Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Biol Evol ; 38(6): 2260-2272, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-33528505

RESUMO

In the course of evolution, pecorans (i.e., higher ruminants) developed a remarkable diversity of osseous cranial appendages, collectively referred to as "headgear," which likely share the same origin and genetic basis. However, the nature and function of the genetic determinants underlying their number and position remain elusive. Jacob and other rare populations of sheep and goats are characterized by polyceraty, the presence of more than two horns. Here, we characterize distinct POLYCERATE alleles in each species, both associated with defective HOXD1 function. We show that haploinsufficiency at this locus results in the splitting of horn bud primordia, likely following the abnormal extension of an initial morphogenetic field. These results highlight the key role played by this gene in headgear patterning and illustrate the evolutionary co-option of a gene involved in the early development of bilateria to properly fix the position and number of these distinctive organs of Bovidae.


Assuntos
Evolução Biológica , Cabras/genética , Proteínas de Homeodomínio/genética , Cornos , Ovinos/genética , Animais , Biometria , Regulação da Expressão Gênica no Desenvolvimento , Cabras/embriologia , Cabras/metabolismo , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos Transgênicos , Mutação , Ovinos/embriologia , Ovinos/metabolismo
2.
PLoS Biol ; 17(4): e3000185, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30947255

RESUMO

Dmrt1 is a highly conserved transcription factor, which is critically involved in regulation of gonad development of vertebrates. In medaka, a duplicate of dmrt1-acting as master sex-determining gene-has a tightly timely and spatially controlled gonadal expression pattern. In addition to transcriptional regulation, a sequence motif in the 3' UTR (D3U-box) mediates transcript stability of dmrt1 mRNAs from medaka and other vertebrates. We show here that in medaka, two RNA-binding proteins with antagonizing properties target this D3U-box, promoting either RNA stabilization in germ cells or degradation in the soma. The D3U-box is also conserved in other germ-cell transcripts, making them responsive to the same RNA binding proteins. The evolutionary conservation of the D3U-box motif within dmrt1 genes of metazoans-together with preserved expression patterns of the targeting RNA binding proteins in subsets of germ cells-suggest that this new mechanism for controlling RNA stability is not restricted to fishes but might also apply to other vertebrates.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Oryzias/genética , Processos de Determinação Sexual/genética , Regiões 3' não Traduzidas/genética , Animais , Evolução Biológica , Feminino , Proteínas de Peixes/genética , Células Germinativas/metabolismo , Masculino , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Estabilidade de RNA/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vertebrados/metabolismo
3.
PLoS Genet ; 15(2): e1007909, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30735494

RESUMO

Gonad differentiation is a crucial step conditioning the future fertility of individuals and most of the master genes involved in this process have been investigated in detail. However, transcriptomic analyses of developing gonads from different animal models have revealed that hundreds of genes present sexually dimorphic expression patterns. DMXL2 was one of these genes and its function in mammalian gonads was unknown. We therefore investigated the phenotypes of total and gonad-specific Dmxl2 knockout mouse lines. The total loss-of-function of Dmxl2 was lethal in neonates, with death occurring within 12 hours of birth. Dmxl2-knockout neonates were weak and did not feed. They also presented defects of olfactory information transmission and severe hypoglycemia, suggesting that their premature death might be due to global neuronal and/or metabolic deficiencies. Dmxl2 expression in the gonads increased after birth, during follicle formation in females and spermatogenesis in males. DMXL2 was detected in both the supporting and germinal cells of both sexes. As Dmxl2 loss-of-function was lethal, only limited investigations of the gonads of Dmxl2 KO pups were possible. They revealed no major defects at birth. The gonadal function of Dmxl2 was then assessed by conditional deletions of the gene in gonadal supporting cells, germinal cells, or both. Conditional Dmxl2 ablation in the gonads did not impair fertility in males or females. By contrast, male mice with Dmxl2 deletions, either throughout the testes or exclusively in germ cells, presented a subtle testicular phenotype during the first wave of spermatogenesis that was clearly detectable at puberty. Indeed, Dmxl2 loss-of-function throughout the testes or in germ cells only, led to sperm counts more than 60% lower than normal and defective seminiferous tubule architecture. Transcriptomic and immunohistochemichal analyses on these abnormal testes revealed a deregulation of Sertoli cell phagocytic activity related to germ cell apoptosis augmentation. In conclusion, we show that Dmxl2 exerts its principal function in the testes at the onset of puberty, although its absence does not compromise male fertility in mice.


