Six of the thirty-six female ricefield eels have undergone sex change during the experiment [18], and from 3 of them serial gonadal samples at the female (F), early intersexual (EI), mid-intersexual (MI), and late intersexual (LI) stages were obtained.

RNA extraction from gonadal tissues

Total RNA was isolated from serial gonadal samples of the three sex changing fish using Trizol reagent (Invitrogen, USA), and resuspended in RNase-free water. RNA quality was checked with agarose gel electrophoresis and further assessed on an Agilent 2100 Bioanalyser (Agilent Technologies, Palo Alto, CA, USA) before RNA-seq.

cDNA library construction and sequencing

The construction of cDNA libraries and sequencing were performed essentially as previously described [34]. Briefly, a total amount of 1 μg RNA per sample was used as input material for RNA sequencing. The sequencing library was generated using a NEBNext® Ultra™ RNA Library Prep Kit for Illumina® (#E7530L, NEB, USA) by following the manufacturer’s recommendation. Briefly, mRNA was enriched from total RNA using Oligo (dT) magnetic beads, and fragmented in NEBNext First Strand Synthesis Reaction Buffer (5 ×) by ultrasound. First strand cDNA was synthesized with ProtoScript II Reverse Transcriptase using NEBNext Random Primers. Second-strand cDNAs were synthesized with the Second Strand Synthesis Enzyme Mix in the Second Strand Synthesis Reaction Buffer. Then double-stranded cDNA fragments were purified with 1.8X Agencourt AMPure XP Beads (NEB), end repaired, and ligated to Illumina sequencing adapters. The ligation products were purified with AMPure XP Beads, PCR amplified, and sequenced by Gene Denovo Biotechnology Co. (Guangzhou, China) using Illumina HiSeq 2500 to generate 150 bp paired-end reads.

Data filtering and annotation

To get high-quality reads, raw data obtained from the sequencing machine were further filtered by fastp (version 0.18.0) to remove reads containing adapters or containing more than 10% of unknown nucleotides (N), and low-quality reads containing more than 50% of low-quality bases (Q-value ≤ 20). At the same time, Q20, Q30, GC content, and the sequence duplication levels of the high-quality data were calculated. All the downstream analysis was based on data of high quality. Short reads alignment tool Bowtie2 [74] (version 2.2.8) was used to map reads to the ribosome RNA (rRNA) database of ricefield eels, and the rRNA mapped reads were removed for gene abundance calculation. An index of the reference genome was built, and paired-end high-quality reads were mapped to the ricefield eel M. albus genome database (https://www.Ncbi.nlm.nih.gov/genome/24053?genome_assembly_id=302095), using HISAT2.2.4 [75] with “RNA—strandness RF” and other parameters set as default. There were some transcripts that were aligned to the unannotated regions in the genome database of ricefield eel, and these transcripts were called novel genes in the present study. Then the novel genes were further aligned by using BLASTx program (http://www.ncbi.nlm.nih.gov/BLAST/) with an E-value threshold of 1e-5 to the NCBI non-redundant protein (Nr) database (http://www.ncbi.nlm.nih.gov), the Swiss-Prot protein database (http://www.expasy.ch/sprot), the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (http://www.genome.jp/kegg), and the Gene Ontology database (http://www.geneontology.org/). Protein functional annotations could then be obtained for the novel genes according to the best alignment results.

Gene expression analysis

The mapped reads of each sample were assembled by using StringTie v1.3.1 [76, 77] in a reference-based approach. For each transcription region, a TPM (transcripts per million) value [78] was calculated to quantify its expression abundance and variations using String Tie software.

A differential gene expression analysis between pairs of samples (F vs. EI, EI vs. MI, MI vs. LI) of three fish was performed by using GLM with negative binomial distribution under paired statistical model in edgeR [79]. The genes/transcripts with the parameter of false discovery rate (FDR) below 0.05 and absolute fold change ≥ 2 were considered to be differentially expressed. All differentially expressed genes (DEGs) were mapped to GO terms in the Gene Ontology database (http://www.geneontology.org/), and gene numbers were calculated for every term. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used to interpret the biological functions of differentially expressed genes. The significantly enriched GO terms and KEGG pathways in differentially expressed genes compared to the genome background were defined by hypergeometric test. The calculated p-value of both enrichment analyses was gone through FDR correction, taking FDR (Q-value) ≤ 0.05 as a threshold. GO terms or KEGG pathways meeting this criterion were defined as significantly enriched in DEGs.

