Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(24)2022 Dec 07.
Article in English | MEDLINE | ID: mdl-36555134

ABSTRACT

Germ cells are pivotal for gonadal sexuality maintenance and reproduction. Sex lethal (sxl), the somatic sex determining gene of Drosophila, is the known regulator and initiator of germ cell femininity in invertebrates. However, the role of the Sxl homologue has rarely been investigated in vertebrates. So, we used medaka to clarify the role of sxl in vertebrate gonadogenesis and sexuality and identified two Sxl homologues, i.e., Sxl1a and Sxl1b. We found that sxl1a specifically expresses in the primordial germ cells (PGC), ovary, (early gonia and oocytes), while sxl1b distributions are ubiquitous. An mRNA overexpression of sxl1a accelerated germ cell numbers in 10 DAH XY fish, and sxl1a knockdown (KD), on the other hand, induced PGC mis-migration, aberrant PGC structuring and ultimately caused significant germ cell reduction in XX fish. Using an in vitro promoter analysis and in vivo steroid treatment, we found a strong link between sxl1a and estrogenic germ cell-population maintenance. Further, using sxl1a-KD and erß2-knockout fish, we determined that sxl1 acts through erß2 and controls PGC sexuality. Cumulatively, our study highlights the novel role of sxl1a in germ cell maintenance and sexual identity assignment and thus might become a steppingstone to understanding the commonalities of animal sexual development.


Subject(s)
Oryzias , Animals , Female , Oryzias/genetics , Genes, Lethal , Gonads , Sex Differentiation , Ovary , Germ Cells
2.
Sci Rep ; 11(1): 11117, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34045599

ABSTRACT

Vertebrates usually exhibit gonochorism, whereby their sex is fixed throughout their lifetime. However, approximately 500 species (~ 2%) of extant teleost fishes change sex during their lifetime. Although phylogenetic and evolutionary ecological studies have recently revealed that the extant sequential hermaphroditism in teleost fish is derived from gonochorism, the evolution of this transsexual ability remains unclear. We revealed in a previous study that the tunica of the ovaries of several protogynous hermaphrodite groupers contain functional androgen-producing cells, which were previously unknown structures in the ovaries of gonochoristic fishes. Additionally, we demonstrated that these androgen-producing cells play critical roles in initiating female-to-male sex change in several grouper species. In the present study, we widened the investigation to include 7 genera and 18 species of groupers and revealed that representatives from most major clades of extant groupers commonly contain these androgen-producing cells, termed testicular-inducing steroidogenic (TIS) cells. Our findings suggest that groupers acquired TIS cells in the tunica of the gonads for successful sex change during their evolution. Thus, TIS cells trigger the evolution of sex change in groupers.


Subject(s)
Hermaphroditic Organisms/physiology , Perciformes/physiology , Sex Determination Processes/physiology , Testis/physiology , Androgens/metabolism , Animals , Female , Male
3.
Curr Biol ; 31(8): 1699-1710.e6, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33639108

ABSTRACT

Male and female animals typically display innate sex-specific mating behaviors, which, in vertebrates, are highly dependent on sex steroid signaling. While estradiol-17ß (E2) signaling through estrogen receptor 2 (ESR2) serves to defeminize male mating behavior in rodents, the available evidence suggests that E2 signaling is not required in teleosts for either male or female mating behavior. Here, we report that female medaka deficient for Esr2b, a teleost ortholog of ESR2, are not receptive to males but rather court females, despite retaining normal ovarian function with an unaltered sex steroid milieu. Thus, contrary to both prevailing views in rodents and teleosts, E2/Esr2b signaling in the brain plays a decisive role in feminization and demasculinization of female mating behavior and sexual preference in medaka. Further behavioral testing showed that mutual antagonism between E2/Esr2b signaling and androgen receptor-mediated androgen signaling in adulthood induces and actively maintains sex-typical mating behaviors and preference. Our results also revealed that the female-biased sexual dimorphism in esr2b expression in the telencephalic and preoptic nuclei implicated in mating behavior can be reversed between males and females by altering the sex steroid milieu in adulthood, likely via mechanisms involving direct E2-induced transcriptional activation. In addition, Npba, a neuropeptide mediating female sexual receptivity, was found to act downstream of E2/Esr2b signaling in these brain nuclei. Collectively, these functional and regulatory mechanisms of E2/Esr2b signaling presumably underpin the neural mechanism for induction, maintenance, and reversal of sex-typical mating behaviors and sexual preference in teleosts, at least in medaka.