Assuntos
Proteínas do Tecido Nervoso/genética , Espermatogênese/genética , Espermatozoides/fisiologia , Animais , Apoptose/genética , Feminino , Fertilidade/genética , Células Germinativas/fisiologia , Gônadas/fisiologia , Infertilidade Feminina/genética , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Túbulos Seminíferos/fisiologia , Células de Sertoli/fisiologia , Testículo/fisiologia
4.
Proc Natl Acad Sci U S A ; 115(50): 12781-12786, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30463951

RESUMO

Evolutionary novelties require rewiring of transcriptional networks and/or the evolution of new gene functions. Sex determination (SD), one of the most plastic evolutionary processes, requires such novelties. Studies on the evolution of vertebrate SD revealed that new master SD genes are generally recruited from genes involved in the downstream SD regulatory genetic network. Only a single exception to this rule is currently known in vertebrates: the intriguing case of the salmonid master SD gene (sdY), which arose from duplication of an immune-related gene. This exception immediately posed the question of how a gene outside from the classical sex differentiation cascade could acquire its function as a male SD gene. Here we show that SdY became integrated in the classical vertebrate sex differentiation cascade by interacting with the Forkhead box domain of the female-determining transcription factor, Foxl2. In the presence of Foxl2, SdY is translocated to the nucleus where the SdY:Foxl2 complex prevents activation of the aromatase (cyp19a1a) promoter in cooperation with Nr5a1 (Sf1). Hence, by blocking a positive loop of regulation needed for the synthesis of estrogens in the early differentiating gonad, SdY disrupts a preset female differentiation pathway, consequently allowing testicular differentiation to proceed. These results also suggest that the evolution of unusual vertebrate master sex determination genes recruited from outside the classical pathway like sdY is strongly constrained by their ability to interact with the canonical gonadal differentiation pathway.


Assuntos
Redes Reguladoras de Genes/genética , Gônadas/fisiologia , Oncorhynchus mykiss/genética , Processos de Determinação Sexual/genética , Diferenciação Sexual/genética , Animais , Aromatase/genética , Diferenciação Celular/genética , Núcleo Celular/genética , Estrogênios/genética , Feminino , Proteína Forkhead Box L2/genética , Masculino , Regiões Promotoras Genéticas/genética , Testículo/metabolismo , Translocação Genética/genética
5.
Nucleic Acids Res ; 45(12): 7191-7211, 2017 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-28472341

RESUMO

In mammalian embryonic gonads, SOX9 is required for the determination of Sertoli cells that orchestrate testis morphogenesis. To identify genetic networks directly regulated by SOX9, we combined analysis of SOX9-bound chromatin regions from murine and bovine foetal testes with sequencing of RNA samples from mouse testes lacking Sox9. We found that SOX9 controls a conserved genetic programme that involves most of the sex-determining genes. In foetal testes, SOX9 modulates both transcription and directly or indirectly sex-specific differential splicing of its target genes through binding to genomic regions with sequence motifs that are conserved among mammals and that we called 'Sertoli Cell Signature' (SCS). The SCS is characterized by a precise organization of binding motifs for the Sertoli cell reprogramming factors SOX9, GATA4 and DMRT1. As SOX9 biological role in mammalian gonads is to determine Sertoli cells, we correlated this genomic signature with the presence of SOX9 on chromatin in foetal testes, therefore equating this signature to a genomic bar code of the fate of foetal Sertoli cells. Starting from the hypothesis that nuclear factors that bind to genomic regions with SCS could functionally interact with SOX9, we identified TRIM28 as a new SOX9 partner in foetal testes.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Morfogênese/genética , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Fatores de Transcrição SOX9/genética , Células de Sertoli/metabolismo , Transcriptoma , Animais , Bovinos , Cromatina/química , Cromatina/metabolismo , Embrião de Mamíferos , Feminino , Feto , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Redes Reguladoras de Genes , Masculino , Camundongos , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOX9/metabolismo , Análise de Sequência de RNA , Células de Sertoli/citologia , Processos de Determinação Sexual , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 28 com Motivo Tripartido
6.
Dev Biol ; 406(2): 158-71, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26358182

