Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.183
Filtrar
1.
BMC Genomics ; 25(1): 464, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38741085

RESUMO

Gonad development includes sex determination and divergent maturation of the testes and ovaries. Recent advances in measuring gene expression in single cells are providing new insights into this complex process. However, the underlying epigenetic regulatory mechanisms remain unclear. Here, we profiled chromatin accessibility in mouse gonadal cells of both sexes from embryonic day 11.5 to 14.5 using single-cell assay for transposase accessible chromatin by sequencing (scATAC-seq). Our results showed that individual cell types can be inferred by the chromatin landscape, and that cells can be temporally ordered along developmental trajectories. Integrative analysis of transcriptomic and chromatin-accessibility maps identified multiple putative regulatory elements proximal to key gonadal genes Nr5a1, Sox9 and Wt1. We also uncover cell type-specific regulatory factors underlying cell type specification. Overall, our results provide a better understanding of the epigenetic landscape associated with the progressive restriction of cell fates in the gonad.


Assuntos
Linhagem da Célula , Cromatina , Gônadas , Fatores de Transcrição SOX9 , Análise de Célula Única , Animais , Cromatina/metabolismo , Cromatina/genética , Camundongos , Linhagem da Célula/genética , Feminino , Masculino , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Gônadas/metabolismo , Gônadas/citologia , Gônadas/embriologia , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo , Testículo/metabolismo , Testículo/citologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Ovário/metabolismo , Ovário/citologia
2.
Gen Comp Endocrinol ; 351: 114476, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38408712

RESUMO

Primordial germ cells (PGCs) are pivotal for gonadal development and reproductive success. Though artificial induction of sterility by targeting PGCs are gaining popularity due to its advantages in fish surrogacy and biodiversity management, it is often skill and time intensive. In this study, we have focused on understanding the role of PGCs and the chemotactic SDF-1/CXCR4 signaling on gonad development of Japanese anchovy (JA, Engraulis japonicus), an upcoming marine model organism with eco-commercial values, with an aim to develop a novel, easy, and versatile gonad sterilization method. Our data showed that PGC migration related genes, i.e., sdf-1a, sdf-1b, cxcr4a, cxcr4b and vasa, are phylogenetically closer relatives of respective herring (Clupea harengus) and zebrafish (Danio rerio) homolog. Subsequently, PGC marking and live tracing experiments confirmed that PGC migration in JA initiates from 16 hours post fertilization (hpf) followed by PGC settlement in the gonadal ridge at 44 hpf. We found that overexpression of zebrafish sdf-1a mRNA in the germ cell suppresses cxcr4a and increases cxcr4b transcription at 8 hpf, dose dependently disrupts PGC migration at 24-48 hpf, induces PGC death and upregulates sdf-1b at 5 days post hatching. 48 h of immersion treatment with CXCR4 antagonist (AMD3100, Abcam) also accelerated PGC mismigration and pushed the PGC away from gonadal ridge in a dose responsive manner, and further when grown to adulthood caused germ cell less gonad formation in some individuals. Cumulatively, our data, for the first time, suggests that JA PGC migration is largely regulated by SDF1/CXCR4 signaling, and modulation of this signaling has strong potential for sterile, germ cell less gonad preparation at a mass scale. However, further in-depth analysis is pertinent to apply this methodology in marine fish species to successfully catapult Japanese anchovy into a true marine fish model.


Assuntos
Gônadas , Mesoderma , Animais , Movimento Celular , Células Germinativas/metabolismo , Gônadas/embriologia , Japão , Peixe-Zebra
3.
Dev Growth Differ ; 66(1): 66-74, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37945353

RESUMO

We previously identified Xenopus tudor domain containing 6/Xenopus tudor repeat (Xtdrd6/Xtr), which was exclusively expressed in the germ cells of adult Xenopus laevis. Western blot analysis showed that the XTdrd6/Xtr protein was translated in St. I/II oocytes and persisted as a maternal factor until the tailbud stage. XTdrd6/Xtr has been reported to be essential for the translation of maternal mRNA involved in oocyte meiosis. In the present study, we examined the distribution of the XTdrd6/Xtr protein during oogenesis and early development, to predict the time point of its action during development. First, we showed that XTdrd6/Xtr is localized to germinal granules in the germplasm by electron microscopy. XTdrd6/Xtr was found to be localized to the origin of the germplasm, the mitochondrial cloud of St. I oocytes, during oogenesis. Notably, XTdrd6/Xtr was also found to be localized around the nuclear membrane of St. I oocytes. This suggests that XTdrd6/Xtr may immediately interact with some mRNAs that emerge from the nucleus and translocate to the mitochondrial cloud. XTdrd6/Xtr was also detected in primordial germ cells and germ cells throughout development. Using transgenic Xenopus expressing XTdrd6/Xtr with a C-terminal FLAG tag produced by homology-directed repair, we found that the zygotic translation of the XTdrd6/Xtr protein began at St. 47/48. As germ cells are surrounded by gonadal somatic cells and are considered to enter a new differentiation stage at this phase, the newly synthesized XTdrd6/Xtr protein may regulate the translation of mRNAs involved in the new steps of germ cell differentiation.


Assuntos
Células Germinativas , Gônadas , Mesoderma , Proteínas de Xenopus , Animais , Células Germinativas/metabolismo , Gônadas/embriologia , Oócitos , Oogênese/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Xenopus laevis/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
4.
Front Immunol ; 12: 790379, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899758

RESUMO

The journey of a hematopoietic stem cell (HSC) involves the passage through successive anatomical sites where HSCs are in direct contact with their surrounding microenvironment, also known as niche. These spatial and temporal cellular interactions throughout development are required for the acquisition of stem cell properties, and for maintaining the HSC pool through balancing self-renewal, quiescence and lineage commitment. Understanding the context and consequences of these interactions will be imperative for our understanding of HSC biology and will lead to the improvement of in vitro production of HSCs for clinical purposes. The aorta-gonad-mesonephros (AGM) region is in this light of particular interest since this is the cradle of HSC emergence during the embryonic development of all vertebrate species. In this review, we will focus on the developmental origin of HSCs and will discuss the novel technological approaches and recent progress made to identify the cellular composition of the HSC supportive niche and the underlying molecular events occurring in the AGM region.


Assuntos
Genômica/tendências , Hematopoese/genética , Células-Tronco Hematopoéticas/fisiologia , Análise de Célula Única/tendências , Nicho de Células-Tronco , Animais , Aorta/embriologia , Técnicas de Cultura de Células/tendências , Linhagem da Célula , Células Cultivadas , Difusão de Inovações , Perfilação da Expressão Gênica/tendências , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Humanos , Mesonefro/embriologia , Fenótipo , Proteômica/tendências , Transdução de Sinais , Transcriptoma
5.
Hum Genet ; 140(12): 1733-1751, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34647195

RESUMO

Mitochondrial disorders are collectively common, genetically heterogeneous disorders in both pediatric and adult populations. They are caused by molecular defects in oxidative phosphorylation, failure of essential bioenergetic supply to mitochondria, and apoptosis. Here, we present three affected individuals from a consanguineous family of Pakistani origin with variable seizures and intellectual disability. Both females display primary ovarian insufficiency (POI), while the male shows abnormal sex hormone levels. We performed whole exome sequencing and identified a recessive missense variant c.694C > T, p.Arg232Cys in TFAM that segregates with disease. TFAM (mitochondrial transcription factor A) is a component of the mitochondrial replisome machinery that maintains mtDNA transcription and replication. In primary dermal fibroblasts, we show depletion of mtDNA and significantly altered mitochondrial function and morphology. Moreover, we observed reduced nucleoid numbers with significant changes in nucleoid size or shape in fibroblasts from an affected individual compared to controls. We also investigated the effect of tfam impairment in zebrafish; homozygous tfam mutants carrying an in-frame c.141_149 deletion recapitulate the mtDNA depletion and ovarian dysgenesis phenotypes observed in affected humans. Together, our genetic and functional data confirm that TFAM plays a pivotal role in gonad development and expands the repertoire of mitochondrial disease phenotypes.


Assuntos
DNA Mitocondrial , Proteínas de Ligação a DNA/genética , Genes Recessivos , Perda Auditiva/genética , Deficiência Intelectual/genética , Proteínas Mitocondriais/genética , Insuficiência Ovariana Primária/genética , Convulsões/genética , Fatores de Transcrição/genética , Animais , Células Cultivadas , Feminino , Gônadas/embriologia , Humanos , Masculino , Linhagem , Peixe-Zebra/genética
6.
Int. j. morphol ; 39(5): 1253-1263, oct. 2021. ilus, tab
Artigo em Inglês | LILACS | ID: biblio-1385493