Subject(s)
Oryzias , Animals , Estradiol , Female , Gonadal Steroid Hormones , Male , Oryzias/genetics , Receptors, Estrogen , Reproduction , Sexual Behavior, Animal
4.
Physiol Rev ; 101(3): 1237-1308, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33180655

ABSTRACT

A diverse array of sex determination (SD) mechanisms, encompassing environmental to genetic, have been found to exist among vertebrates, covering a spectrum from fixed SD mechanisms (mammals) to functional sex change in fishes (sequential hermaphroditic fishes). A major landmark in vertebrate SD was the discovery of the SRY gene in 1990. Since that time, many attempts to clone an SRY ortholog from nonmammalian vertebrates remained unsuccessful, until 2002, when DMY/dmrt1by was discovered as the SD gene of a small fish, medaka. Surprisingly, however, DMY/dmrt1by was found in only 2 species among more than 20 species of medaka, suggesting a large diversity of SD genes among vertebrates. Considerable progress has been made over the last 3 decades, such that it is now possible to formulate reasonable paradigms of how SD and gonadal sex differentiation may work in some model vertebrate species. This review outlines our current understanding of vertebrate SD and gonadal sex differentiation, with a focus on the molecular and cellular mechanisms involved. An impressive number of genes and factors have been discovered that play important roles in testicular and ovarian differentiation. An antagonism between the male and female pathway genes exists in gonads during both sex differentiation and, surprisingly, even as adults, suggesting that, in addition to sex-changing fishes, gonochoristic vertebrates including mice maintain some degree of gonadal sexual plasticity into adulthood. Importantly, a review of various SD mechanisms among vertebrates suggests that this is the ideal biological event that can make us understand the evolutionary conundrums underlying speciation and species diversity.


Subject(s)
Gonads/physiology , Sex Determination Processes/physiology , Sex Differentiation/physiology , Vertebrates/physiology , Animals , Female , Male
5.
Elife ; 92020 08 12.
Article in English | MEDLINE | ID: mdl-32783809

ABSTRACT

Recent studies in mice demonstrate that a subset of neurons in the medial preoptic area (MPOA) that express galanin play crucial roles in regulating parental behavior in both sexes. However, little information is available on the function of galanin in social behaviors in other species. Here, we report that, in medaka, a subset of MPOA galanin neurons occurred nearly exclusively in males, resulting from testicular androgen stimulation. Galanin-deficient medaka showed a greatly reduced incidence of male-male aggressive chases. Furthermore, while treatment of female medaka with androgen induced male-typical aggressive acts, galanin deficiency in these females attenuated the effect of androgen on chases. Given their male-biased and androgen-dependent nature, the subset of MPOA galanin neurons most likely mediate androgen-dependent male-male chases. Histological studies further suggested that variability in the projection targets of the MPOA galanin neurons may account for the species-dependent functional differences in these evolutionarily conserved neural substrates.


Subject(s)
Androgens/metabolism , Galanin/metabolism , Oryzias/metabolism , Sex Characteristics , Animals , Animals, Genetically Modified , Cell Line , Female , Galanin/genetics , Gene Expression , HEK293 Cells , Humans , Male , Neurons/metabolism , Preoptic Area/cytology , Preoptic Area/metabolism , Receptors, Galanin/metabolism , Reproduction , Signal Transduction , Steroids/metabolism
6.
Cell Death Differ ; 27(11): 3117-3130, 2020 11.
Article in English | MEDLINE | ID: mdl-32483382