Validation of differentially expressed genes from transcriptomes with real-time quantitative PCR

The relative mRNA levels of 12 differentially expressed genes identified from transcriptomes were examined by real-time quantitative PCR (qPCR) to verify their expression profiles in ricefield eel gonads during sexual change from female to late intersexual stages. The main purpose of analyzing the expression of these 12 genes by qPCR was to test if their expression profiles parallel with those inferred from RNA-seq data. Three biological replicates were employed for each stage and processed similarly as reported [34]. Briefly, total RNA (1 μg) from each sample was first treated with RNase-free DNase I and then reverse transcribed with random primers by using the RevertAid H Minus First Strand cDNA Synthesis Kit (Fermentas, Vilnius, Lithuania) according to the manufacturer’s instruction. The qPCR reaction was performed on the Roche Light Cycler 480 in a reaction volume of 10 μl containing 0.3 µM of each primer, 1 µL of cDNA template, and 5 µL of 2 × Bestar® SYBR Green qPCR Master Mix (DBI® Bioscience, Germany). The oligonucleotide primers (Table 1) were designed to span exons when possible. The cycling parameters were: 95 °C for 5 min, followed by 40 cycles of amplification at 95 °C for 10 s, 58 °C for 15 s and 72 °C for 20 s. The specificity of qPCR amplification was confirmed by melt-curve analysis, agarose gel electrophoresis, and sequencing of PCR products.

Table 1 Sequences of primers used for real-time quantitative PCR analysis

The quantification of the mRNA expression level was conducted as previously described [80], using a standard curve with tenfold serial dilutions of the plasmid containing the corresponding cDNA fragment which ranges from 101 to 109 copies. The correlation coefficients were not less than 0.98, and the PCR efficiencies ranged from 1.9 to 2.0. To minimize variation due to the differences in RNA loading, the geometric mean expression levels of actb, gapdh, and hprt1 were used to normalize the expression levels of the target genes. One-way analysis of variance, followed by the Duncan multiple comparison test, was used to estimate significant differences (p < 0.05) in gene expression across sexual stages except apoeb and amh, for which Dunnett T3 was used for the multiple comparison test due to the heterogeneity of data. 

Availability of data and materials

The Illumina HiSeq 2500 was used to generate transcriptomic data on gonadal samples. All data were deposited at the NCBI under Bioproject PRJNA764057 and are publicly available (https://www.ncbi.nlm.nih.gov/search/all/?term = PRJNA764057).

References

  1. Capel B. Vertebrate sex determination: evolutionary plasticity of a fundamental switch. Nat Rev Genet. 2017;18(11):675–89.

    Article  CAS  PubMed  Google Scholar 

  2. DeFalco T, Capel B. Gonad morphogenesis in vertebrates: divergent means to a convergent end. Annu Rev Cell Dev Biol. 2009;25:457–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Avise JC, Mank JE. Evolutionary perspectives on hermaphroditism in fishes. Sexual Development. 2009;3(2–3):152–63.

    Article  CAS  PubMed  Google Scholar 

  4. Erisman BE, Petersen CW, Hastings PA, Warner RR. Phylogenetic perspectives on the evolution of functional hermaphroditism in teleost fishes. Integr Comp Biol. 2013;53(4):736–54.

    Article  PubMed  Google Scholar 

  5. Gemmell NJ, Todd EV, Goikoetxea A, Ortega-Recalde O, Hore TA. Natural sex change in fish. Curr Top Dev Biol. 2019;134:71–117.

    Article  PubMed  Google Scholar 

  6. Todd EV, Liu H, Muncaster S, Gemmell NJ. Bending genders: the biology of natural sex change in fish. Sexual Development. 2016;10(5–6):223–41.

    Article  CAS  PubMed  Google Scholar 

  7. Liu CK. Rudimentary hermaphroditism in the symbranchoid eel Monopterus javanensis. Sinensia. 1944;15:1–8.

    Google Scholar 

  8. Supiwong W, Pinthong K, Seetapan K, Saenjundaeng P, Bertollo LAC, de Oliveira EA, et al. Karyotype diversity and evolutionary trends in the Asian swamp eel Monopterus albus (Synbranchiformes, Synbranchidae): a case of chromosomal speciation? BMC Evol Biol. 2019;19(1):73.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Feng K, Luo H, Li Y, Chen J, Wang Y, Sun Y, et al. High efficient gene targeting in rice field eel Monopterus albus by transcription activator-like effector nucleases. Sci Bull. 2017;62(3):162–4.

    Article  CAS  Google Scholar 

  10. Tao YX, Lin HR, Glen VDK, Richard EP. Hormonal induction of precocious sex reversal in the ricefield eel Monopterus albus. Aquaculture. 1993;118:131–40.

    Article  CAS  Google Scholar 

  11. Zhang Y, Zhang W, Yang H, Zhou W, Hu C, Zhang L. Two cytochrome P450 aromatase genes in the hermaphrodite ricefield eel Monopterus albus: mRNA expression during ovarian development and sex change. J Endocrinol. 2008;199(2):317–31.

    Article  CAS  PubMed  Google Scholar 

  12. Huang X, Guo Y, Shui Y, Gao S, Yu H, Cheng H, et al. Multiple alternative splicing and differential expression of dmrt1 during gonad transformation of the rice field eel. Biol Reprod. 2005;73(5):1017–24.

    Article  CAS  PubMed  Google Scholar 

  13. Hu Q, Guo W, Gao Y, Tang R, Li D. Molecular cloning and analysis of gonadal expression of Foxl2 in the rice-field eel Monopterus albus. Sci Rep. 2014;4:6884.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhu Y, Wang C, Chen X, Guan G. Identification of gonadal soma-derived factor involvement in Monopterus albus (protogynous rice field eel) sex change. Mol Biol Rep. 2016;43(7):629–37.

    Article  CAS  PubMed  Google Scholar 

  15. Chi W, Gao Y, Hu Q, Guo W, Li D. Genome-wide analysis of brain and gonad transcripts reveals changes of key sex reversal-related genes expression and signaling pathways in three stages of Monopterus albus. PLoS ONE. 2017;12(3):e0173974.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zhang Y, Zhang S, Liu Z, Zhang L, Zhang W. Epigenetic modifications during sex change repress gonadotropin stimulation of cyp19a1a in a teleost ricefield eel (Monopterus albus). Endocrinology. 2013;154(8):2881–90.

    Article  CAS  PubMed  Google Scholar 

  17. Gao Y, Guo W, Hu Q, Zou M, Tang R, Chi W, et al. Characterization and differential expression patterns of conserved microRNAs and mRNAs in three genders of the rice field eel (Monopterus albus). Sexual Development. 2014;8(6):387–98.

    Article  CAS  PubMed  Google Scholar 

  18. Fan M, Yang W, Sun S, Li Z, Zhang LH, Zhang WM. Natural sex change of the “virgin” female ricefield eel Monopterus albus. Acta Hydrobiol Sin. 2021;45(2):387–96.

    Google Scholar 

  19. Chan STH, Wai-Sum O, Tang F, Lofts B. Biopsy studies on the natural sex reversal in Monopterus albus. Zool J Linn Soc. 1972;167:415–21.

    Article  Google Scholar 

  20. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Liem KF. Sex reversal as a natural process in the synbranchiform fish Monopterus albus. Copeia. 1963;2:303–12.

    Article  Google Scholar 

  22. Cheng H, Guo Y, Yu Q, Zhou R. The rice field eel as a model system for vertebrate sexual development. Cytogenet Genome Res. 2003;101(3–4):274–7.

    Article  CAS  PubMed  Google Scholar 

  23. Hilles AR, Hashim R, Mahmood S. The secret behind the natural sex reversal in ricefield eel (Monopterus albus) remains unknown. Advances in Complementary & Alternative medicine. 2018;2(2):1–2.

    Google Scholar 

  24. Kokokiris L, Fostier A, Athanassopoulou F, Petridis D, Kentouri M. Gonadal changes and blood sex steroids levels during natural sex inversion in the protogynous Mediterranean red porgy, Pagrus pagrus (Teleostei: Sparidae). Gen Comp Endocrinol. 2006;149(1):42–8.

    Article  CAS  PubMed  Google Scholar 

  25. Nakamura M, Hourigan TF, Yamauchi K, Nagahama Y, Grau EG. Histological and ultrastructural evidence for the role of gonadal steroid hormones in sex change in the protogynous wrasse Thalassoma duperrey. Environ Biol Fishes. 1989;24(2):117–36.

    Article  Google Scholar 

  26. Guiguen Y, Fostier A, Piferrer F, Chang CF. Ovarian aromatase and estrogens: a pivotal role for gonadal sex differentiation and sex change in fish. Gen Comp Endocrinol. 2010;165(3):352–66.

    Article  CAS  PubMed  Google Scholar 

  27. Wang DS, Kobayashi T, Zhou LY, Paul-Prasanth B, Ijiri S, Sakai F, et al. 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(3):712–25.

    Article  CAS  PubMed  Google Scholar 

  28. Bhandari RK, Komuro H, Higa M, Nakamura M. Sex inversion of sexually immature honeycomb grouper (Epinephelus merra) by aromatase inhibitor. Zoolog Sci. 2004;21(3):305–10.

    Article  CAS  PubMed  Google Scholar 

  29. Liu JF, Guiguen Y, Liu SJ. Aromatase (P450arom) and 11beta-hydroxylase (P45011beta) genes are differentially expressed during the sex change process of the protogynous rice field eel Monopterus albus. Fish Physiol Biochem. 2009;35(3):511–8.

    Article  CAS  PubMed  Google Scholar 

  30. Yan T, Lu H, Sun C, Peng Y, Meng F, Gan R, et al. Nr5a homologues in the ricefield eel Monopterus albus: Alternative splicing, tissue-specific expression, and differential roles on the activation of cyp19a1a promoter in vitro. Gen Comp Endocrinol. 2021;312:113871.

    Article  CAS  PubMed  Google Scholar 

  31. Matson CK, Murphy MW, Sarver AL, Griswold MD, Bardwell VJ, Zarkower D. DMRT1 prevents female reprogramming in the postnatal mammalian testis. Nature. 2011;476(7358):101–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang DS, Zhou LY, Kobayashi T, Matsuda M, Shibata Y, Sakai F, et al. Doublesex- and Mab-3-related transcription factor-1 repression of aromatase transcription, a possible mechanism favoring the male pathway in tilapia. Endocrinology. 2010;151(3):1331–40.

    Article  CAS  PubMed  Google Scholar 

  33. Gao Y, Jia D, Hu Q, Li D. Foxl3, a Target of miR-9, Stimulates Spermatogenesis in Spermatogonia During Natural Sex Change in Monopterus albus. Endocrinology. 2016;157(11):4388–99.

    Article  PubMed  Google Scholar 

  34. Meng FY, Sun S, Xu X, Yu WX, Gan RP, Zhang LH, et al. Transcriptomic analysis provides insights into the growth and maturation of ovarian follicles in the ricefield eel (Monopterus albus). Aquaculture. 2022;555:738251.

    Article  CAS  Google Scholar 

  35. Todd EV, Ortega-Recalde O, Liu H, Lamm MS, Rutherford KM, Cross H, et al. Stress, novel sex genes, and epigenetic reprogramming orchestrate socially controlled sex change. Sci Adv. 2019;5(7):eaaw7006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Glasauer SM, Neuhauss SC. Whole-genome duplication in teleost fishes and its evolutionary consequences. Mol Genet Genomics. 2014;289(6):1045–60.

    Article  CAS  PubMed  Google Scholar 

  37. Chassot AA, Ranc F, Gregoire EP, Roepers-Gajadien HL, Taketo MM, Camerino G, et al. Activation of beta-catenin signaling by Rspo1 controls differentiation of the mammalian ovary. Hum Mol Genet. 2008;17(9):1264–77.

    Article  CAS  PubMed  Google Scholar 

  38. Nicol B, Guiguen Y. Expression profiling of Wnt signaling genes during gonadal differentiation and gametogenesis in rainbow trout. Sexual Development. 2011;5(6):318–29.

    Article  CAS  PubMed  Google Scholar 

  39. Kim Y, Kobayashi A, Sekido R, DiNapoli L, Brennan J, Chaboissier MC, et al. Fgf9 and Wnt4 act as antagonistic signals to regulate mammalian sex determination. PLoS Biol. 2006;4(6):e187.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Watanabe A, Kobayashi E, Ogawa T, Onitake K. Fibroblast growth factor may regulate the initiation of oocyte growth in the developing ovary of the Medaka Oryzias latipes. Zoological Science. 1998;15(4):531–6.

    CAS  PubMed  Google Scholar 

  41. Whitehead GG, Makino S, Lien CL, Keating MT. FGF20 is essential for initiating zebrafish fin regeneration. Science. 2005;310:1957–60.

    Article  CAS  PubMed  Google Scholar 

  42. Sun YL, Zeng S, Ye K, Yang C, Li MH, Huang BF, et al. Involvement of FGF9/16/20 subfamily in female germ cell development of the Nile tilapia Oreochromis niloticus. Fish Physiol Biochem. 2012;38(5):1427–39.

    Article  CAS  PubMed  Google Scholar 

  43. Zerbinatti CV, Mayer LP, Audet RG, Dyer CA. Apolipoprotein E is a putative autocrine regulator of the rat ovarian theca cell compartment. Biol Reprod. 2001;64(4):1080–9.

    Article  CAS  PubMed  Google Scholar 

  44. Carvalho E, Oliveira WF, Coelho L, Correia MTS. Lectins as mitosis stimulating factors: Briefly reviewed. Life Sci. 2018;207:152–7.

    Article  CAS  PubMed  Google Scholar 

  45. Frisca F, Colquhoun D, Goldshmit Y, Anko ML, Pebay A, Kaslin J. Role of ectonucleotide pyrophosphatase/phosphodiesterase 2 in the midline axis formation of zebrafish. Sci Rep. 2016;6:37678.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. DeFalco T, Takahashi S, Capel B. Two distinct origins for Leydig cell progenitors in the fetal testis. Dev Biol. 2011;352(1):14–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tian Y, Xiao YH, Geng T, Sun C, Gu J, Tang KF, et al. Clusterin suppresses spermatogenic cell apoptosis to alleviate diabetes-induced testicular damage by inhibiting autophagy via the PI3K/AKT/mTOR axis. Biol Cell. 2021;113(1):14–27.

    Article  CAS  PubMed  Google Scholar 

  48. Wang XJ, Su HM, Liang Y, Wang YF, Guo XD, Wang ZG, et al. Stable transfection and identification of a hair follicle-specific expression vector of IGFBP-5 in goat fetal fibroblasts. Genet Mol Res. 2014;13(1):1885–92.

    Article  CAS  PubMed  Google Scholar 

  49. Zhong W, Oguljahan B, Xiao Y, Nelson J, Hernandez L, Garcia-Barrio M, et al. Serum and glucocorticoid-regulated kinase 1 promotes vascular smooth muscle cell proliferation via regulation of beta-catenin dynamics. Cell Signal. 2014;26(12):2765–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Reijo R, Alagappan RK, Patrizio P, Page DC. Severe oligozoospermia resulting from deletions of azoospermia factor gene on Y chromosome. Lancet. 1996;347(9011):1290–3.

    Article  CAS  PubMed  Google Scholar 

  51. Moore FL, Jaruzelska J, Dorfman DM, Reijo-Pera RA. Identification of a novel gene, DZIP (DAZ-interacting protein), that encodes a protein that interacts with DAZ (deleted in azoospermia) and is expressed in embryonic stem cells and germ cells. Genomics. 2004;83(5):834–43.

    Article  CAS  PubMed  Google Scholar 

  52. Curry BJ, Holt JE, McLaughlin EA, Aitken RJ. Characterization of structure and expression of the Dzip1 gene in the rat and mouse. Genomics. 2006;87(2):275–85.

    Article  CAS  PubMed  Google Scholar 

  53. Wu Y, Chen X, Wang S, Jiang M, Zheng B, Zhou Q, et al. Flotillin-2 is an acrosome-related protein involved in mouse spermiogenesis. J Biomed Res. 2012;26(4):278–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Schneyer A, Sidis Y, Xia Y, Saito S, del Re E, Lin HY, et al. Differential actions of follistatin and follistatin-like 3. Mol Cell Endocrinol. 2004;225(1–2):25–8.

    Article  CAS  PubMed  Google Scholar 

  55. Itman C, Mendis S, Barakat B, Loveland KL. All in the family: TGF-beta family action in testis development. Reproduction. 2006;132(2):233–46.

    Article  CAS  PubMed  Google Scholar 

  56. Drummond AE, Findlay JK, Ireland JJ. Animal models of inhibin action. Semin Reprod Med. 2004;22(3):243–52.

    Article  CAS  PubMed  Google Scholar 

  57. O’Connor AE, De Kretser DM. Inhibins in normal male physiology. Semin Reprod Med. 2004;22(3):177–85.

    Article  CAS  PubMed  Google Scholar 

  58. Rato L, Alves MG, Socorro S, Duarte AI, Cavaco JE, Oliveira PF. Metabolic regulation is important for spermatogenesis. Nat Rev Urol. 2012;9(6):330–8.

    Article  CAS  PubMed  Google Scholar 

  59. Bernardino RL, D’Souza WN, Rato L, Rothstein JL, Dias TR, Chui D, et al. Knockout of MCT1 results in total absence of spermatozoa, sex hormones dysregulation, and morphological alterations in the testicular tissue. Cell Tissue Res. 2019;378(2):333–9.

    Article  PubMed  Google Scholar 

  60. Perez-Escuredo J, Van Hee VF, Sboarina M, Falces J, Payen VL, Pellerin L, et al. Monocarboxylate transporters in the brain and in cancer. Biochim Biophys Acta. 2016;1863(10):2481–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Godwin J, Sawby R, Warner RR, Crews D, Grober MS. Hypothalamic arginine vasotocin mRNA abundance variation across sexes and with sex change in a coral reef fish. Brain Behav Evol. 2000;55:77–84.

    Article  CAS  PubMed  Google Scholar 

  62. Semsar K, Godwin J. Social influences on the arginine vasotocin system are independent of gonads in a sex-changing fish. J Neurosci. 2003;23(10):4386–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kobayashi Y, Alam MA, Horiguchi R, Shimizu A, Nakamura M. Sexually dimorphic expression of gonadotropin subunits in the pituitary of protogynous honeycomb grouper (Epinephelus merra): evidence that follicle-stimulating hormone (FSH) induces gonadal sex change. Biol Reprod. 2010;82(6):1030–6.

    Article  CAS  PubMed  Google Scholar 

  64. Huang M, Chen J, Liu Y, Chen H, Yu Z, Ye Z, et al. New Insights Into the Role of Follicle-Stimulating Hormone in Sex Differentiation of the Protogynous Orange-Spotted Grouper, Epinephelus coioides. Front Endocrinol (Lausanne). 2019;10:304.

    Article  PubMed  Google Scholar 

  65. Tao YX, Lin HR. Studies on the natural sex reversal of paddy field eel Monopterus albus. Acta Hydrobiologica Sinica. 1991;15(3):274–8.

    Google Scholar 

  66. Yeung WSB, Chen H, Chan STH. In vivo effects of LH and LHRH-analog on sex reversal and plasma sex steroid profiles in the female Monopterus albus. Gen Comp Endocrinol. 1993;90(1):23–30.

    Article  CAS  PubMed  Google Scholar 

  67. Chen D, Yang W, Han S, Yang H, Cen X, Liu J, et al. A Type IIb, but Not Type IIa, GnRH Receptor Mediates GnRH-Induced Release of Growth Hormone in the Ricefield Eel. Front Endocrinol (Lausanne). 2018;9:721.

    Article  PubMed  Google Scholar 

  68. Zhang Y, Sun X, Zhang L, Zhang W. Testicular Dnmt3 expression and global DNA methylation are down-regulated by gonadotropin releasing hormones in the ricefield eel Monopterus albus. Sci Rep. 2017;7:43158.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Jiang YJ, Luo HR, Hou MX, Chen J, Tao BB, Zhu ZY, et al. Aromatase inhibitor induces sex reversal in the protogynous hermaphroditic rice field eel (Monopterus albus). Aquaculture. 2022;551:737960.

  70. Wu C, Patino R, Davis KB, Chang X. Localization of estrogen receptor alpha and beta RNA in germinal and nongerminal epithelia of the channel catfish testis. Gen Comp Endocrinol. 2001;124(1):12–20.

    Article  CAS  PubMed  Google Scholar 

  71. Choi CY, Habibi HR. Molecular cloning of estrogen receptor α and expression pattern of estrogen receptor subtypes in male and female goldfish. Mol Cell Endocrinol. 2003;204(1–2):169–77.

    Article  CAS  PubMed  Google Scholar 

  72. Miura C, Ohta T, Nader MR, Todo T, Yamauchi K. Estradiol-17b stimulates the renewal of spermatogonialstem cells in males. Biochem Biophys Res Commun. 1999;264:230–4.

    Article  CAS  PubMed  Google Scholar 

  73. Percie du Sert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, et al. The ARRIVE guidelines 20: Updated guidelines for reporting animal research. PLoS Biol. 2020;18(7):3000410.

    Article  Google Scholar 

  74. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9(4):357–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12(4):357–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Pertea M, Kim D, Pertea GM, Leek JT, Salzberg SL. Transcript-level expression analysis of RNA-seq experiments with HISAT StringTie and Ballgown. Nat Protoc. 2016;11(9):1650–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33(3):290–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Wagner GP, Kin K, Lynch VJ. Measurement of mRNA abundance using RNA-seq data: RPKM measure is inconsistent among samples. Theor Biosci. 2012;131(4):281–5.

    Article  CAS  Google Scholar 

  79. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40.

    Article  CAS  PubMed  Google Scholar 

  80. He Z, Wu Y, Xie J, Wang T, Zhang L, Zhang W. Growth differentiation factor 9 (Gdf9) was localized in the female as well as male germ cells in a protogynous hermaphroditic teleost fish, ricefield eel Monopterus albus. Gen Comp Endocrinol. 2012;178(2):355–62.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Shu Sun and Youwei Zhang for their help in raising ricefield eels.

Funding

This work was supported by grants from National Key Research and Development Program (2018YFD0900203), the National Natural Science Foundation of China (32072956 and 31572604), and the Project of Guangdong Basic and Applied Basic Research Foundation (2020A1515111185).

Author information

Authors and Affiliations

Authors

Contributions

WZ, LZ and MF conceived of the study and wrote the manuscript. MF and WY collected gonadal samples for transcriptomic analysis. MF, WY and WZ analyzed the transcriptomic data. MF and LZ analyzed the in vivo effects of letrozole. The author(s) read and approved the final manuscript. 

Corresponding authors

Correspondence to Weimin Zhang or Lihong Zhang.

Ethics declarations

Ethics approval and consent to participate

All procedures and investigations conformed to the ARRIVE guidelines (72), which were also reviewed and approved by the Center for Laboratory Animals of Sun Yat-sen University, and were performed in accordance with the Guiding Principles for the care and use of laboratory animals.

Consent for publication

Not applicable.

Competing interests

The authors declare that no competing interests exist.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

Figure S1. Representative total RNA extracted from gonadal tissues of ricefield eels at stages of female (F), early intersex (EI), mid-intersex (MI), and late intersex (LI). Figure S2. Gene ontology classifications of DEGs in the transcriptome data of ricefield eel gonads during sex change. A: EI vs. F. B: MI vs. EI. C: LI vs MI. X-axis shows the GO term. Y-axis shows the number of DEGs. F: female; EI: early intersex; MI: mid-intersex; LI: late intersex.

Additional file 2:

Table S1. Quality control parameters of the sequencing data. TableS2. Summary of high-quality reads mapped to the ribosome database. Table S3. Overview of mapping status for gonadal transcriptomes during sexual change of ricefield eel. Table S4. Number of reference genes and new genes in sequencing data. Table S5. Annotation of novel genes.Table S6. GO Enrichment Analysis of DEGs between EI vs F. Table S7. GO Enrichment Analysis of DEGs between MI vs EI. Table S8. GO Enrichment Analysis of DEGs between LI vs MI. Table S9. KEGG enrichment analysis of DEGs between EI vs F. Table S10. KEGG enrichment analysis of DEGs between MI vs EI. Table S11. KEGG enrichment analysis of DEGs between LI vs MI. Table S12. The expression of down-regulated DEGs for EI vs F during sex change. Table S13. The expression of up-regulated for DEGs for EI vs F during sex change.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fan, M., Yang, W., Zhang, W. et al. The ontogenic gonadal transcriptomes provide insights into sex change in the ricefield eel Monopterus albus. BMC Zool 7, 56 (2022). https://doi.org/10.1186/s40850-022-00155-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40850-022-00155-4

Keywords