RESUMO

SUMMARY: The rabbit is considered an ideal animal model for studies that describe abnormalities in the testicles due to the similar morphogenetic mechanisms of sexual development and diseases commonly found in humans. The aim of this study was to determine the male sexual differentiation of the New Zealand rabbit (Oryctolagus cuniculus) through development. The gestational age was estimated and classified as 9, 12, 14, 16, 18, 20, 23 and 28 gestational days. The morphological and sexual determination were performed by histological analysis of the reproductive tract in the embryos and fetuses (9-28 days) as well as by immunohistochemistry- Desert hedgehog-Dhh- (testis-specific protein on Y chromosome- 16, 20, 23 days and adult rabbits). Gonads were observed from the 14th day in an undifferentiated stage and with homogeneous aspect. Sexual differentiation was observed from the 16th day with presence of cells forming gonadal cords and Dhh+ cells in the gonadal parenchyma. From the 18th gestational day testicular cords were observed, which evolved into organized seminiferous tubules. The formation of the efferent ducts and ductus deferens and epididymis was observed on the 20th and 23rd days, respectively. The differentiation of the external genitalia occurred from the 23rd days from the anogenital distance and was identified to identify the penile structures. In summary, the features of the sexual differentiation were determined by observation of the Dhh+ protein in embryos from the 16th day to adulthood, and the morphological particularities observed from the 18th gestational day, determined by differentiation of the external genitalia from the 23rd day.


RESUMEN: El conejo se considera un modelo animal ideal para estudios que describen anomalías a nivel testícular debido a que presenta mecanismos morfogenéticos similares al desa- rrollo sexual y enfermedades que se encuentran comúnmente en los seres humanos. El objetivo de este estudio fue determinar la diferenciación sexual masculina del conejo de Nueva Zelanda (Oryctolagus cuniculus) a través del desarrollo. La edad gestacional se estimó y clasificó en 9, 12, 14, 16, 18, 20, 23 y 28 días gestacionales. La determinación morfológica y sexual se realizó mediante análisis histológico del tracto reproductivo en los embriones y fetos (9 - 28 días) así como mediante inmunohistoquímica -Desert hedgehog-Dhh- (proteína testicular específica en el cromosoma Y- 16, 20, 23 días y conejos adultos). Las gónadas se observaron a partir del día 14 en un estadio indiferenciado y con aspecto homogéneo. Se observó diferenciación sexual a partir del día 16 con presencia de células formadoras de cordones gonadales y células Dhh+ en el parénquima gonadal. A partir del día 18 de gestación se observaron cordones testiculares, que evolucionaron a túbulos seminíferos organizados. La formación de los conductos eferentes, deferentes y del epidídimo se observó a los 20 y 23 días, respectivamente. La diferenciación de los genitales externos ocurrió a partir del día 23 desde la distancia anogenital y se utilizó para identificar las estructuras del pene. En conclusión, las características de la diferenciación sexual se determinaron mediante la observación de la proteína Dhh en embriones desde el día 16 hasta la edad adulta, y las particularidades morfológicas observadas a partir del día 18 de gestación, determinadas por diferenciación de los genitales externos a partir del día 23.


Assuntos
Animais , Masculino , Coelhos , Diferenciação Celular , Desenvolvimento Embrionário e Fetal , Gônadas/crescimento & desenvolvimento , Gônadas/embriologia , Túbulos Seminíferos , Diferenciação Sexual , Imuno-Histoquímica
7.
Sci Rep ; 11(1): 12882, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145300

RESUMO

The monitoring of anthropogenic chemicals in the aquatic environment including their potential effects on aquatic organisms, is important for protecting life under water, a key sustainable development goal. In parallel with monitoring the concentrations of chemicals of concern, sentinel species are often used to investigate the biological effects of contaminants. Among these, bivalve molluscs such as mussels are filter-feeding and sessile, hence an excellent model system for measuring localized pollution. This study investigates the relationship between the metabolic state of the blue mussel (Mytilus edulis) and its physiology in different environments. We developed a computational model based on a reference site (relatively unpolluted) and integrated seasonal dynamics of metabolite relative concentrations with key physiological indicators and environmental parameters. The analysis of the model revealed that changes in metabolite levels during an annual cycle are influenced by water temperature and are linked to gonadal development. This work supports the importance of data-driven biology and its potential in environmental monitoring.


Assuntos
Biomarcadores , Meio Ambiente , Gônadas/embriologia , Gônadas/metabolismo , Metaboloma , Mytilus edulis/fisiologia , Desenvolvimento Sexual , Animais , Biologia Computacional/métodos , Metabolômica/métodos , Modelos Teóricos , Mytilus edulis/embriologia , Estações do Ano , Fatores Sexuais , Desenvolvimento Sexual/genética
8.
Biol Reprod ; 105(2): 345-358, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-33889937

RESUMO

We hypothesized that sexually dimorphic differences exist in the expression of miRNAs in amniotic fluid (AF) and maternal blood plasma (MP) in association with the process of sex determination and gonad differentiation in cattle. Amniotic fluid and MP were collected from six pregnant heifers (three carrying a single male and three a single female embryo) following slaughter on Day 39 postinsemination, coinciding with the peak of SRY expression. Samples (six AF and six MP) were profiled using an miRNA Serum/Plasma Focus PCR Panel. Differentially expressed (DE) miRNAs were identified in AF (n = 5) and associated MP (n = 56) of male vs. female embryos (P < 0.05). Functional analysis showed that inflammatory and immune response were among the 13 biological processes enriched by miRNAs DE in MP in the male group (FDR < 0.05), suggesting that these sex-dependent DE miRNAs may be implicated in modulating the receptivity of the dam to a male embryo. Further, we compared the downstream targets of the sex-dependent DE miRNAs detected in MP with genes previously identified as DE in male vs. female genital ridges. The analyses revealed potential targets that might be important during this developmental stage such as SHROOM2, DDX3Y, SOX9, SRY, PPP1CB, JARID2, USP9X, KDM6A, and EIF2S3. Results from this study highlight novel aspects of sex determination and embryo-maternal communication in cattle such as the potential role of miRNAs in gonad development as well as in the modulation of the receptivity of the dam to a male embryo.


Assuntos
Líquido Amniótico/química , Gônadas/embriologia , MicroRNAs/metabolismo , Plasma/química , Diferenciação Sexual/genética , Animais , Bovinos , Feminino , Masculino
9.
Genetics ; 217(2)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33724412

RESUMO

People with NR5A1 mutations experience testicular dysgenesis, ovotestes, or adrenal insufficiency, but we do not completely understand the origin of this phenotypic diversity. NR5A1 is expressed in gonadal soma precursor cells before expression of the sex-determining gene SRY. Many fish have two co-orthologs of NR5A1 that likely partitioned ancestral gene subfunctions between them. To explore ancestral roles of NR5A1, we knocked out nr5a1a and nr5a1b in zebrafish. Single-cell RNA-seq identified nr5a1a-expressing cells that co-expressed genes for steroid biosynthesis and the chemokine receptor Cxcl12a in 1-day postfertilization (dpf) embryos, as does the mammalian adrenal-gonadal (interrenal-gonadal) primordium. In 2dpf embryos, nr5a1a was expressed stronger in the interrenal-gonadal primordium than in the early hypothalamus but nr5a1b showed the reverse. Adult Leydig cells expressed both ohnologs and granulosa cells expressed nr5a1a stronger than nr5a1b. Mutants for nr5a1a lacked the interrenal, formed incompletely differentiated testes, had no Leydig cells, and grew far larger than normal fish. Mutants for nr5a1b formed a disorganized interrenal and their gonads completely disappeared. All homozygous mutant genotypes lacked secondary sex characteristics, including male breeding tubercles and female sex papillae, and had exceedingly low levels of estradiol, 11-ketotestosterone, and cortisol. RNA-seq showed that at 21dpf, some animals were developing as females and others were not, independent of nr5a1 genotype. By 35dpf, all mutant genotypes greatly under-expressed ovary-biased genes. Because adult nr5a1a mutants form gonads but lack an interrenal and conversely, adult nr5a1b mutants lack a gonad but have an interrenal, the adrenal, and gonadal functions of the ancestral nr5a1 gene partitioned between ohnologs after the teleost genome duplication, likely owing to reciprocal loss of ancestral tissue-specific regulatory elements. Identifying such elements could provide hints to otherwise unexplained cases of Differences in Sex Development.


Assuntos
Glândulas Suprarrenais/metabolismo , Proteínas de Ligação a DNA/genética , Disgenesia Gonadal/genética , Gônadas/metabolismo , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/genética , Glândulas Suprarrenais/embriologia , Animais , Proteínas de Ligação a DNA/metabolismo , Feminino , Gônadas/embriologia , Masculino , Fenótipo , Processos de Determinação Sexual , Fatores de Transcrição/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
10.
Gene ; 774: 145423, 2021 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-33434625

RESUMO

Sox9 gene, a crucial member of the Sox gene family, is present in various organisms and involved in many physiological processes, especially in sex determination and gonad development. In this study, we reported a sox9 gene (designated as Spsox9) from Scylla paramamosain through analyzing published gonad transcriptome data. Meanwhile, the accuracy was validated by PCR technology, and the 3' sequences were cloned with 3' RACE technology. The full-length cDNA of Spsox9 is 2843 bp, consisting of a 243 bp 5' UTR, an 1124 bp 3' UTR, and a 1476 bp ORF encoding 491 amino acids. Furthermore, to better understand its conservation among crustacean species, the sox9 gene ortholog was identified in several other crustaceans species with their published transcriptome data, respectively. All of the Sox9 proteins identified in the current study had the common feature of Sox proteins (HMG domain) and were highly conserved among analyzed crustacean species. In all examined tissues, the Spsox9 was mainly expressed in the gonad (testis and ovary), eyestalk, and cerebral ganglion. During embryo development, Spsox9 was highly expressed in 5 pairs of appendages, 7 pairs of appendages, and eye-pigment formation stage. During ovary development, the expression level of Spsox9 remained stable in the first 4 stages (O1-O4) and decreased in the tertiary vitellogenesis (O5) stage. During testis development, the expression level of Spsox9 was highest in the spermatid stage (T2) and was significantly different from that in the spermatocyte stage (T1) and mature sperm stage (T3) (p < 0.05). In addition, Spsox9 exhibited a sex-biased expression pattern in T1 and O1. These present results indicated that the Spsox9 gene might play crucial roles in the gonad and embryo development of mud crab.


Assuntos
Proteínas de Artrópodes/genética , Braquiúros/genética , Fatores de Transcrição SOX9/genética , Sequência de Aminoácidos , Animais , Braquiúros/embriologia , Clonagem Molecular , Sequência Conservada , DNA Complementar , Feminino , Perfilação da Expressão Gênica , Gônadas/embriologia , Humanos , Masculino , Homologia de Sequência de Aminoácidos , Caracteres Sexuais , Especificidade da Espécie , Vitelogeninas/genética
11.
Integr Zool ; 16(2): 160-169, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32762015

RESUMO

Many egg-laying reptiles possess temperature-dependent sex determination (TSD) in which outcome of gonadogenesis is determined by incubation temperature during a temperature-sensitive period of development. Prior studies on Malayemys macrocephala showed that incubation temperatures influence gonadal development and suggested that M. macrocephala exhibits TSD. However, information on the temperature-sensitivity period in this species was unknown until the current study. Turtle eggs were collected from rice fields in central Thailand from December 2016 to February 2017. In the laboratory, eggs were incubated at male-biased temperature (26 °C) and shifted to female-biased temperature (32 °C), or vice versa. Single shift experiments were performed systematically during embryonic stages 13-21. After hatching, sex of individual turtles was determined by histological analysis. We found that the sex determination of M. macrocephala is affected by temperature up to stage 16 of embryonic development.


Assuntos
Desenvolvimento Embrionário/fisiologia , Diferenciação Sexual/fisiologia , Temperatura , Tartarugas/embriologia , Animais , Embrião não Mamífero/fisiologia , Feminino , Gônadas/embriologia , Masculino , Tartarugas/fisiologia
12.
Gene Expr Patterns ; 39: 119163, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33359643

RESUMO

Desert hedgehog (dhh) is a gene that is crucial for spermatogenesis and Leydig cell differentiation, but little is known regarding its influence on gonadal differentiation and development in fish. To understand its function, we cloned and characterized the dhh gene from Cynoglossus semilaevis (csdhh). The full length csdhh cDNA was 2473 bp, including a 1386 bp open reading frame (ORF), a 475 bp 5'-UTR, and a 612 bp 3'-UTR, encoding a predicted protein of 461 amino acid residues. Phylogenetic analysis showed that the putative protein belongs to the hedgehog (HH) family, and contains typical HH-N and HH-C domains. Amino acid sequence analysis revealed that CsDhh shares many features with Dhh analogues in other teleost species. Real-time quantitative PCR showed that csdhh was detected in eight different tissues in male and female tongue sole. During early embryonic development, the relative expression of the csdhh was significantly higher in the neural stage than in other embryonic developmental stages (P < 0.05). csdhh was detected at 20 days after hatching (dah) and at the critical period of male gonadal differentiation (80-95 dah), the relative expression of the csdhh was significantly higher in the male gonads than the female gonads. In 5, 8, and 12 month old gonads, the relative expression of the csdhh was significantly higher in male and pseudo-male than in female fish. The in situ hybridization (ISH) results showed that the hybridization signal was strongly expressed in primary and secondary spermatocytes, spermatids, and sertoli cells of the 1-year-old fish testis, with only weak signal expression in the corresponding ovarian tissue. These results suggest that csdhh is highly conserved in evolution and plays an important role in spermatogenesis in males and pseudo-males.


Assuntos
Proteínas de Peixes/genética , Linguados/genética , Proteínas Hedgehog/genética , Animais , Clonagem Molecular , Feminino , Proteínas de Peixes/metabolismo , Linguados/embriologia , Linguados/metabolismo , Células Germinativas/metabolismo , Gônadas/embriologia , Gônadas/metabolismo , Proteínas Hedgehog/metabolismo , Masculino
13.
Sci Rep ; 10(1): 20073, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33257723

RESUMO

Research on avian sex determination has focused on the chicken. In this study, we established the utility of another widely used animal model, the Japanese quail (Coturnix japonica), for clarifying the molecular mechanisms underlying gonadal sex differentiation. In particular, we performed comprehensive gene expression profiling of embryonic gonads at three stages (HH27, HH31 and HH38) by mRNA-seq. We classified the expression patterns of 4,815 genes into nine clusters according to the extent of change between stages. Cluster 2 (characterized by an initial increase and steady levels thereafter), including 495 and 310 genes expressed in males and females, respectively, contained five key genes involved in gonadal sex differentiation. A GO analysis showed that genes in this cluster are related to developmental processes including reproductive structure development and developmental processes involved in reproduction were significant, suggesting that expression profiling is an effective approach to identify novel candidate genes. Based on RNA-seq data and in situ hybridization, the expression patterns and localization of most key genes for gonadal sex differentiation corresponded well to those of the chicken. Our results support the effectiveness of the Japanese quail as a model for studies gonadal sex differentiation in birds.


Assuntos
Coturnix/genética , Caracteres Sexuais , Diferenciação Sexual/genética , Animais , Feminino , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Gônadas/embriologia , Masculino , Ovário/embriologia , Codorniz/genética , Reprodução/genética , Testículo/embriologia , Transcriptoma/genética
14.
Int J Mol Sci ; 21(21)2020 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158211

RESUMO

Cadherins are a group of membrane proteins responsible for cell adhesion. They are crucial for cell sorting and recognition during the morphogenesis, but they also play many other roles such as assuring tissue integrity and resistance to stretching, mechanotransduction, cell signaling, regulation of cell proliferation, apoptosis, survival, carcinogenesis, etc. Within the cadherin superfamily, E- and N-cadherin have been especially well studied. They are involved in many aspects of sexual development and reproduction, such as germline development and gametogenesis, gonad development and functioning, and fertilization. E-cadherin is expressed in the primordial germ cells (PGCs) and also participates in PGC migration to the developing gonads where they become enclosed by the N-cadherin-expressing somatic cells. The differential expression of cadherins is also responsible for the establishment of the testis or ovary structure. In the adult testes, N-cadherin is responsible for the integrity of the seminiferous epithelium, regulation of sperm production, and the establishment of the blood-testis barrier. Sex hormones regulate the expression and turnover of N-cadherin influencing the course of spermatogenesis. In the adult ovaries, E- and N-cadherin assure the integrity of ovarian follicles and the formation of corpora lutea. Cadherins are expressed in the mature gametes and facilitate the capacitation of sperm in the female reproductive tract and gamete contact during fertilization. The germ cells and accompanying somatic cells express a series of different cadherins; however, their role in gonads and reproduction is still unknown. In this review, we show what is known and unknown about the role of cadherins in the germline and gonad development, and we suggest topics for future research.


Assuntos
Caderinas/fisiologia , Fertilidade/fisiologia , Gônadas/crescimento & desenvolvimento , Reprodução/fisiologia , Animais , Feminino , Fertilidade/genética , Gametogênese/genética , Células Germinativas/fisiologia , Gônadas/embriologia , Humanos , Masculino , Morfogênese/genética , Reprodução/genética
15.
J Vis Exp ; (164)2020 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-33135688

RESUMO

The Drosophila melanogaster male embryonic gonad is an advantageous model to study various aspects of developmental biology including, but not limited to, germ cell development, piRNA biology, and niche formation. Here, we present a dissection technique to live-image the gonad ex vivo during a period when in vivo live-imaging is highly ineffective. This protocol outlines how to transfer embryos to an imaging dish, choose appropriately-staged male embryos, and dissect the gonad from its surrounding tissue while still maintaining its structural integrity. Following dissection, gonads can be imaged using a confocal microscope to visualize dynamic cellular processes. The dissection procedure requires precise timing and dexterity, but we provide insight on how to prevent common mistakes and how to overcome these challenges. To our knowledge this is the first dissection protocol for the Drosophila embryonic gonad, and will permit live-imaging during an otherwise inaccessible window of time. This technique can be combined with pharmacological or cell-type specific transgenic manipulations to study any dynamic processes occurring within or between the cells in their natural gonadal environment.


Assuntos
Dissecação , Drosophila melanogaster/embriologia , Embrião não Mamífero/diagnóstico por imagem , Gônadas/diagnóstico por imagem , Gônadas/embriologia , Imageamento Tridimensional , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Drosophila melanogaster/citologia , Embrião não Mamífero/citologia , Gônadas/citologia , Masculino
16.
Biosci Rep ; 40(10)2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-32990306

RESUMO

Cytochrome P450 Family 19 SubFamily A member 1 (CYP19A1) gene encodes an aromatase which regulates the sexual differentiation in vertebrates by initiating and maintaining 17ß-Estradiol (E2) synthesis. Here, we described the spatiotemporal expression pattern of CYP19A1 and its functional role in the embryonic gonad development in amphoteric chickens (Gallus gallus). Results showed that CYP19A1 exhibited a sexually dimorphic expression pattern in female gonads early at embryonic day 5.5 (HH 28) and robustly expressed within the cytoplasm in ovarian medullas. Most importantly, we induced the gonadal sex reversal by ectopically delivering the aromatase inhibitor (AI) or estradiol (E2) into chicken embryos. To further explore the role of CYP19A1 in chicken embryonic sexual differentiation, we successfully developed an effective method to deliver lentiviral particles with CYP19A1 manipulation into chicken embryos via embryonic intravascular injection. The analysis of interference and overexpression of CYP19A1 provided solid evidences that CYP19A1 is both necessary and sufficient to initiate sex differentiation toward female in chicken embryos. Collectively, this work demonstrates that CYP19A1 is a crucial sex differentiation gene in the embryonic development, which provides a foundation for understanding the mechanism of sex determination and differentiation in chickens.


Assuntos
Aromatase/metabolismo , Galinhas/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Processos de Determinação Sexual/genética , Animais , Aromatase/genética , Embrião de Galinha , Galinhas/genética , Feminino , Masculino
17.
Gene Expr Patterns ; 38: 119148, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32980455

RESUMO

Inhibin plays important roles in vertebrate reproduction and development. In this study, we have cloned two genes encoding inhibin subunits, inhα and ihnßb, in Chinese tongue sole. inhα consists of 1032 bp, encoding a 343 amino-acid protein. inhßb is composed of 1275 bp, encoding a 424 amino-acid protein. Phylogenetic tree analysis indicated that INHα and INHßB were independently evolved. qPCR showed that inhα expression of in male testis was higher than that in ovary and pseudomale testis, while the expression of inhßb in ovary was higher than that in male and pseudomale testis. During gonadal developmental stages, inhα expression reached highest at 120 days post hatching (dph) both in ovary and testis, then showed decline in ovary but it was first decreased and then increased in the testis. Similarly, inhßb expression in ovary was low at 50-80 dph. At 120 dph, its expression was significantly increased to the peak level, and then gradually decreased. inhßb expression in testis maintained at a low level. During the embryonic developmental stages, inhα displayed the highest expression at 32-cell stage, whereas inhßb reached the highest expression at blastula stages. In situ hybridization data showed that both of inhα and inhßb were detected in oocytes of all stages. In male testis, inhα and inhßb was localized in spermatogonia, spermatocytes, spermatozoa, sertoli and leydig cells. In pseudomale testis, inhα showed the similar pattern in male testis, while the inhßb was detected in spermatocytes and spermatozoa. These data suggested that inhα may participate the spermatogenesis and oogenesis of Chinese tongue sole, while inhßb might predominantly function in oogenesis.


Assuntos
Proteínas de Peixes/genética , Peixes/genética , Regulação da Expressão Gênica no Desenvolvimento , Inibinas/genética , Animais , Feminino , Proteínas de Peixes/química , Proteínas de Peixes/metabolismo , Peixes/crescimento & desenvolvimento , Peixes/metabolismo , Gônadas/embriologia , Gônadas/metabolismo , Inibinas/química , Inibinas/metabolismo , Masculino , Oogênese , Domínios Proteicos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Espermatogênese
18.
Int J Mol Sci ; 21(18)2020 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-32927658

RESUMO

The gonads are unique among the body's organs in having a developmental choice: testis or ovary formation. Gonadal sex differentiation involves common progenitor cells that form either Sertoli and Leydig cells in the testis or granulosa and thecal cells in the ovary. Single-cell analysis is now shedding new light on how these cell lineages are specified and how they interact with the germline. Such studies are also providing new information on gonadal maturation, ageing and the somatic-germ cell niche. Furthermore, they have the potential to improve our understanding and diagnosis of Disorders/Differences of Sex Development (DSDs). DSDs occur when chromosomal, gonadal or anatomical sex are atypical. Despite major advances in recent years, most cases of DSD still cannot be explained at the molecular level. This presents a major pediatric concern. The emergence of single-cell genomics and transcriptomics now presents a novel avenue for DSD analysis, for both diagnosis and for understanding the molecular genetic etiology. Such -omics datasets have the potential to enhance our understanding of the cellular origins and pathogenesis of DSDs, as well as infertility and gonadal diseases such as cancer.


Assuntos
Transtornos do Desenvolvimento Sexual/etiologia , Gônadas/embriologia , Desenvolvimento Sexual , Análise de Célula Única , Transtornos do Desenvolvimento Sexual/diagnóstico , Genômica , Humanos , Análise de Sequência de RNA
19.
J Clin Endocrinol Metab ; 105(9)2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32726409

RESUMO

CONTEXT: Insulin-like growth factor (IGF) signaling is crucial for sex differentiation and development of Leydig and Sertoli cells in fetal mice testes. No such information is available for human embryonic and fetal testes and ovaries. OBJECTIVE: To investigate presence and activity of the IGF signaling system during human embryonic and fetal ovarian and testicular development. DESIGN: Human embryonic and fetal gonads were obtained following legal terminations of pregnancies. Gene expression was assessed by microarray and qPCR transcript analyses. Proteins of the IGF system components were detected with immunohistochemistry and immunofluorescence analyses. Specimens were included from 2010 to 2017. SETTING: University Hospital. PATIENTS/PARTICIPANTS: Ovaries and testes from a total of 124 human embryos and fetuses aged 5 to 17 postconception weeks were obtained from healthy women aged 16 to 47 years resident in Denmark or Scotland. MAIN OUTCOME MEASURES: Gene expression analysis using microarray was performed in 46 specimens and qPCR analysis in 56 specimens, both sexes included. Protein analysis included 22 specimens (11 ovaries, 11 testes). RESULTS: IGF system members were detected in embryonic and fetal testes and ovaries, both at gene transcript and protein level. A higher expression of IGF regulators was detected in testes than ovaries, with a preferred localization to Leydig cells. CONCLUSIONS: These data indicate that the IGF system is active during very early gestation, when it may have a regulatory role in Leydig cells.


Assuntos
Gônadas , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Primeiro Trimestre da Gravidez/genética , Segundo Trimestre da Gravidez/genética , Receptores de Somatomedina/genética , Somatomedinas/genética , Adolescente , Adulto , Embrião de Mamíferos , Feminino , Feto/metabolismo , Feto/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Gônadas/embriologia , Gônadas/metabolismo , Gônadas/patologia , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Gravidez , Primeiro Trimestre da Gravidez/metabolismo , Segundo Trimestre da Gravidez/metabolismo , Receptores de Somatomedina/metabolismo , Transdução de Sinais/genética , Somatomedinas/metabolismo , Adulto Jovem
20.
Dev Dyn ; 249(11): 1365-1376, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32506585

RESUMO

BACKGROUND: The nuclear progesterone receptor (Pgr) is a ligand-dependent transcription factor primarily responsible for mediating progesterone actions relevant for reproduction across vertebrates. Information on the cellular localization of Pgr expression in the reproductive system is required for developing a comprehensive approach to elucidate the role of Pgr in reproduction. RESULTS: We generated transgenic zebrafish Tg(pgr:eGFP) that express enhanced green fluorescent protein (eGFP) driven by promoter sequence of pgr gene. The tissue distribution pattern of egfp mRNA is consistent with the pgr mRNA expression in Tg(pgr:eGFP). In the pituitary, GFP signals are found in the proximal pars distalis. In order to better discern the cellular localization of GFP signals in gonads, Tg(pgr:eGFP) line was crossed with Tg(gsdf:nfsB-mCherry) line, specifically expressing nitroreductase-mCherry fusion protein in granulosa and Sertoli cells in ovary and testis, respectively. Imaging of testis tissue showed that GFP expression was confined to Leydig cells. In the ovary, GFP expression colocalized with the mCherry signal in granulosa cells. Intriguingly, we also identified some non-granulosa cells close to where blood vessels branched, expressing stronger GFP signals than granulosa cells. CONCLUSIONS: Analyzing Tg(pgr:eGFP) expression in zebrafish provided leads toward new routes to study the role of Pgr in reproduction.


Assuntos
Animais Geneticamente Modificados , Embrião não Mamífero/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Proteínas de Fluorescência Verde , Hipófise/embriologia , Regiões Promotoras Genéticas , Receptores de Progesterona/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/genética , Feminino , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Masculino , Peixe-Zebra/embriologia , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...