ABSTRACT

Autophagy, or cellular self-digestion, is an essential cellular process imperative for energy homeostasis, development, differentiation, and survival. However, the intrinsic factors that bring about the sex-biased differences in liver autophagy are still unknown. In this work, we found that autophagic genes variably expresses in the steroidogenic tissues, mostly abundant in liver, and is influenced by the individual's sexuality. Starvation-induced autophagy in a time-dependent female-dominated manner, and upon starvation, a strong gender responsive circulating steroid-HK2 relation was observed, which highlighted the importance of estrogen in autophagy regulation. This was further confirmed by the enhanced or suppressed autophagy upon estrogen addition (male) or blockage (female), respectively. In addition, we found that estrogen proved to be the common denominator between stress management, glucose metabolism, and autophagic action in female fish. To understand further, we used estrogen receptor (ER)α- and ER-ß2-knockout (KO) medaka and found ER-specific differences in sex-biased autophagy. Interestingly, starvation resulted in significantly elevated mTOR transcription (compared with control) in male ERα-KO fish while HK2 and ULK activation was greatly decreased in both KO fish in a female oriented fashion. Later, ChIP analysis confirmed that, NRF2, an upstream regulator of mTOR, only binds to ERα, while both ERα and ERß2 are effectively pulled down the HK2 and LC3. FIHC data show that, in both ER-KO fish, LC3 nuclear-cytoplasmic transport and its associated pathways involving SIRT1 and DOR were greatly affected. Cumulatively, our data suggest that, ERα-KO strongly affected the early autophagic initiation and altered the LC3 nuclear-cytoplasmic translocation, thereby influencing the sex-biased final autophagosome formation in medaka. Thus, existence of steroid responsive autophagy regulatory-switches and sex-biased steroid/steroid receptor availability influences the gender-skewed autophagy. Expectedly, this study may furnish newer appreciation for gender-specific medicine research and therapeutics.


Subject(s)
Autophagy , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Liver/metabolism , Sex Differentiation , Animals , Female , Fishes , Male , Receptors, Thyroid Hormone/metabolism , Sirtuin 1/metabolism , TOR Serine-Threonine Kinases/metabolism
7.
Elife ; 82019 08 06.
Article in English | MEDLINE | ID: mdl-31383257

ABSTRACT

Male and female animals display innate sex-specific mating behaviors. In teleost fish, altering the adult sex steroid milieu can effectively reverse sex-typical mating behaviors, suggesting remarkable sexual lability of their brains as adults. In the teleost medaka, neuropeptide B (NPB) is expressed female-specifically in the brain nuclei implicated in mating behavior. Here, we demonstrate that NPB is a direct mediator of estrogen action on female mating behavior, acting in a female-specific but reversible manner. Analysis of regulatory mechanisms revealed that the female-specific expression of NPB is dependent on direct transcriptional activation by estrogen via an estrogen-responsive element and is reversed in response to changes in the adult sex steroid milieu. Behavioral studies of NPB knockouts revealed that female-specific NBP mediates female receptivity to male courtship. The female-specific NPB signaling identified herein is presumably a critical element of the neural circuitry underlying sexual dimorphism and lability of mating behaviors in teleosts.


Subject(s)
Neuropeptides/metabolism , Oryzias/physiology , Sexual Behavior, Animal/drug effects , Animals , Estrogens/metabolism , Female , Gene Expression Regulation/drug effects
8.
Stem Cell Reports ; 13(2): 419-433, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412286

ABSTRACT

In vertebrates, estrogen receptors are essential for estrogen-associated early gonadal sex development. Our previous studies revealed sexual dimorphic expression of estrogen receptor ß2 (ERß2) during embryogenesis of medaka, and here we investigated the functional importance of ERß2 in female gonad development and maintenance using a transgenerational ERß2-knockdown (ERß2-KD) line and ERß2-null mutants. We found that ERß2 reduction favored male-biased gene transcription, suppressed female-responsive gene expression, and affected SDF1a and CXCR4b co-assisted chemotactic primordial germ cell (PGC) migration. Co-overexpression of SDF1a and CXXR4b restored the ERß2-KD/KO associated PGC mismigration. Further analysis confirmed that curtailment of ERß2 increased intracellular Ca2+ concentration, disrupted intra- and extracellular calcium homeostasis, and instigated autophagic germ cell degradation and germ cell loss, which in some cases ultimately affected the XX female sexual development. This study is expected improve our understanding of germ cell maintenance and sex spectrum, and hence open new avenues for reproductive disorder management.


Subject(s)
Estrogen Receptor beta/metabolism , Fish Proteins/metabolism , Gonads/growth & development , Sex Differentiation , Animals , Calcium/metabolism , Cell Proliferation , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Embryo, Nonmammalian/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Female , Fish Proteins/antagonists & inhibitors , Fish Proteins/genetics , Gene Expression Regulation, Developmental , Germ Cells/cytology , Germ Cells/metabolism , Gonads/metabolism , Male , Oryzias/growth & development , Oryzias/metabolism , RNA, Antisense/genetics , RNA, Antisense/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism
9.
Mol Cell Endocrinol ; 460: 104-122, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28711606

ABSTRACT

Although estrogens have been generally considered to play a critical role in ovarian differentiation in non-mammalian vertebrates, the specific functions of estrogens during ovarian differentiation remain unclear. We isolated two mutants with premature stops in the ovarian aromatase (cyp19a1) gene from an N-ethyl-N-nitrosourea-based gene-driven mutagenesis library of the medaka, Oryzias latipes. In XX mutants, gonads first differentiated into normal ovaries containing many ovarian follicles that failed to accumulate yolk. Subsequently, ovarian tissues underwent extensive degeneration, followed by the appearance of testicular tissues on the dorsal side of ovaries. In the newly formed testicular tissue, strong expression of gsdf was detected in sox9a2-positive somatic cells surrounding germline stem cells suggesting that gsdf plays an important role in testicular differentiation during estrogen-depleted female-to-male sex reversal. We conclude that endogenous estrogens synthesized after fertilization are not essential for early ovarian differentiation but are critical for the maintenance of adult ovaries.


Subject(s)
Loss of Function Mutation/genetics , Oryzias/genetics , Ovary/pathology , Sex Determination Processes , Sexual Maturation , Amino Acid Sequence , Animals , Aromatase/chemistry , Aromatase/genetics , Base Sequence , Cell Lineage , Down-Regulation/genetics , Estrogens/biosynthesis , Female , Gene Expression Profiling , Male , Ovarian Follicle/pathology , Sex Determination Processes/genetics , Testis/pathology , Up-Regulation/genetics , Vitellogenins/metabolism
10.
Sci Rep ; 7(1): 3183, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28600501

ABSTRACT

Environmental stressors, gonadal degenerative diseases and tumour development can significantly alter the oocyte physiology, and species fertility and fitness. To expand the molecular understanding about oocyte degradation, we isolated several spliced variants of Japanese anchovy hatching enzymes (AcHEs; ovastacin homologue) 1 and 2, and analysed their potential in oocyte sustenance. Particularly, AcHE1b, an ovary-specific, steroid-regulated, methylation-dependent, stress-responsive isoform, was neofunctionalized to regulate autophagic oocyte degeneration. AcHE1a and 2 triggered apoptotic degeneration in vitellogenic and mature oocytes, respectively. Progesterone, starvation, and high temperature elevated the total degenerating oocyte population and AcHE1b transcription by hyper-demethylation. Overexpression, knockdown and intracellular zinc ion chelation study confirmed the functional significance of AcHE1b in autophagy induction, possibly to mitigate the stress effects in fish, via ion-homeostasis. Our finding chronicles the importance of AcHEs in stress-influenced apoptosis/autophagy cell fate decision and may prove significant in reproductive failure assessments, gonadal health maintenance and ovarian degenerative disease therapy.


Subject(s)
Gonads/chemistry , Metalloendopeptidases/chemistry , Oocytes/chemistry , Protein Isoforms/genetics , Animals , Apoptosis , Autophagy , Cell Lineage/genetics , DNA Fragmentation , Fertility/genetics , Gonads/growth & development , Metalloendopeptidases/genetics , Oocytes/growth & development , Progesterone/genetics , Protein Isoforms/chemistry , Proteolysis , Vertebrates/growth & development
11.
Endocrinology ; 158(3): 627-639, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27967235

ABSTRACT

The maturation-inducing hormone 17α,20ß-dihydroxy-4-pregnen-3-one (DHP) was first identified in the amago salmon. Although carbonyl reductase-like 20ß-hydroxysteroid dehydrogenase (CR/20ß-HSD) was reported to convert 17α-hydroxyprogesterone (17OHP) to DHP in rainbow trout, we previously found that CR/20ß-HSD messenger RNA (mRNA) was not upregulated in stimulated granulosa cells from masu salmon, which suggested that DHP is synthesized by a different enzyme. Accordingly, the current study aimed to identify the specific 20ß-hydroxysteroid dehydrogenase (20ß-HSD) responsible for DHP production by granulosa cells during final oocyte maturation in masu salmon. RNA sequencing was performed on granulosa layers that were isolated from ovarian follicles at 1 month before ovulation and incubated with or without forskolin, which was used to mimic luteinizing hormone, and ∼12 million reads were obtained, which yielded 71,062 contigs of >100 bp. tBlastx analysis identified 1 contig (#f103496) as similar to 17ß-hydroxysteroid dehydrogenase type 12 (hsd17ß12); however, because the full-length #f103496 sequence was different from hsd17ß12, it was termed hsd17ß12-like (hsd17ß12l). We found that mammalian cells transfected with full-length hsd17ß12l exhibited considerable 20ß-HSD activity, as indicated by efficient conversion of exogenous 17OHP to DHP. In addition, we found that hsd17ß12l mRNA levels were consistently low in follicles during vitellogenic growth; however, the levels increased significantly during final oocyte maturation. The levels of hsd17ß12l mRNA were also considerably increased in granulosa layers in which 20ß-HSD activity was induced by salmon pituitary extract. Therefore, we suggest that hsd17ß12l, not CR/20ß-HSD, is the 20ß-HSD responsible for DHP production by granulosa cells in masu salmon during final oocyte maturation.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Fish Proteins/metabolism , Hydroxyprogesterones/metabolism , Oocytes/growth & development , Animals , Base Sequence , Female , Granulosa Cells/metabolism , Molecular Sequence Data , Salmon , Seasons , Sequence Analysis, RNA
12.
Sci Rep ; 6: 19738, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26813267

ABSTRACT

Sex is pivotal for reproduction, healthcare and evolution. In the fish medaka, the Y-chromosomal dmy (also dmrt1bY) serves the sex determiner, which activates dmrt1 for male sex maintenance. However, how dmy makes the male decision via initiating testicular differentiation has remained unknown. Here we report that autosomal gsdf serves a male sex initiator. Gene addition and deletion revealed that gsdf was necessary and sufficient for maleness via initiating testicular differentiation. We show that gsdf transcription is activated directly by dmy. These results establish the autosomal gsdf as the first male sex initiator. We propose that dmy determines maleness through activating gsdf and dmrt1 without its own participation in developmental processes of sex initiation and maintenance. gsdf may easily become a sex determiner or other autosomal genes can be recruited as new sex determiners to initiate gsdf expression. Our findings offer new insights into molecular mechanisms underlying sex development and evolution of sex-controlling genes in vertebrates.


Subject(s)
Oryzias/genetics , Sex Determination Processes/genetics , Y Chromosome , Animals , Female , Gene Deletion , Gene Expression Regulation, Developmental , Gonads/embryology , Gonads/metabolism , Male , Phenotype
13.
Sci Rep ; 6: 19543, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26782368

ABSTRACT

In contrast to our understanding of testicular differentiation, ovarian differentiation is less well understood in vertebrates. In mammals, R-spondin1 (Rspo1), an activator of Wnt/ß-catenin signaling pathway, is located upstream of the female sex determination pathway. However, the functions of Rspo1 in ovarian differentiation remain unclear in non-mammalian species. In order to elucidate the detailed functions of Rspo/Wnt signaling pathway in fish sex determination/differentiation, the ectopic expression of the Rspo1 gene was performed in XY medaka (Oryzias latipes). The results obtained demonstrated that the gain of Rspo1 function induced femininity in XY fish. The overexpression of Rspo1 enhanced Wnt4b and ß-catenin transcription, and completely suppressed the expression of male-biased genes (Dmy, Gsdf, Sox9a2 and Dmrt1) as well as testicular differentiation. Gonadal reprograming of Rspo1-over-expressed-XY (Rspo1-OV-XY) fish, induced the production of female-biased genes (Cyp19a1a and Foxl2), estradiol-17ß production and further female type secondary sexuality. Moreover, Rspo1-OV-XY females were fertile and produced successive generations. Promoter analyses showed that Rspo1 transcription was directly regulated by DM domain genes (Dmy, the sex-determining gene, and Dmrt1) and remained unresponsive to Foxl2. Taken together, our results strongly suggest that Rspo1 is sufficient to activate ovarian development and plays a decisive role in the ovarian differentiation in medaka.


Subject(s)
Cell Differentiation/physiology , Fish Proteins/genetics , Oryzias/growth & development , Oryzias/genetics , Ovary/physiology , Sex Differentiation/genetics , Animals , Cell Differentiation/genetics , Female , Gene Expression Regulation, Developmental/genetics , Male , Oryzias/physiology , Promoter Regions, Genetic/genetics , Sex Determination Processes/genetics , Sex Determination Processes/physiology , Sex Differentiation/physiology , Testis/physiology , Transcription, Genetic/genetics , Wnt4 Protein/genetics , beta Catenin/genetics
14.
PLoS One ; 10(12): e0145182, 2015.
Article in English | MEDLINE | ID: mdl-26700177

ABSTRACT

In fish, oocyte meiotic maturation is regulated by 17α, 20ß-dihydroxy-progesterone through cAMP. To study the role of cAMP response element binding protein (CREB) in meiotic maturation, we cloned and characterized the expression pattern of CREBs from two fish models, the Nile tilapia and catfish. In the Nile tilapia three different CREBs were identified where in CREB1 was found in many tissues including gonads with abundant expression in testis. CREB2, few amino acids shorter than CREB1, was expressed in several tissues with abundant expression in ovary. In addition, a 3'UTR variant form, CREB3 was exclusively found in ovary. During natural 14-day ovarian cycle of the Nile tilapia, CREB1 expression was stable throughout vitellogenesis with a sharp decrease on the day of spawning. In contrast, CREB2 remain unchanged throughout the ovarian cycle, however elevated in 11-day full-grown immature ovarian follicle and after hCG-induction. Interestingly, CREB3 expression was induced three folds on the day of spawning as well as during hCG-induced oocyte maturation. Based on the synergistic expression pattern, CREB1 is likely to control oocyte growth, whereas CREB 2 and 3 contribute to oocyte maturation in tilapia and the latter seems to be critical. In catfish, a single form of CREB showed a maximum expression during spawning phase and hCG-induced maturation both in vivo and in vitro augmented CREB expression. These results suggest that spatial and temporal expression of CREBs seems to be important for final oocyte maturation and may also regulate oocyte growth in fish.


Subject(s)
Catfishes/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Fish Proteins/metabolism , Oocytes/metabolism , Tilapia/metabolism , Amino Acid Sequence , Animals , Catfishes/genetics , Cloning, Molecular , Cyclic AMP Response Element-Binding Protein/genetics , DNA, Complementary/chemistry , Female , Fish Proteins/genetics , Gene Expression Profiling , Male , Molecular Sequence Data , Oocytes/growth & development , Phylogeny , Sequence Alignment , Tilapia/genetics , Vitellogenesis
15.
PLoS Genet ; 11(11): e1005678, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26588702

ABSTRACT

Variation in the TGF-ß signaling pathway is emerging as an important mechanism by which gonadal sex determination is controlled in teleosts. Here we show that amhy, a Y-specific duplicate of the anti-Müllerian hormone (amh) gene, induces male sex determination in Nile tilapia. amhy is a tandem duplicate located immediately downstream of amhΔ-y on the Y chromosome. The coding sequence of amhy was identical to the X-linked amh (amh) except a missense SNP (C/T) which changes an amino acid (Ser/Leu92) in the N-terminal region. amhy lacks 5608 bp of promoter sequence that is found in the X-linked amh homolog. The amhΔ-y contains several insertions and deletions in the promoter region, and even a 5 bp insertion in exonVI that results in a premature stop codon and thus a truncated protein product lacking the TGF-ß binding domain. Both amhy and amhΔ-y expression is restricted to XY gonads from 5 days after hatching (dah) onwards. CRISPR/Cas9 knockout of amhy in XY fish resulted in male to female sex reversal, while mutation of amhΔ-y alone could not. In contrast, overexpression of Amhy in XX fish, using a fosmid transgene that carries the amhy/amhΔ-y haplotype or a vector containing amhy ORF under the control of CMV promoter, resulted in female to male sex reversal, while overexpression of AmhΔ-y alone in XX fish could not. Knockout of the anti-Müllerian hormone receptor type II (amhrII) in XY fish also resulted in 100% complete male to female sex reversal. Taken together, these results strongly suggest that the duplicated amhy with a missense SNP is the candidate sex determining gene and amhy/amhrII signal is essential for male sex determination in Nile tilapia. These findings highlight the conserved roles of TGF-ß signaling pathway in fish sex determination.


Subject(s)
Anti-Mullerian Hormone/genetics , Gene Duplication , Mutation, Missense , Polymorphism, Single Nucleotide , Sex Determination Processes , Tilapia/genetics , Y Chromosome , Animals , Animals, Genetically Modified , Female , Genetic Linkage , Male
16.
Mol Cell Endocrinol ; 415: 87-99, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26265450

ABSTRACT

The Nile tilapia, Oreochromis niloticus, is a gonochoristic teleost fish with an XX/XY genetic system and is an excellent model for gonadal sex differentiation. In the present study, we screened novel genes that were expressed predominantly in either XY or XX undifferentiated gonads during the critical period for differentiation of gonads into ovaries or testes using microarray screening. We focused on one of the isolated 12 candidate genes, #9475, which was an ortholog of gsdf (gonadal soma-derived factor), a member of the transforming growth factor-beta superfamily. #9475/gsdf showed sexual dimorphism in expression in XY gonads before any other testis differentiation-related genes identified in this species thus far. We also overexpressed the #9475/gsdf gene in XX tilapia, and XX tilapia bearing the #9475/gsdf gene showed normal testis development, which suggests that #9475/gsdf plays an important role in male determination and/or differentiation in tilapia.


Subject(s)
Cichlids/genetics , Testis/growth & development , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Animals , Cichlids/growth & development , Cichlids/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Male , Oligonucleotide Array Sequence Analysis/methods , Phylogeny , Sex Characteristics , Sex Differentiation , Testis/metabolism
17.
Article in English | MEDLINE | ID: mdl-26188170

ABSTRACT

Dietary compromises, especially food restrictions, possess species-specific effects on the health status and infection control in several organisms, including fish. To understand the starvation-mediated physiological responses in Edwardsiella tarda infected red sea bream, especially in the liver, we performed a 20-day starvation experiment using 4 treatment (2 fed and 2 starved) groups, namely, fed-placebo, starved-placebo, fed-infected, and starved-infected, wherein bacterial exposure was done on the 11th day. In the present study, the starved groups showed reduced hepatosomatic index and drastic depletion in glycogen storage and vacuole formation. The fed-infected fish showed significant (P<0.05) increase in catalase and superoxide dismutase activity in relation to its starved equivalent. Significant (P<0.05) alteration in glucose and energy metabolism, as evident from hexokinase and glucose-6-phosphate dehydrogenase activity, was recorded in the starved groups. Interestingly, coinciding with the liver histology, PPAR (peroxisome proliferator activated receptors) α transcription followed a time-dependent activation in starved groups while PPARγ exhibited an opposite pattern. The transcription of hepcidin 1 and transferrin, initially increased in 0dai (days after infection) starved fish but reduced significantly (P<0.05) at later stages. Two-color immunohistochemistry and subsequent cell counting showed significant increase in P63-positive cells at 0dai and 5dai but later reduced slightly at 10dai. Similar results were also obtained in the lysosomal (cathepsin D) and non-lysosomal (ubiquitin) gene transcription level. All together, our data suggest that starvation exerts multidirectional responses, which allows for better physiological adaptations during any infectious period, in red sea bream.


Subject(s)
Edwardsiella tarda/growth & development , Enterobacteriaceae Infections/physiopathology , Fish Diseases/physiopathology , Liver/physiopathology , Sea Bream/physiology , Starvation , Animals , Catalase/metabolism , Edwardsiella tarda/physiology , Energy Metabolism , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Fish Diseases/metabolism , Fish Diseases/microbiology , Fish Proteins/genetics , Fish Proteins/metabolism , Food , Gene Expression , Glucose/metabolism , Glycogen/metabolism , Host-Pathogen Interactions , Liver/metabolism , Liver/microbiology , PPAR alpha/genetics , PPAR gamma/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sea Bream/metabolism , Sea Bream/microbiology , Superoxide Dismutase/metabolism , Vacuoles/metabolism
18.
Gen Comp Endocrinol ; 222: 144-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26163025

ABSTRACT

Relaxin-like gonad-stimulating peptide (RGP) of starfish Asterina pectinifera was the first invertebrate gonadotropin to have its chemical structure identified. However, it is unclear whether gonadotropic hormones in other species starfish are relaxin-like peptides. Thus, this study tried to identify the molecular structure of gonadotropic hormone in Asterias amurensis. As a result, we identified A. amurensis gonadotropic hormone as the RGP (AamRGP). The DNA sequence encoding AamRGP consisted of 330 base pairs with an open reading frame encoding a peptide of 109 amino acids (aa), including a signal peptide (26 aa), B-chain (20 aa), C-peptide (38 aa) and A-chain (25 aa). Comparing with A. pectinifera RGP (ApeRGP), the amino acid identity levels between AmaRGP and ApeRGP were 58% for the A-chain and 73% for the B-chain. Furthermore, chemical synthetic AamRGP induced gamete spawning and oocyte maturation in ovarian fragments of A. amurensis. In contrast, the ovary of A. pectinifera failed to respond to the AamRGP. This suggested that AamRGP is a new relaxin-like peptide.


Subject(s)
Gonads/metabolism , Relaxin/metabolism , Starfish/metabolism , Animals , Female , Humans
19.
PLoS One ; 10(4): e0123900, 2015.
Article in English | MEDLINE | ID: mdl-25894586

ABSTRACT

Figla (factor in the germ line, alpha), a female germ cell-specific transcription factor, had been shown to activate genetic hierarchies in oocytes. The ectopic expression of Figla was known to repress spermatogenesis-associated genes in male mice. However, the potential role of Figla in other vertebrates remains elusive. The present work was aimed to identify and characterize the functional relevance of Figla in the ovarian development of Nile tilapia (Oreochromis niloticus). Tissue distribution and ontogeny analysis revealed that tilapia Figla gene was dominantly expressed in the ovary from 30 days after hatching. Immunohistochemistry analysis also demonstrated that Figla was expressed in the cytoplasm of early primary oocytes. Intriguingly, over-expression of Figla in XY fish resulted in the disruption of spermatogenesis along with the depletion of meiotic spermatocytes and spermatids in testis. Dramatic decline of sycp3 (synaptonemal complex protein 3) and prm (protamine) expression indicates that meiotic spermatocytes and mature sperm production are impaired. Even though Sertoli cell (dmrt1) and Leydig cell (star and cyp17a1) marker genes remained unaffected, hsd3b1 expression and 11-KT production were enhanced in Figla-transgene testis. Taken together, our data suggest that fish Figla might play an essential role in the ovarian development by antagonizing spermatogenesis.


Subject(s)
Cell Differentiation , Cichlids/metabolism , Ovary/cytology , Spermatogenesis , Transcription Factors/metabolism , Androgens , Animals , Animals, Genetically Modified , Base Sequence , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Male , Ovary/metabolism , Phylogeny , Subcellular Fractions/metabolism , Synteny/genetics , Transcription Factors/genetics , Transgenes
20.
Mol Cell Endocrinol ; 400: 61-70, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25458697

ABSTRACT

Interferon gamma (IFNγ) is an active player in estrogen dependent immuno-regulation of fish. The present work was aimed to characterize the alternatively spliced isoforms of IFNγ2 in the gonadal sex development in medaka. Phylogenetic analysis demonstrated that IFNγ2a and 2b were clustered with fish specific interferon gamma. Our in vitro promoter and mini-genome analysis data confirmed that alternative splicing of IFNγ2 is regulated by estrogens and androgens. Tissue distribution, quantitative PCR and ISH data demonstrated ubiquitous expression of IFNγ2a, while IFNγ2b was only expressed predominantly in female germ cells than males. This was further confirmed by germ cell specific GFP signals in the IFNγ2b-GFP over-expressed embryos and specific induction of IFNγ2b expression in the BrdU positive cells. All together our data suggest that steroid responsive alternatively spliced IFNγ2b isoforms might have some indirect roles in germ cell proliferation and thus can be an important candidate for immuno-reproductive interaction studies.


Subject(s)
Alternative Splicing/drug effects , Fish Proteins/genetics , Interferon-gamma/genetics , Oryzias/genetics , Ovary/metabolism , Testis/metabolism , Androgens/metabolism , Androgens/pharmacology , Animals , Cell Proliferation , Embryo, Nonmammalian , Estrogens/metabolism , Estrogens/pharmacology , Female , Fish Proteins/metabolism , Interferon-gamma/metabolism , Male , Organ Specificity , Oryzias/classification , Oryzias/growth & development , Ovary/cytology , Ovary/drug effects , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sex Factors , Testis/cytology , Testis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...