RESUMO

Topaz1 (Testis and Ovary-specific PAZ domain gene 1) is a germ cell specific gene highly conserved in vertebrates. The putative protein TOPAZ1 contains a PAZ domain, specifically found in PIWI, Argonaute and Zwille proteins. Consequently, Topaz1 is supposed to have a role during gametogenesis and may be involved in the piRNA pathway and contribute to silencing of transposable elements and maintenance of genome integrity. Here we report Topaz1 inactivation in mouse. Female fertility was not affected, but male sterility appeared exclusively in homozygous mutants in accordance with the high expression of Topaz1 in male germ cells. Pachytene Topaz1--deficient spermatocytes progress through meiosis without either derepression of retrotransposons or MSCI dysfunction, but become arrested before the post-meiotic round spermatid stage with extensive apoptosis. Consequently, an absence of spermatids and spermatozoa was observed in Topaz1(-/-) testis. Histological analysis also revealed that disturbances of spermatogenesis take place between post natal days 15 and 20, during the first wave of male meiosis and before the generation of haploid germ cells. Transcriptomic analysis at these two stages showed that TOPAZ1 influences the expression of one hundred transcripts, most of which are up-regulated in mutant testis at post natal day 20. Our results also showed that 10% of these transcripts are long non-coding RNA. This suggests that a highly regulated balance of lncRNAs seems to be essential during spermatogenesis for induction of appropriate male gamete production.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Infertilidade Masculina/genética , Meiose/genética , Animais , Perfilação da Expressão Gênica , Técnicas Histológicas , Masculino , Meiose/fisiologia , Camundongos , Camundongos Knockout , RNA Longo não Codificante/genética , Testículo/metabolismo
7.
BMC Genomics ; 16: 316, 2015 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-25895751

RESUMO

BACKGROUND: Cattle populations are characterized by regular outburst of genetic defects as a result of the extensive use of elite sires. The causative genes and mutations can nowadays be rapidly identified by means of genome-wide association studies combined with next generation DNA sequencing, provided that the causative mutations are conventional loss-of-function variants. We show in this work how the combined use of next generation DNA and RNA sequencing allows for the rapid identification of otherwise difficult to identify splice-site variants. RESULTS: We report the use of haplotype-based association mapping to identify a locus on bovine chromosome 10 that underlies autosomal recessive arthrogryposis in Belgian Blue Cattle. We identify 31 candidate mutations by resequencing the genome of four cases and 15 controls at ~10-fold depth. By analyzing RNA-Seq data from a carrier fetus, we observe skipping of the second exon of the PIGH gene, which we confirm by RT-PCR to be fully penetrant in tissues from affected calves. We identify - amongst the 31 candidate variants - a C-to-G transversion in the first intron of the PIGH gene (c211-10C > G) that is predicted to affect its acceptor splice-site. The resulting PIGH protein is likely to be non-functional as it lacks essential domains, and hence to cause arthrogryposis. CONCLUSIONS: This work illustrates how the growing arsenal of genome exploration tools continues to accelerate the identification of an even broader range of disease causing mutations, therefore improving the management and control of genetic defects in livestock.


Assuntos
Artrogripose/genética , Proteínas de Membrana/genética , Splicing de RNA , Animais , Artrogripose/etiologia , Artrogripose/veterinária , Bélgica , Bovinos , Mapeamento Cromossômico , Éxons , Genoma , Estudo de Associação Genômica Ampla , Genótipo , Glicosilfosfatidilinositóis/metabolismo , Haplótipos , Sequenciamento de Nucleotídeos em Larga Escala , Íntrons , Proteínas de Membrana/metabolismo , Polimorfismo de Nucleotídeo Único , Sítios de Splice de RNA , Análise de Sequência de DNA , Análise de Sequência de RNA
8.
Development ; 139(23): 4461-72, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23095882

RESUMO

The gonad arises from the thickening of the coelomic epithelium and then commits into the sex determination process. Testis differentiation is activated by the expression of the Y-linked gene Sry, which promotes cell proliferation and differentiation of Sertoli cells, the supporting cells of the testis. In absence of Sry (XX individuals), activation of WNT/CTNNB1 signalling, via the upregulation of Rspo1 and Wnt4, promotes ovarian differentiation. However, Rspo1 and Wnt4 are expressed in the early undifferentiated gonad of both sexes, and Axin2-lacZ, a reporter of canonical WNT/CTNNB1 signalling, is expressed in the coelomic region of the E11.5 gonadal primordium, suggesting a role of these factors in early gonadal development. Here, we show that simultaneous ablation of Rspo1 and Wnt4 impairs proliferation of the cells of the coelomic epithelium, reducing the number of progenitors of Sertoli cells in XY mutant gonads. As a consequence, in XY Wnt4(-/-); Rspo1(-/-) foetuses, this leads to the differentiation of a reduced number of Sertoli cells and the formation of a hypoplastic testis exhibiting few seminiferous tubules. Hence, this study identifies Rspo1 and Wnt4 as two new regulators of cell proliferation in the early gonad regardless of its sex, in addition to the specific role of these genes in ovarian differentiation.


Assuntos
Gônadas/embriologia , Processos de Determinação Sexual , Trombospondinas/metabolismo , Proteína Wnt4/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células , Feminino , Gônadas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovário/embriologia , Fatores de Transcrição SOX9/biossíntese , Células de Sertoli/metabolismo , Transdução de Sinais , Testículo/embriologia , Trombospondinas/genética , Proteína Wnt4/genética
9.
Biol Reprod ; 91(6): 153, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25395674

RESUMO

FOXL2 loss of function in goats leads to the early transdifferentiation of ovaries into testes, then to the full sex reversal of XX homozygous mutants. By contrast, Foxl2 loss of function in mice induces an arrest of follicle formation after birth, followed by complete female sterility. In order to understand the molecular role of FOXL2 during ovarian differentiation in the goat species, putative FOXL2 target genes were determined at the earliest stage of gonadal sex-specific differentiation by comparing the mRNA profiles of XX gonads expressing the FOXL2 protein or not. Of these 163 deregulated genes, around two-thirds corresponded to testicular genes that were up-regulated when FOXL2 was absent, and only 19 represented female-associated genes, down-regulated in the absence of FOXL2. FOXL2 should therefore be viewed as an antitestis gene rather than as a female-promoting gene. In particular, the key testis-determining gene DMRT1 was found to be up-regulated ahead of SOX9, thus suggesting in goats that SOX9 primary up-regulation may require DMRT1. Overall, our results equated to FOXL2 being an antitestis gene, allowing us to propose an alternative model for the sex-determination process in goats that differs slightly from that demonstrated in mice.


Assuntos
Transtornos Testiculares 46, XX do Desenvolvimento Sexual/genética , Fatores de Transcrição Forkhead/genética , Genitália Feminina/metabolismo , Cabras/fisiologia , Fatores de Transcrição SOX9/genética , Fatores de Transcrição/genética , Transtornos Testiculares 46, XX do Desenvolvimento Sexual/veterinária , Animais , Animais Geneticamente Modificados , Transdiferenciação Celular , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genitália Feminina/embriologia , Cabras/genética , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Ovário/metabolismo , Processos de Determinação Sexual/genética , Testículo/metabolismo , Regulação para Cima
10.
Nat Commun ; 15(1): 5574, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38956430

RESUMO

The biomedical research community addresses reproducibility challenges in animal studies through standardized nomenclature, improved experimental design, transparent reporting, data sharing, and centralized repositories. The ARRIVE guidelines outline documentation standards for laboratory animals in experiments, but genetic information is often incomplete. To remedy this, we propose the Laboratory Animal Genetic Reporting (LAG-R) framework. LAG-R aims to document animals' genetic makeup in scientific publications, providing essential details for replication and appropriate model use. While verifying complete genetic compositions may be impractical, better reporting and validation efforts enhance reliability of research. LAG-R standardization will bolster reproducibility, peer review, and overall scientific rigor.


Assuntos
Animais de Laboratório , Guias como Assunto , Animais , Animais de Laboratório/genética , Reprodutibilidade dos Testes , Projetos de Pesquisa , Experimentação Animal/normas , Pesquisa Biomédica/normas
11.
Elife ; 122023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37847154

RESUMO

DMRT1 is the testis-determining factor in several species of vertebrates, but its involvement in mammalian testes differentiation, where SRY is the testis-determining gene, remains ambiguous. So far, DMRT1 loss-of-function has been described in two mammalian species and induces different phenotypes: Disorders of Sex Development (46, XY DSD) in men and male infertility in mice. We thus abolished DMRT1 expression by CRISPR/Cas9 in a third species of mammal, the rabbit. First, we observed that gonads from XY DMRT1-/- rabbit fetuses differentiated like ovaries, highlighting that DMRT1 is involved in testis determination. In addition to SRY, DMRT1 is required in the supporting cells to increase the expression of the SOX9 gene, which heads the testicular genetic cascade. Second, we highlighted another function of DMRT1 in the germline since XX and XY DMRT1-/- ovaries did not undergo meiosis and folliculogenesis. XX DMRT1-/- adult females were sterile, showing that DMRT1 is also crucial for female fertility. To conclude, these phenotypes indicate an evolutionary continuum between non-mammalian vertebrates such as birds and non-rodent mammals. Furthermore, our data support the potential involvement of DMRT1 mutations in different human pathologies, such as 46, XY DSD as well as male and female infertility.


Animals that reproduce sexually have organs called gonads, the ovaries and testes, which produce eggs and sperm. These organs, which are different in males and females, originate from the same cells during the development of the embryo. As a general rule, the chromosomal sex of an embryo, which gets determined at fertilization, leads to the activation and repression of specific genes. This in turn, controls whether the cells that will form the gonads will differentiate to develop testes or ovaries. Disruption of the key genes involved in the differentiation of the gonads can lead to fertility problems, and in some cases, it can cause the gonads to develop in the 'opposite' direction, resulting in a sex reversal. Identifying these genes is therefore essential to know how to maintain or restore fertility. DMRT1 is a gene that drives the differentiation of gonadal cells into the testicular pathway in several species of animals with backbones, including species of fish, frogs and birds. However, its role in mammals ­ where testis differentiation is driven by a different gene called SRY ­ is not well understood. Indeed, when DMRT1 is disrupted in male humans it leads to disorders of sex development, while disrupting this gene in male mice causes infertility. To obtain more information about the roles of DMRT1 in mammalian species, Dujardin et al. disrupted the gene in a third species of mammal: the rabbit. Dujardin et al. observed that chromosomally-male rabbits lacking DMRT1 developed ovaries instead of testes, showing that in rabbits, both SRY and DMRT1 are both required to produce testes. Additionally, this effect is similar to what is seen in humans, suggesting that rabbits may be a better model for human gonadal differentiation than mice are. Additionally, Dujardin et al. were also able to show that in female rabbits, lack of DMRT1 led to infertility, an effect that had not been previously described in other species. The results of Dujardin et al. may lead to better models for gonadal development in humans, involving DMRT1 in the differentiation of testes. Interestingly, they also suggest the possibility that mutations in this gene may be responsible for some cases of infertility in women. Overall, these findings indicate that DMRT1 is a key fertility gene.


Assuntos
Transtorno 46,XY do Desenvolvimento Sexual , Testículo , Animais , Feminino , Masculino , Coelhos , Transtorno 46,XY do Desenvolvimento Sexual/genética , Transtorno 46,XY do Desenvolvimento Sexual/metabolismo , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/metabolismo , Mamíferos/genética , Processos de Determinação Sexual/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Testículo/metabolismo
12.
Transgenic Res ; 21(6): 1183-90, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22528467

RESUMO

Compared to experiments involving pigs, cows and/or sheep, transgenesis applied to goats is probably less advertised. However, recent successes and increasing amount of dedicated research make this species of special interest for ongoing biological and physiological questions on genome engineering in large animals. This short review aims at highlighting the current applications and limitations of the goat genome manipulation.


Assuntos
Animais Geneticamente Modificados , Modelos Animais de Doenças , Doenças das Cabras/genética , Cabras/genética , Animais , Genoma
13.
Sex Dev ; 16(2-3): 194-206, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35835064

RESUMO

Recently, a series of recurrent missense variants in the RNA-helicase DHX37 have been reported associated with either 46,XY gonadal dysgenesis, 46,XY testicular regression syndrome (TRS), or anorchia. All affected children have non-syndromic forms of disorders/differences of sex development (DSD). These variants, which involve highly conserved amino acids within known functional domains of the protein, are predicted by in silico tools to have a deleterious effect on helicase function. DHX37 is required for ribosome biogenesis in eukaryotes, and how these variants cause DSD is unclear. The relationship between DHX37 and human congenital disorders is complex as compound heterozygous as well as de novo heterozygous missense variants in DHX37 are also associated with a complex congenital developmental syndrome (NEDBAVC, neurodevelopmental disorder with brain anomalies and with or without vertebral or cardiac anomalies; OMIM 618731), consisting of microcephaly, global developmental delay, seizures, facial dysmorphia, and kidney and cardiac anomalies. Here, we will give a brief overview of ribosome biogenesis and the role of DHX37 in this process. We will discuss variants in DHX37, their contribution to human disease in the general context of human ribosomopathies, and the possible disease mechanisms that may be involved.


Assuntos
Disgenesia Gonadal 46 XY , RNA Helicases , Humanos , Disgenesia Gonadal 46 XY/genética , Heterozigoto , Mutação de Sentido Incorreto , RNA Helicases/genética
14.
Genes (Basel) ; 13(11)2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36360307

RESUMO

Estrogens are steroid hormones produced by the aromatization of androgens by the aromatase enzyme, encoded by the CYP19A1 gene. Although generally referred to as "female sex hormones", estrogen is also produced in the adult testes of many mammals, including humans. To better understand the function of estrogens in the male, we used the rabbit model which is an important biomedical model. First, the expression of CYP19A1 transcripts was localized mainly in meiotic germ cells. Thus, testicular estrogen appears to be produced inside the seminiferous tubules. Next, the cells expressing ESR1 and ESR2 were identified, showing that estrogens could exert their function on post-meiotic germ cells in the tubules and play a role during sperm maturation, since ESR1 and ESR2 were detected in the cauda epididymis. Then, CRISPR/Cas9 CYP19A1-/- genetically modified rabbits were analyzed. CYP19A1-/- males showed decreased fertility with lower sperm count associated with hypo-spermatogenesis and lower spermatid number. Germ/sperm cell DNA methylation was unchanged, while sperm parameters were affected as CYP19A1-/- males exhibited reduced sperm motility associated with increased flagellar defects. In conclusion, testicular estrogens could be involved in the spermatocyte-spermatid transition in the testis, and in the acquisition of sperm motility in the epididymis.


Assuntos
Sêmen , Testículo , Humanos , Animais , Masculino , Coelhos , Feminino , Testículo/metabolismo , Sêmen/metabolismo , Motilidade dos Espermatozoides/genética , Espermatogênese/genética , Estrogênios/metabolismo , Mamíferos
15.
Endocrinology ; 163(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34614143

RESUMO

AROMATASE is encoded by the CYP19A1 gene and is the cytochrome enzyme responsible for estrogen synthesis in vertebrates. In most mammals, a peak of CYP19A1 gene expression occurs in the fetal XX gonad when sexual differentiation is initiated. To elucidate the role of this peak, we produced 3 lines of TALEN genetically edited CYP19A1 knockout (KO) rabbits that were devoid of any estradiol production. All the KO XX rabbits developed as females with aberrantly small ovaries in adulthood, an almost empty reserve of primordial follicles, and very few large antrum follicles. Ovulation never occurred. Our histological, immunohistological, and transcriptomic analyses showed that the estradiol surge in the XX fetal rabbit gonad is not essential to its determination as an ovary, or for meiosis. However, it is mandatory for the high proliferation and differentiation of both somatic and germ cells, and consequently for establishment of the ovarian reserve.


Assuntos
Estrogênios/metabolismo , Ovário/embriologia , Ovário/fisiologia , Processos de Determinação Sexual/fisiologia , Animais , Hormônio Antimülleriano/metabolismo , Diferenciação Celular , Proliferação de Células , Família 19 do Citocromo P450/metabolismo , Estradiol/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Gônadas , Mutação INDEL , Folículo Ovariano/fisiologia , Ovulação , Fenótipo , Coelhos , Diferenciação Sexual/fisiologia , Testosterona/metabolismo
16.
Med Sci (Paris) ; 27(10): 859-65, 2011 Oct.
Artigo em Francês | MEDLINE | ID: mdl-22027423

RESUMO

Our knowledge on sex differentiation in mammals has considerably progressed during the last decennials, beginning with the discovery of the testis-determining factor. Here, the morphogenetic processes involved in the early gonadic switch will be presented, together with the major genes involved in testis and ovary formation. Existing differences between the widely used mouse model and other mammals, such as human and goat, will be highlighted.


Assuntos
Processos de Determinação Sexual/fisiologia , Diferenciação Sexual/fisiologia , Animais , Feminino , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/fisiologia , Dosagem de Genes , Genes sry , Cabras/fisiologia , Hormônios/fisiologia , Humanos , Masculino , Mamíferos/fisiologia , Camundongos/fisiologia , Modelos Biológicos , Morfogênese , Ovário/embriologia , Ovário/crescimento & desenvolvimento , Caracteres Sexuais , Cromossomos Sexuais/fisiologia , Processos de Determinação Sexual/genética , Diferenciação Sexual/genética , Especificidade da Espécie , Testículo/embriologia , Testículo/crescimento & desenvolvimento
17.
Dev Dyn ; 239(12): 3324-35, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20941779

RESUMO

The testis-determining gene SRY is not well-conserved among mammals, and particularly between mouse and other mammals. To evaluate SRY function in a nonrodent species, we produced an antibody against goat SRY and used it to investigate the expression pattern of SRY throughout goat testicular development. By contrast with the mouse, SRY is primarily expressed in most cells of XY genital-ridges and not solely in pre-Sertoli cells. Between cord formation and prepuberty, SRY remains expressed in both Sertoli and germinal cells. During adulthood, SRY expression declines and then disappears from meiotic germ cells, only remaining present at low levels in some spermatogonia. Unlike the germinal lineage, SRY continues to be highly expressed in adult Sertoli cells with a typical nuclear staining. Our data indicate that in goat, the role of SRY may not be limited to testis determination and could have other functions in testicular maintenance and hence male fertility.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteína da Região Y Determinante do Sexo/metabolismo , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Feminino , Cabras , Masculino , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Diferenciação Sexual/genética , Diferenciação Sexual/fisiologia , Proteína da Região Y Determinante do Sexo/genética , Espermatogônias/citologia , Espermatogônias/metabolismo , Testículo/embriologia
18.
Front Cell Dev Biol ; 9: 700290, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34277642

RESUMO

Spermatogenesis involves coordinated processes, including meiosis, to produce functional gametes. We previously reported Topaz1 as a germ cell-specific gene highly conserved in vertebrates. Topaz1 knockout males are sterile with testes that lack haploid germ cells because of meiotic arrest after prophase I. To better characterize Topaz1 -/- testes, we used RNA-sequencing analyses at two different developmental stages (P16 and P18). The absence of TOPAZ1 disturbed the expression of genes involved in microtubule and/or cilium mobility, biological processes required for spermatogenesis. Moreover, a quarter of P18 dysregulated genes are long non-coding RNAs (lncRNAs), and three of them are testis-specific and located in spermatocytes, their expression starting between P11 and P15. The suppression of one of them, 4939463O16Rik, did not alter fertility although sperm parameters were disturbed and sperm concentration fell. The transcriptome of P18-4939463O16Rik -/- testes was altered and the molecular pathways affected included microtubule-based processes, the regulation of cilium movement and spermatogenesis. The absence of TOPAZ1 protein or 4930463O16Rik produced the same enrichment clusters in mutant testes despite a contrasted phenotype on male fertility. In conclusion, although Topaz1 is essential for the meiosis in male germ cells and regulate the expression of numerous lncRNAs, these studies have identified a Topaz1 regulated lncRNA (4930463O16Rik) that is key for both sperm production and motility.

19.
Hum Mol Genet ; 17(7): 1010-9, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18158309

RESUMO

Polyalanine (polyAla) tract expansions have been associated with an increasing number of human diseases. Here, we have undertaken a functional study of the effects of polyAla expansions in the context of the transcription factor FOXL2, involved in cranio-facial and ovarian development. Using two cellular models, we show that FOXL2 polyAla expansions lead to protein mislocalization and aggregation in a length-dependent manner. The fraction of cells containing cytoplasmic staining displays a sigmoidal relationship with respect to the length of the polyAla tract, suggesting the existence of a threshold length above which protein mislocalization occurs. The existence of such a threshold might be rationalized if we consider that the longer the polyAla tract is, the higher its tendency to misfolding or to inducing spurious interactions with cytoplasmic components. To study the intranuclear dynamics of polyAla-expanded FOXL2, we performed fluorescence recovery after photobleaching experiments. The most unexpected result concerned the pathogenic protein containing 19 Ala residues in the run, which was virtually immobile, although this variant does not present a classical aggregation pattern. Luciferase assays and real time RT-PCR of many potential target genes showed that polyAla expansions induce different losses of activity according to the target promoters tested. We provide molecular explanations for these findings. Although our main focus is the mechanisms of pathogenesis of polyAla-expanded proteins, we discuss the potential relevance of polyAla length variation in micro- and macroevolution because polyAla-containing proteins tend to be transcription factors.


Assuntos
Anormalidades Craniofaciais/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Ovário/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Expansão das Repetições de Trinucleotídeos , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Citoplasma/química , Citoplasma/metabolismo , Feminino , Recuperação de Fluorescência Após Fotodegradação , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/química , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/metabolismo , Humanos , Espaço Intranuclear/metabolismo , Microscopia de Fluorescência , Ovário/anormalidades , Ovário/embriologia , Peptídeos/metabolismo , Transporte Proteico , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/análise , Transfecção
20.
Sci Adv ; 6(21): eaaz1261, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32494737

RESUMO

In mammals, the timing of meiosis entry is regulated by signals from the gonadal environment. All-trans retinoic acid (ATRA) signaling is considered the key pathway that promotes Stra8 (stimulated by retinoic acid 8) expression and, in turn, meiosis entry. This model, however, is debated because it is based on analyzing the effects of exogenous ATRA on ex vivo gonadal cultures, which not accurately reflects the role of endogenous ATRA. Aldh1a1 and Aldh1a2, two retinaldehyde dehydrogenases synthesizing ATRA, are expressed in the mouse ovaries when meiosis initiates. Contrary to the present view, here, we demonstrate that ATRA-responsive cells are scarce in the ovary. Using three distinct gene deletion models for Aldh1a1;Aldh1a2;Aldh1a3, we show that Stra8 expression is independent of ATRA production by ALDH1A proteins and that germ cells progress through meiosis. Together, these data demonstrate that ATRA signaling is dispensable for instructing meiosis initiation in female germ cells.


Assuntos
Meiose , Ovário , Animais , Feminino , Células Germinativas/metabolismo , Mamíferos/metabolismo , Camundongos , Ovário/metabolismo , Proteínas/metabolismo , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA