Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
Biol Reprod ; 110(5): 985-999, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38376238

RESUMO

Sry on the Y-chromosome upregulates Sox9, which in turn upregulates a set of genes such as Fgf9 to initiate testicular differentiation in the XY gonad. In the absence of Sry expression, genes such as Rspo1, Foxl2, and Runx1 support ovarian differentiation in the XX gonad. These two pathways antagonize each other to ensure the development of only one gonadal sex in normal development. In the B6.YTIR mouse, carrying the YTIR-chromosome on the B6 genetic background, Sry is expressed in a comparable manner with that in the B6.XY mouse, yet, only ovaries or ovotestes develop. We asked how testicular and ovarian differentiation pathways interact to determine the gonadal sex in the B6.YTIR mouse. Our results showed that (1) transcript levels of Sox9 were much lower than in B6.XY gonads while those of Rspo1 and Runx1 were as high as B6.XX gonads at 11.5 and 12.5 days postcoitum. (2) FOXL2-positive cells appeared in mosaic with SOX9-positive cells at 12.5 days postcoitum. (3) SOX9-positive cells formed testis cords in the central area while those disappeared to leave only FOXL2-positive cells in the poles or the entire area at 13.5 days postcoitum. (4) No difference was found at transcript levels of all genes between the left and right gonads up to 12.5 days postcoitum, although ovotestes developed much more frequently on the left than the right at 13.5 days postcoitum. These results suggest that inefficient Sox9 upregulation and the absence of Rspo1 repression prevent testicular differentiation in the B6.YTIR gonad.


Assuntos
Fatores de Transcrição SOX9 , Processos de Determinação Sexual , Testículo , Trombospondinas , Regulação para Cima , Animais , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Masculino , Feminino , Camundongos , Trombospondinas/genética , Trombospondinas/metabolismo , Processos de Determinação Sexual/genética , Processos de Determinação Sexual/fisiologia , Testículo/metabolismo , Gônadas/metabolismo , Ovário/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Diferenciação Sexual/genética , Camundongos Endogâmicos C57BL
2.
Science ; 382(6670): 600-606, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37917714

RESUMO

Sex determination in mammals depends on the differentiation of the supporting lineage of the gonads into Sertoli or pregranulosa cells that govern testis and ovary development, respectively. Although the Y-linked testis-determining gene Sry has been identified, the ovarian-determining factor remains unknown. In this study, we identified -KTS, a major, alternatively spliced isoform of the Wilms tumor suppressor WT1, as a key determinant of female sex determination. Loss of -KTS variants blocked gonadal differentiation in mice, whereas increased expression, as found in Frasier syndrome, induced precocious differentiation of ovaries independently of their genetic sex. In XY embryos, this antagonized Sry expression, resulting in male-to-female sex reversal. Our results identify -KTS as an ovarian-determining factor and demonstrate that its time of activation is critical in gonadal sex differentiation.


Assuntos
Ovário , Processos de Determinação Sexual , Proteínas WT1 , Animais , Feminino , Masculino , Camundongos , Ovário/crescimento & desenvolvimento , Processos de Determinação Sexual/genética , Proteína da Região Y Determinante do Sexo/genética , Proteína da Região Y Determinante do Sexo/metabolismo , Testículo/crescimento & desenvolvimento , Proteínas WT1/genética , Proteínas WT1/metabolismo , Isoformas de Proteínas
3.
BMC Genomics ; 24(1): 183, 2023 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-37024792

RESUMO

BACKGROUND: Red-tail catfish (Hemibagrus wyckioides) is an important commercially farmed catfish in southern China. Males of red-tail catfish grow faster than females, suggesting that all-male catfish will produce more significant economic benefits in aquaculture practice. However, little research has been reported on sex determination and gonadal development in red-tail catfish. RESULTS: In this study, we performed the first transcriptomic analysis of male and female gonads at four developmental stages at 10, 18, 30, and 48 days post hatching (dph) using RNA-seq technology. A total of 23,588 genes were screened in 24 sequenced samples, of which 28, 213, 636, and 1381 differentially expressed genes (DEGs) were detected at four developmental stages, respectively. Seven candidate genes of sex determination and differentiation were further identified. Real-time quantitative PCR (RT-qPCR) further confirmed that anti-Mullerian hormone (amh), growth differentiation factor 6a (gdf6a), testis-specific gene antigen 10 (tsga10), and cytochrome P450 family 17 subfamily A (cyp17a) were highly expressed mainly in the male, while cytochrome P450 family 19 subfamily A polypeptide 1b (cyp19a1b), forkhead box L2 (foxl2), and hydroxysteroid 17-beta dehydrogenase 1 (hsd17b1) were highly expressed in the female. The KEGG pathway enrichment data showed that these identified DEGs were mainly involved in steroid hormone biosynthesis and TGF-ß signaling pathways. CONCLUSIONS: Based on RNA-seq data of gonads at the early developmental stages, seven DEGs shared by the four developmental stages were identified, among which amh and gdf6a may be the male-biased expression genes, while foxl2, cyp19a1b and hsd17b1 may be the female-biased expression genes in red-tail catfish. Our study will provide crucial genetic information for the research on sex control in red-tail catfish, as well as for exploring the evolutionary processes of sex determination mechanisms in fish.


Assuntos
Peixes-Gato , Perciformes , Animais , Feminino , Masculino , Transcriptoma , Peixes-Gato/genética , Gônadas/metabolismo , Ovário/metabolismo , Perfilação da Expressão Gênica , Perciformes/genética , Diferenciação Sexual/genética , Regulação da Expressão Gênica no Desenvolvimento , Processos de Determinação Sexual/genética
4.
Genetics ; 224(1)2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-36947451

RESUMO

Estrogen signaling exerts a decisive role in female sex determination and differentiation in chicken and fish. Aromatase encoded by Cyp19a1 is the key enzyme that catalyzes the conversion of androgen to estrogen. Correlative analyses implicate the potential involvement of aromatase in reptilian sexual development, however, the direct genetic evidence is lacking. Herein, we found that Cyp19a1 exhibited temperature-dependent sexually dimorphic expression, and located in the medullary somatic cells in early female embryos of the red-eared slider turtle (Trachemys scripta elegans), before the gonad is distinct. To determine the functional role of Cyp19a1 in turtle ovarian determination, we established loss- and gain-of-function models through in ovo lentivirus-mediated genetic manipulation. At female-producing temperature, inhibition of aromatase or knockdown of Cyp19a1 in turtle embryos resulted in female-to-male sex reversal, with the formation of a testis-like structure and a male distribution pattern of germ cells, as well as ectopic expression of male-specific markers (SOX9 and AMH) and disappearance of ovarian regulator FOXL2. On the contrary, overexpression of Cyp19a1 at male-producing temperature led to male-to-female sex reversal. In conclusion, our results suggest that Cyp19a1 is both necessary and sufficient for ovarian determination in the red-eared slider turtle, establishing causality and a direct genetic link between aromatase and reptilian sex determination and differentiation.


Assuntos
Tartarugas , Animais , Feminino , Masculino , Tartarugas/genética , Aromatase/genética , Aromatase/metabolismo , Processos de Determinação Sexual/genética , Mutação com Ganho de Função , Estrogênios/metabolismo , Temperatura , Diferenciação Sexual/genética
5.
J Exp Biol ; 225(16)2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35860927

RESUMO

Variation in developmental conditions can affect a variety of embryonic processes and shape a number of phenotypic characteristics that can affect offspring throughout their lives. This is particularly true of oviparous species where development typically occurs outside of the female, and studies have shown that traits such as survival and behavior can be altered by both temperature and exposure to steroid hormones during development. In species with temperature-dependent sex determination (TSD), the fate of gonadal development can be affected by temperature and by maternal estrogens present in the egg at oviposition, and there is evidence that these factors can affect gene expression patterns. Here, we explored how thermal fluctuations and exposure to an estrogen metabolite, estrone sulfate, affect the expression of several genes known to be involved in sexual differentiation: Kdm6b, Dmrt1, Sox9, FoxL2 and Cyp19A1. We found that most of the genes responded to both temperature and estrone sulfate exposure, but that the responses to these factors were not identical, in that estrone sulfate effects occur downstream of temperature effects. Our findings demonstrate that conjugated hormones such as estrone sulfate are capable of influencing temperature-dependent pathways to potentially alter how embryos respond to temperature, and highlight the importance of studying the interaction of maternal hormone and temperature effects.


Assuntos
Processos de Determinação Sexual , Tartarugas , Animais , Estrona/análogos & derivados , Estrona/metabolismo , Feminino , Expressão Gênica , Hormônios , Processos de Determinação Sexual/genética , Diferenciação Sexual/fisiologia , Temperatura , Tartarugas/fisiologia
6.
Biol Reprod ; 107(4): 1125-1138, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-35594452

RESUMO

In mammals, testis development is triggered by the expression of the sex-determining Y-chromosome gene SRY to commit the Sertoli cell (SC) fate at gonadal sex determination in the fetus. Several genes have been identified to be required to promote the testis pathway following SRY activation (i.e., SRY box 9 (SOX9)) in an embryo; however, it largely remains unknown about the genes and the mechanisms involved in stabilizing the testis pathway after birth and throughout adulthood. Herein, we report postnatal males with SC-specific deletion of Raptor demonstrated the absence of SC unique identity and adversely acquired granulosa cell-like characteristics, along with loss of tubular architecture and scattered distribution of SCs and germ cells. Subsequent genome-wide analysis by RNA sequencing revealed a profound decrease in the transcripts of testis genes (i.e., Sox9, Sox8, and anti-Mullerian hormone (Amh)) and, conversely, an increase in ovary genes (i.e., LIM/Homeobox gene 9 (Lhx9), Forkhead box L2 (Foxl2) and Follistatin (Fst)); these changes were further confirmed by immunofluorescence and quantitative reverse-transcription polymerase chain reaction. Importantly, co-immunofluorescence demonstrated that Raptor deficiency induced SCs dedifferentiation into a progenitor state; the Raptor-mutant gonads showed some ovarian somatic cell features, accompanied by enhanced female steroidogenesis and elevated estrogen levels, yet the zona pellucida 3 (ZP3)-positive terminally feminized oocytes were not observed. In vitro experiments with primary SCs suggested that Raptor is likely involved in the fibroblast growth factor 9 (FGF9)-induced formation of cell junctions among SCs. Our results established that Raptor is required to maintain SC identity, stabilize the male pathway, and promote testis development.


Assuntos
Aves Predatórias , Células de Sertoli , Animais , Hormônio Antimülleriano/genética , Estrogênios/metabolismo , Feminino , Fator 9 de Crescimento de Fibroblastos/genética , Folistatina/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Homeodomínio LIM/genética , Masculino , Mamíferos/genética , Camundongos , Aves Predatórias/genética , Aves Predatórias/metabolismo , Fatores de Transcrição SOX9/genética , Células de Sertoli/metabolismo , Processos de Determinação Sexual/genética , Testículo/metabolismo , Fatores de Transcrição/genética
7.
Artigo em Inglês | MEDLINE | ID: mdl-34920111

RESUMO

The fruitless (fru) gene has an important function in the courtship behavior and sex determination pathway of Drosophila melanogaster; however, the fru gene has never been reported in shrimps. In this study, the fruitless-like gene was identified in Cherax quadricarinatus (Cqfru) and is reported here for the first time. A sequence analysis revealed a conserved BTB domain in Cqfru which is the same as fru in D. melanogaster. An analysis of the expression level of Cqfru showed that it was highly expressed in the gastrula stage during embryonic development. Furthermore, in situ hybridization and expression distribution in tissues showed that its sexually dimorphic expression may be focused on the hepatopancreas, brains, and gonads. The gonads, brains, and hepatopancreas of males had a higher expression level of Cqfru than those of females; however, the expression level of the abdominal ganglion was found to be higher in females than in males in this study. The results of an RNA interference treatment showed that a knockdown of Cqfru reduced the expression of the insulin-like androgenic gland hormone (IAG) and tumor necrosis factor (TNF). The characteristic fru gene in shrimps is reported here for the first time, with the results providing basic information for research into the sex-determination mechanism in C. quadricarinatus.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Animais , Astacoidea/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Masculino , Proteínas do Tecido Nervoso/genética , Caracteres Sexuais , Processos de Determinação Sexual/genética , Fatores de Transcrição/metabolismo
8.
Endocrinology ; 162(12)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34581801

RESUMO

In teleost fish, sex steroids are involved in sex determination, sex differentiation, and fertility. Cyp17a1 (Cytochrome P450 family 17 subfamily A member 1) is thought to play essential roles in fish steroidogenesis. Therefore, to further understand its roles in steroidogenesis, sex determination, and fertility in fish, we constructed a cyp17a1 gene mutant in Nile tilapia (Oreochromis niloticus). In XX fish, mutation of the cyp17a1 gene led to a female-to-male sex reversal with a significant decline in 17ß-estradiol (E2) and testosterone (T) production, and ectopic expression of male-biased markers (Dmrt1 and Gsdf) in gonads from the critical window of sex determination. Sex reversal was successfully rescued via T or E2 administration, and ovarian characteristics were maintained after termination of E2 supplementation in the absence of endogenous estrogen production in cyp17a1-/- XX fish. Likewise, deficiencies in T and 11-ketotestosterone (11-KT) production in both cyp17a1-/- XX sex-reversed males and cyp17a1-/- XY mutants resulted in meiotic initiation delays, vas deferens obstruction and sterility due to excessive apoptosis and abnormal mitochondrial morphology. However, 11-KT treatment successfully rescued the dysspermia to produce normal sperm in cyp17a1-/- male fish. Significant increases in gonadotropic hormone (gth) and gth receptors in cyp17a1-/- mutants may excessively upregulate steroidogenic gene expression in Leydig cells through a feedback loop. Taken together, our findings demonstrate that Cyp17a1 is indispensable for E2 production, which is fundamental for female sex determination and differentiation in XX tilapia. Additionally, Cyp17a1 is essential for T and 11-KT production, which further promotes spermatogenesis and fertility in XY males.


Assuntos
Ciclídeos/fisiologia , Família 17 do Citocromo P450/fisiologia , Hormônios Esteroides Gonadais/biossíntese , Infertilidade Masculina/genética , Processos de Determinação Sexual/genética , Animais , Animais Geneticamente Modificados , Ciclídeos/genética , Ciclídeos/metabolismo , Família 17 do Citocromo P450/genética , Feminino , Fertilidade/genética , Peixes/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Infertilidade Masculina/veterinária , Masculino , Redes e Vias Metabólicas/genética
9.
Nucleic Acids Res ; 49(16): 9097-9116, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34403484

RESUMO

Sex is a modulator of health that has been historically overlooked in biomedical research. Recognizing this knowledge gap, funding agencies now mandate the inclusion of sex as a biological variable with the goal of stimulating efforts to illuminate the molecular underpinnings of sex biases in health and disease. DNA methylation (DNAm) is a strong molecular candidate for mediating such sex biases; however, a robust and well characterized annotation of sex differences in DNAm is yet to emerge. Beginning with a large (n = 3795) dataset of DNAm profiles from normative adult whole blood samples, we identified, validated and characterized autosomal sex-associated co-methylated genomic regions (sCMRs). Strikingly, sCMRs showed consistent sex differences in DNAm over the life course and a subset were also consistent across cell, tissue and cancer types. sCMRs included sites with known sex differences in DNAm and links to health conditions with sex biased effects. The robustness of sCMRs enabled the generation of an autosomal DNAm-based predictor of sex with 96% accuracy. Testing this tool on blood DNAm profiles from individuals with sex chromosome aneuploidies (Klinefelter [47,XXY], Turner [45,X] and 47,XXX syndrome) revealed an intimate relationship between sex chromosomes and sex-biased autosomal DNAm.


Assuntos
Metilação de DNA , Transtornos do Cromossomo Sexual no Desenvolvimento Sexual/genética , Processos de Determinação Sexual/genética , Cromossomos/genética , Ilhas de CpG , Feminino , Humanos , Masculino
10.
Philos Trans R Soc Lond B Biol Sci ; 376(1832): 20200089, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34247507

RESUMO

Several hypotheses explain the prevalence of undifferentiated sex chromosomes in poikilothermic vertebrates. Turnovers change the master sex determination gene, the sex chromosome or the sex determination system (e.g. XY to WZ). Jumping master genes stay main triggers but translocate to other chromosomes. Occasional recombination (e.g. in sex-reversed females) prevents sex chromosome degeneration. Recent research has uncovered conserved heteromorphic or even homomorphic sex chromosomes in several clades of non-avian and non-mammalian vertebrates. Sex determination in sturgeons (Acipenseridae) has been a long-standing basic biological question, linked to economical demands by the caviar-producing aquaculture. Here, we report the discovery of a sex-specific sequence from sterlet (Acipenser ruthenus). Using chromosome-scale assemblies and pool-sequencing, we first identified an approximately 16 kb female-specific region. We developed a PCR-genotyping test, yielding female-specific products in six species, spanning the entire phylogeny with the most divergent extant lineages (A. sturio, A. oxyrinchus versus A. ruthenus, Huso huso), stemming from an ancient tetraploidization. Similar results were obtained in two octoploid species (A. gueldenstaedtii, A. baerii). Conservation of a female-specific sequence for a long period, representing 180 Myr of sturgeon evolution, and across at least one polyploidization event, raises many interesting biological questions. We discuss a conserved undifferentiated sex chromosome system with a ZZ/ZW-mode of sex determination and potential alternatives. This article is part of the theme issue 'Challenging the paradigm in sex chromosome evolution: empirical and theoretical insights with a focus on vertebrates (Part I)'.


Assuntos
Evolução Molecular , Peixes/genética , Genoma , Cromossomos Sexuais/genética , Processos de Determinação Sexual/genética , Animais , Feminino , Filogenia
11.
Sex Dev ; 15(1-3): 69-79, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33902053

RESUMO

In species with temperature-dependent sex determination (TSD), incubation temperatures regulate the expression of genes involved in gonadal differentiation and determine whether the gonads develop into ovaries or testes. For most species, natural incubation conditions result in transient exposure to thermal cues for both ovarian and testis development, but how individuals respond to this transient exposure varies and can drive variation in the resulting sex ratios. Here, we argue that variation in the timing to respond to temperature cues, or thermal responsiveness, is a trait needing further study. Recent work in the red-eared slider turtle (Trachemys scripta) has found that when embryos experience transient exposure to warm conditions (i.e., heatwaves), some embryos show high responsiveness, requiring only short exposures to commit to ovarian development, while others show low responsiveness, developing testes even after more extended exposures to warm conditions. We discuss how maternal estrogens might influence thermal responsiveness for organisms that develop under thermal fluctuations. Examining the interplay of molecular responses to more subtle thermal and endocrine environments may reveal significant insights into the process of sex determination in species with TSD.


Assuntos
Estrogênios , Tartarugas , Animais , Feminino , Gônadas/metabolismo , Masculino , Processos de Determinação Sexual/genética , Diferenciação Sexual/genética , Temperatura , Tartarugas/genética
12.
PLoS Genet ; 17(3): e1009468, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33788836

RESUMO

Doublesex (Dsx) and Fruitless (Fru) are the two downstream transcription factors that actuate Drosophila sex determination. While Dsx assists Fru to regulate sex-specific behavior, whether Fru collaborates with Dsx in regulating other aspects of sexual dimorphism remains unknown. One important aspect of sexual dimorphism is found in the gonad stem cell (GSC) niches, where male and female GSCs are regulated to create large numbers of sperm and eggs. Here we report that Fru is expressed male-specifically in the GSC niche and plays important roles in the development and maintenance of these cells. Unlike previously-studied aspects of sex-specific Fru expression, which are regulated by Transformer (Tra)-mediated alternative splicing, we show that male-specific expression of fru in the gonad is regulated downstream of dsx, and is independent of tra. fru genetically interacts with dsx to support maintenance of the niche throughout development. Ectopic expression of fru inhibited female niche formation and partially masculinized the ovary. fru is also required autonomously for cyst stem cell maintenance and cyst cell survival. Finally, we identified a conserved Dsx binding site upstream of fru promoter P4 that regulates fru expression in the niche, indicating that fru is likely a direct target for transcriptional regulation by Dsx. These findings demonstrate that fru acts outside the nervous system to influence sexual dimorphism and reveal a new mechanism for regulating sex-specific expression of fru that is regulated at the transcriptional level by Dsx, rather than by alternative splicing by Tra.


Assuntos
Proteínas de Drosophila/genética , Regulação da Expressão Gênica , Gônadas/citologia , Gônadas/metabolismo , Proteínas do Tecido Nervoso/genética , Caracteres Sexuais , Processos de Determinação Sexual/genética , Nicho de Células-Tronco/genética , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Animais , Proteínas de Drosophila/metabolismo , Evolução Molecular , Feminino , Imunofluorescência , Ordem dos Genes , Loci Gênicos , Masculino , Proteínas do Tecido Nervoso/metabolismo , Testículo , Fatores de Transcrição/metabolismo
13.
Sci China Life Sci ; 64(1): 77-87, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32529288

RESUMO

Polyploids in vertebrates are generally associated with unisexual reproduction, but the direct consequences of polyploidy on sex determination system and reproduction mode remain unknown. Here, we synthesized a group of artificial octoploids between unisexual gynogenetic hexaploid Carassius gibelio and sexual tetraploid Carassius auratus. The synthetic octoploids were revealed to have more than 200 chromosomes, in which 50 chromosomes including the X/Y sex determination system were identified to transfer from sexual tetraploid C. auratus into the unisexual gynogenetic hexaploid C. gibelio. Significantly, a few synthetic octoploid males were found to be fertile, and one octoploid male was confirmed to regain sexual reproduction ability, which exhibits characteristics that are the same to sexual reproduction tetraploid males, such as 1:1 sex ratio occurrence, meiosis completion and euploid sperm formation in spermatogenesis, as well as normal embryo development and gene expression pattern during embryogenesis. Therefore, the current finding provides a unique case to explore the effect of sex determination system incorporation on reproduction mode transition from unisexual gynogenesis to sexual reproduction along with genome synthesis of recurrent polyploidy in vertebrates.


Assuntos
Carpas/genética , Genoma/genética , Carpa Dourada/genética , Poliploidia , Animais , Cromossomos/genética , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Feminino , Fertilidade/genética , Perfilação da Expressão Gênica/métodos , Hibridização in Situ Fluorescente/métodos , Masculino , Meiose/genética , Reprodução/genética , Processos de Determinação Sexual/genética , Razão de Masculinidade , Espermatogênese/genética , Espermatozoides/metabolismo
14.
Int J Mol Sci ; 21(21)2020 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-33171657

RESUMO

The increasing incidence of testicular dysgenesis syndrome-related conditions and overall decline in human fertility has been linked to the prevalence of oestrogenic endocrine disrupting chemicals (EDCs) in the environment. Ectopic activation of oestrogen signalling by EDCs in the gonad can impact testis and ovary function and development. Oestrogen is the critical driver of ovarian differentiation in non-mammalian vertebrates, and in its absence a testis will form. In contrast, oestrogen is not required for mammalian ovarian differentiation, but it is essential for its maintenance, illustrating it is necessary for reinforcing ovarian fate. Interestingly, exposure of the bi-potential gonad to exogenous oestrogen can cause XY sex reversal in marsupials and this is mediated by the cytoplasmic retention of the testis-determining factor SOX9 (sex-determining region Y box transcription factor 9). Oestrogen can similarly suppress SOX9 and activate ovarian genes in both humans and mice, demonstrating it plays an essential role in all mammals in mediating gonad somatic cell fate. Here, we review the molecular control of gonad differentiation and explore the mechanisms through which exogenous oestrogen can influence somatic cell fate to disrupt gonad development and function. Understanding these mechanisms is essential for defining the effects of oestrogenic EDCs on the developing gonads and ultimately their impacts on human reproductive health.


Assuntos
Disruptores Endócrinos/efeitos adversos , Estrogênios/efeitos adversos , Gônadas/efeitos dos fármacos , Gônadas/crescimento & desenvolvimento , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Transtornos do Desenvolvimento Sexual/etiologia , Estrogênios/fisiologia , Feminino , Gônadas/citologia , Humanos , Masculino , Camundongos , Modelos Biológicos , Gravidez , Saúde Reprodutiva , Fatores de Transcrição SOX9/metabolismo , Processos de Determinação Sexual/genética , Processos de Determinação Sexual/fisiologia , Diferenciação Sexual/efeitos dos fármacos , Diferenciação Sexual/genética , Diferenciação Sexual/fisiologia
15.
Arch Toxicol ; 94(12): 4143-4158, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32975586

RESUMO

In contrast to established zebrafish gene annotations, the question of sex determination has still not been conclusively clarified for developing zebrafish, Danio rerio, larvae, 28 dpf or earlier. Recent studies indicate polygenic sex determination (PSD), with the genes being distributed throughout the genome. Early genetic markers of sex in zebrafish help unravel co-founding sex-related differences to apply to human health and environmental toxicity studies. A qPCR-based method was developed for six genes: cytochrome P450, family 17, subfamily A, polypeptide 1 (cyp17a1); cytochrome P450, family 19, subfamily A, polypeptide 1a (cyp19a1a); cytochrome P450, family 19, subfamily A, polypeptides 1b (cyp19a1b); vitellogenin 1 (vtg1); nuclear receptor subfamily 0, group B, member 1 (nr0b1), sry (sex-determining region Y)-box 9b (sox9b) and actin, beta 1 (actb1), the reference gene. Sry-box 9a (Sox9a), insulin-like growth factor 3 (igf3) and double sex and mab-3 related transcription factor 1 (dmrt1), which are also known to be associated with sex determination, were used in gene expression tests. Additionally, Next-Generation-Sequencing (NGS) sequenced the genome of two adult female and male and two juveniles. PCR analysis of adult zebrafish revealed sex-specific expression of cyp17a1, cyp19a1a, vtg1, igf3 and dmrt1, the first four strongly expressed in female zebrafish and the last one highly expressed in male conspecifics. From NGS, nine female and four male-fated genes were selected as novel for assessing zebrafish sex, 28 dpf. Differences in transcriptomes allowed allocation of sex-specific genes also expressed in juvenile zebrafish.


Assuntos
Processos de Determinação Sexual , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Embrião não Mamífero/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Reação em Cadeia da Polimerase , Processos de Determinação Sexual/genética , Transcriptoma , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
16.
Mol Cell Endocrinol ; 504: 110689, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31891771

RESUMO

Cyp19a1a is a key gene responsible for the production of estradiol-17ß (E2), the main functional estrogen and a major downstream regulator of reproduction in teleost fish. It is widely known that CYP19 gene expression, aromatase activity, and E2 production can influence gonadal differentiation and sex reversal in teleost fish, but the feedback mechanisms whereby E2 regulates cyp19a1a remain poorly understood, especially regarding the potential roles of endogenous small RNA molecules (miRNAs). Here, we identified miR-26a-5p as a regulatory factor of its predicted target gene (cyp19a1a). In vitro and in vivo studies showed that miR-26a-5p can decrease cyp19a1a expression. Furthermore, high doses of E2 act as a repressor of miR-26a-5p. This study proposes a regulatory feedback loop whereby E2 regulates cyp19a1a through miR-26a-5p, and suggests that this positive feedback is an important aspect of the control of E2 production.


Assuntos
Aromatase/genética , Bass , Transtornos do Desenvolvimento Sexual , Estradiol/farmacologia , MicroRNAs/genética , Animais , Aromatase/metabolismo , Bass/genética , Bass/metabolismo , Transtornos do Desenvolvimento Sexual/genética , Transtornos do Desenvolvimento Sexual/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Feminino , Gônadas/efeitos dos fármacos , Gônadas/metabolismo , Gônadas/fisiologia , Masculino , MicroRNAs/metabolismo , Processos de Determinação Sexual/efeitos dos fármacos , Processos de Determinação Sexual/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
17.
Sci Rep ; 9(1): 20099, 2019 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-31882810

RESUMO

Approximately 0.5-1.4% of natal males and 0.2-0.3% of natal females meet DSM-5 criteria for gender dysphoria, with many of these individuals self-describing as transgender men or women. Despite recent improvements both in social acceptance of transgender individuals as well as access to gender affirming therapy, progress in both areas has been hampered by poor understanding of the etiology of gender dysphoria. Prior studies have suggested a genetic contribution to gender dysphoria, but previously proposed candidate genes have not yet been verified in follow-up investigation. In this study, we expand on the topic of gender identity genomics by identifying rare variants in genes associated with sexually dimorphic brain development and exploring how they could contribute to gender dysphoria. To accomplish this, we performed whole exome sequencing on the genomic DNA of 13 transgender males and 17 transgender females. Whole exome sequencing revealed 120,582 genetic variants. After filtering, 441 variants in 421 genes remained for further consideration, including 21 nonsense, 28 frameshift, 13 splice-region, and 225 missense variants. Of these, 21 variants in 19 genes were found to have associations with previously described estrogen receptor activated pathways of sexually dimorphic brain development. These variants were confirmed by Sanger Sequencing. Our findings suggest a new avenue for investigation of genes involved in estrogen signaling pathways related to sexually dimorphic brain development and their relationship to gender dysphoria.


Assuntos
Sequenciamento do Exoma , Variação Genética , Estudo de Associação Genômica Ampla , Pessoas Transgênero , Processamento Alternativo , Mapeamento Cromossômico , Feminino , Mutação da Fase de Leitura , Estudo de Associação Genômica Ampla/métodos , Humanos , Masculino , Mutação de Sentido Incorreto , Análise de Sequência de DNA , Processos de Determinação Sexual/genética
18.
J Assist Reprod Genet ; 36(7): 1329-1337, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31214882

RESUMO

PURPOSE: The human Y chromosome plays a central role in sex determination and spermatogenesis. The azoospermia factor (AZF) loci on the Y chromosome contain genes that were thought to be testis specific with their deletions leading to spermatogenic failure. However, beyond the testis, the AZF genes (mainly those in AZFa and AZFb loci) are widely expressed in multiple tissues. Further, these genes are predicted to play roles in processes such as gene regulation and protein synthesis. These observations suggest that the AZF genes may have functions beyond regulation of fertility. RESULTS: Three major areas have emerged where alternations in AZF genes have effects beyond infertility. (1) Poor-quality embryos are generated in assisted reproduction when sperm from men harboring Y chromosome microdeletions are used, (2) a higher preponderance of neuropsychiatry disorders is observed in men with deletions in AZF genes, and (3) copy number variations and altered expression of AZF genes are found in several cancers. CONCLUSION: While our data is preliminary and observational in nature, systematic studies are required to address how genetic alterations in the Y chromosome can affect the health of men beyond infertility. This information will provide a different perspective in the area of androgenetics and have implications in devising strategies for maintaining the overall well-being of infertile males.


Assuntos
Azoospermia/genética , Cromossomos Humanos Y/genética , Deleção de Genes , Infertilidade Masculina/genética , Azoospermia/patologia , Humanos , Infertilidade Masculina/patologia , Masculino , Processos de Determinação Sexual/genética , Espermatogênese/genética , Espermatozoides/patologia
19.
Mol Biol Rep ; 46(3): 2971-2978, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30879272

RESUMO

Two sisters phenotypically normal females, presenting with tumor abdominal mass with histopathological findings of teratoma and gonadoblastoma associated to 46,XY male-to-female sex reversal syndrome, secondary to a duplication in DAX-1, possibly inherited of maternal gonadal mosaicism. Copy number variation and functional effects of the duplication were done by MLPA multiplex ligation-dependent probe amplification and real time PCR. DAX-1, also known as dosage sensitive sex reversal gene (DSS), is considered the most likely candidate gene involved in XY gonadal dysgenesis when overexpressed. The excess of DAX-1 gene disturbs testicular development by down regulation of SF-1, WT1, and SOX9. This is the first report of 46,XY sex reversal in two siblings who have a maternally inherited duplication of DAX-1 associated with reduced levels of expression of downstream genes as SOX9-SF1.


Assuntos
Receptor Nuclear Órfão DAX-1/genética , Disgenesia Gonadal/genética , Processos de Determinação Sexual/genética , Adolescente , Criança , Receptor Nuclear Órfão DAX-1/metabolismo , Variações do Número de Cópias de DNA , Proteínas de Ligação a DNA/genética , Feminino , Dosagem de Genes/genética , Duplicação Gênica , Disgenesia Gonadal 46 XY/genética , Gonadoblastoma/genética , Humanos , Linhagem , Análise para Determinação do Sexo/métodos , Diferenciação Sexual , Maturidade Sexual/genética , Irmãos , Teratoma , Testículo/anormalidades
20.
BMC Biol ; 17(1): 6, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30683095

RESUMO

BACKGROUND: Sex determination mechanisms in teleost fish broadly differ from mammals and birds, with sex chromosomes that are far less differentiated and recombination often occurring along the length of the X and Y chromosomes, posing major challenges for the identification of specific sex determination genes. Here, we take an innovative approach of comparative genome analysis of the genomic sequences of the X chromosome and newly sequenced Y chromosome in the channel catfish. RESULTS: Using a YY channel catfish as the sequencing template, we generated, assembled, and annotated the Y genome sequence of channel catfish. The genome sequence assembly had a contig N50 size of 2.7 Mb and a scaffold N50 size of 26.7 Mb. Genetic linkage and GWAS analyses placed the sex determination locus within a genetic distance less than 0.5 cM and physical distance of 8.9 Mb. However, comparison of the channel catfish X and Y chromosome sequences showed no sex-specific genes. Instead, comparative RNA-Seq analysis between females and males revealed exclusive sex-specific expression of an isoform of the breast cancer anti-resistance 1 (BCAR1) gene in the male during early sex differentiation. Experimental knockout of BCAR1 gene converted genetic males (XY) to phenotypic females, suggesting BCAR1 as a putative sex determination gene. CONCLUSIONS: We present the first Y chromosome sequence among teleost fish, and one of the few whole Y chromosome sequences among vertebrate species. Comparative analyses suggest that sex-specific isoform expression through alternative splicing may underlie sex determination processes in the channel catfish, and we identify BCAR1 as a potential sex determination gene.


Assuntos
Ictaluridae/genética , Processos de Determinação Sexual/genética , Cromossomo Y , Animais , Mapeamento Cromossômico , Feminino , Ligação Genética , Genoma , Masculino , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA