Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 389
Filtrar
1.
Front Endocrinol (Lausanne) ; 15: 1343759, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38752176

RESUMO

Syndromic autism spectrum conditions (ASC), such as Klinefelter syndrome, also manifest hypogonadism. Compared to the popular Extreme Male Brain theory, the Enhanced Perceptual Functioning model explains the connection between ASC, savant traits, and giftedness more seamlessly, and their co-emergence with atypical sexual differentiation. Overexcitability of primary sensory inputs generates a relative enhancement of local to global processing of stimuli, hindering the abstraction of communication signals, in contrast to the extraordinary local information processing skills in some individuals. Weaker inhibitory function through gamma-aminobutyric acid type A (GABAA) receptors and the atypicality of synapse formation lead to this difference, and the formation of unique neural circuits that process external information. Additionally, deficiency in monitoring inner sensory information leads to alexithymia (inability to distinguish one's own emotions), which can be caused by hypoactivity of estrogen and oxytocin in the interoceptive neural circuits, comprising the anterior insular and cingulate gyri. These areas are also part of the Salience Network, which switches between the Central Executive Network for external tasks and the Default Mode Network for self-referential mind wandering. Exploring the possibility that estrogen deficiency since early development interrupts GABA shift, causing sensory processing atypicality, it helps to evaluate the co-occurrence of ASC with attention deficit hyperactivity disorder, dyslexia, and schizophrenia based on phenotypic and physiological bases. It also provides clues for understanding the common underpinnings of these neurodevelopmental disorders and gifted populations.


Assuntos
Androgênios , Transtorno do Espectro Autista , Estrogênios , Humanos , Androgênios/deficiência , Androgênios/metabolismo , Estrogênios/metabolismo , Estrogênios/deficiência , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Masculino , Diferenciação Sexual/fisiologia , Síndrome de Klinefelter/fisiopatologia , Síndrome de Klinefelter/metabolismo , Percepção/fisiologia , Encéfalo/metabolismo
2.
Gen Comp Endocrinol ; 353: 114512, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38582176

RESUMO

Eels are gonochoristic species whose gonadal differentiation initiates at the yellow eel stage and is influenced by environmental factors. We revealed some sex-related genes were sex dimorphically expressed in gonads during gonadal sex differentiation of Japanese eel (Anguilla japonica); however, the expression of sex-related genes in the brain-pituitary during gonadal sex differentiation in eels is still unclear. This study aimed to investigate the sex-related gene expressions in the brain-pituitary and tried to clarify their roles in the brain and gonads during gonadal sex differentiation. Based on our previous histological study, the control eels developed as males, and estradiol-17ß (E2) was used for feminization. Our results showed that during testicular differentiation, the brain cyp19a1 transcripts and aromatase proteins were increased significantly; moreover, the cyp19a1, sf-1, foxl2s, and esrs (except gperb) transcripts in the midbrain/pituitary also were increased significantly. Forebrain gnrh1 transcripts increased slightly during gonadal differentiation of both sexes, but the gnrhr1b and gnrhr2 transcripts in the midbrain/pituitary were stable during gonadal differentiation. The expression levels of gths and gh in the midbrain/pituitary were significantly increased during testicular differentiation and were much higher in males than in E2-feminized females. These results implied that endogenous estrogens might play essential roles in the brain/pituitary during testicular differentiation, sf-1, foxl2s, and esrs may have roles in cyp19a1 regulation in the midbrain/pituitary of Japanese eels. For the GnRH-GTH axis, gths, especially fshb, may be regulated by esrs and involved in regulating testicular differentiation and development in Japanese eels.


Assuntos
Aromatase , Encéfalo , Hipófise , Diferenciação Sexual , Animais , Diferenciação Sexual/genética , Diferenciação Sexual/fisiologia , Masculino , Aromatase/genética , Aromatase/metabolismo , Feminino , Encéfalo/metabolismo , Hipófise/metabolismo , Anguilla/genética , Anguilla/metabolismo , Anguilla/crescimento & desenvolvimento , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Testículo/metabolismo , Gônadas/metabolismo , Gônadas/crescimento & desenvolvimento
3.
Front Endocrinol (Lausanne) ; 13: 919670, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35909548

RESUMO

The Sertoli cells of the testes play an essential role during gonadal development, in addition to supporting subsequent germ cell survival and spermatogenesis. Anti-Müllerian hormone (AMH) is a member of the TGF-ß superfamily, which is secreted by immature Sertoli cells from the 8th week of fetal gestation. lnhibin B is a glycoprotein, which is produced by the Sertoli cells from early in fetal development. In people with a Difference or Disorder of Sex Development (DSD), these hormones may be useful to determine the presence of testicular tissue and potential for spermatogenesis. However, fetal Sertoli cell development and function is often dysregulated in DSD conditions and altered production of Sertoli cell hormones may be detected throughout the life course in these individuals. As such this review will consider the role of AMH and inhibin B in individuals with DSD.


Assuntos
Hormônio Antimülleriano , Transtornos do Desenvolvimento Sexual , Inibinas , Células de Sertoli , Diferenciação Sexual , Hormônio Antimülleriano/genética , Hormônio Antimülleriano/metabolismo , Transtornos do Desenvolvimento Sexual/genética , Transtornos do Desenvolvimento Sexual/metabolismo , Humanos , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/metabolismo , Inibinas/genética , Inibinas/metabolismo , Masculino , Células de Sertoli/metabolismo , Diferenciação Sexual/fisiologia , Espermatogênese/fisiologia , Testículo/metabolismo
4.
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
5.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34958103

RESUMO

The specific role of gonadotropin-releasing hormone (GnRH) on brain sexual differentiation remains unclear. To investigate whether gonadotropin and, in turn, testosterone (T) secretion is regulated by GnRH during the critical period for brain differentiation in sheep fetuses, we attempted to selectively suppress pituitary-testicular activation during midgestation with the long-acting GnRH antagonist degarelix. Fetuses received subcutaneous injections of the antagonist or vehicle on day 62 of gestation. After 2 to 3 weeks we examined consequences of the intervention on baseline and GnRH-stimulated plasma luteinizing hormone (LH) and T levels. In addition, we measured the effect of degarelix-treatment on messenger RNA (mRNA) expression for the pituitary gonadotropins and key gonadal steroidogenic enzymes. Baseline and GnRH-stimulated plasma LH levels were significantly suppressed in degarelix-treated male and female fetuses compared to control values. Similarly, T concentrations were suppressed in degarelix-treated males. The percentage of LHß-immunoreactive cells colocalizing c-fos was significantly reduced by degarelix treatment indicating that pituitary sensitivity was inhibited. Degarelix treatment also led to the significant suppression of mRNA expression coding for the pituitary gonadotropin subunits and for the gonadal enzymes involved in androgen synthesis. These findings demonstrate that pharmacologic inhibition of GnRH early in gestation results in suppression of LH secretion and deficits in the plasma T levels of male lamb fetuses. We conclude that GnRH signaling plays a pivotal role for regulating T exposure during the critical period of sheep gestation when the brain is masculinized. Thus, disturbance to gonadotropin secretion during this phase of gestation could have long-term consequence on adult sexual behaviors and fertility.


Assuntos
Idade Gestacional , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Gonadotropinas Hipofisárias/metabolismo , Oligopeptídeos/administração & dosagem , Adeno-Hipófise/embriologia , Ovinos/embriologia , Animais , Encéfalo/embriologia , Feminino , Sangue Fetal/química , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/fisiologia , Gonadotropinas Hipofisárias/genética , Injeções Subcutâneas/veterinária , Hormônio Luteinizante/sangue , Masculino , Ovário/química , Ovário/embriologia , Adeno-Hipófise/química , Adeno-Hipófise/efeitos dos fármacos , Gravidez , RNA Mensageiro/análise , Diferenciação Sexual/fisiologia , Testículo/química , Testículo/embriologia , Testosterona/sangue
6.
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
7.
Endocrinology ; 162(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34478502

RESUMO

Significant sex differences exist across cellular, tissue organization, and body system scales to serve the distinct sex-specific functions required for reproduction. They are present in all animals that reproduce sexually and have widespread impacts on normal development, aging, and disease. Observed from the moment of fertilization, sex differences are patterned by sexual differentiation, a lifelong process that involves mechanisms related to sex chromosome complement and the epigenetic and acute activational effects of sex hormones. In this mini-review, we examine evidence for sex differences in cellular responses to DNA damage, their underlying mechanisms, and how they might relate to sex differences in cancer incidence and response to DNA-damaging treatments.


Assuntos
Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Diferenciação Sexual/fisiologia , Adaptação Fisiológica/fisiologia , Envelhecimento/fisiologia , Animais , Feminino , Hormônios Esteroides Gonadais/metabolismo , Hormônios Esteroides Gonadais/fisiologia , Humanos , Masculino , Caracteres Sexuais
8.
Gene ; 805: 145910, 2021 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-34419567

RESUMO

Ethylene is an important regulatory phytohormone for sex differentiation and flower development. As the rate-limiting enzyme encoding genes in ethylene biosynthesis, ACS gene family has been well studied in cucumber; however, little is known in other cucurbit crops, such as melon and watermelon, which show diverse sex types in the field. Here, we identified and characterized eight ACS genes each in the genomes of melon and watermelon. According to the conserved serine residues at C-terminal, all the ACS genes could be characterized into three groups, which were supported by the exon-intron organizations and conserved motif distributions. ACS genes displayed diverse tissue-specific expression patterns among four melon and three watermelon sex types. Furthermore, a comparative expression analysis in the shoot apex identified orthologous pairs with potential functions in sex determination, e.g., ACS1s and ACS6s. All ACS orthologs in melon and watermelon exhibited similar expression patterns in monoecious and gynoecious genotypes, except for ACS11s and ACS12s. As expected, the majority of ACS genes were responsive to exogenous ethephon; however, some orthologs exhibited opposite expression patterns, such as ACS1s, ACS9s, and ACS10s. Collectively, our findings provide valuable ACS candidates related to flower development in various sex types of melon and watermelon.


Assuntos
Cucurbitaceae/genética , Etilenos/metabolismo , Liases/metabolismo , Diferenciação Sexual/genética , Citrullus/genética , Citrullus/metabolismo , Cucumis sativus/genética , Cucumis sativus/metabolismo , Cucurbitaceae/metabolismo , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica de Plantas/genética , Genes de Plantas/genética , Genótipo , Liases/genética , Filogenia , Proteínas de Plantas/genética , Diferenciação Sexual/fisiologia
9.
Endocrinology ; 162(7)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33963381

RESUMO

The WNT family of proteins is crucial in numerous developmental pathways and tissue homeostasis. WNT4, in particular, is uniquely implicated in the development of the female phenotype in the fetus, and in the maintenance of müllerian and reproductive tissues. WNT4 dysfunction or dysregulation can drive sex-reversal syndromes, highlighting the key role of WNT4 in sex determination. WNT4 is also critical in gynecologic pathologies later in life, including several cancers, uterine fibroids, endometriosis, and infertility. The role of WNT4 in normal decidualization, implantation, and gestation is being increasingly appreciated, while aberrant activation of WNT4 signaling is being linked both to gynecologic and breast cancers. Notably, single-nucleotide polymorphisms (SNPs) at the WNT4 gene locus are strongly associated with these pathologies and may functionally link estrogen and estrogen receptor signaling to upregulation and activation of WNT4 signaling. Importantly, in each of these developmental and disease states, WNT4 gene expression and downstream WNT4 signaling are regulated and executed by myriad tissue-specific pathways. Here, we review the roles of WNT4 in women's health with a focus on sex development, and gynecologic and breast pathologies, and our understanding of how WNT4 signaling is controlled in these contexts. Defining WNT4 functions provides a unique opportunity to link sex-specific signaling pathways to women's health and disease.


Assuntos
Doenças dos Genitais Femininos , Genitália Feminina , Proteína Wnt4/fisiologia , Saúde da Mulher , Animais , Neoplasias da Mama/genética , Feminino , Doenças dos Genitais Femininos/genética , Humanos , Glândulas Mamárias Humanas/fisiologia , Camundongos , Mutação , Polimorfismo de Nucleotídeo Único/genética , Gravidez , Diferenciação Sexual/fisiologia , Desenvolvimento Sexual/fisiologia , Útero/fisiologia , Proteína Wnt4/genética
10.
Indian J Pathol Microbiol ; 64(2): 390-393, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33851643

RESUMO

Gonadal dysgenesis is a distinct variety of Disorders of Sexual Differentiation (DSD) characterised by incomplete or defective formation of the gonads due to either structural or numerical anomalies of the sex chromosomes or mutations in the genes involved in the development of the gland. Here we present two such rare cases that presented during childhood. Both patients presented with ambiguous genitalia with a 45XO/46XY mosaic chromosome pattern. First case, an infant underwent laparoscopic excision of streak gonad, and a single stage hypospadias repair later. Second case, an adolescent who underwent gonadectomy as a child, presented with a mass which was excised and found to contain uterine and ovarian tissue; second stage hypospadias repair is being planned. Mixed gonadal dysgenesis usually presents with a unilateral testis, a streak gonad on the contralateral side and persistent mullerian structures. The most common karyotype noted is 45XO/46XY. These cases are known to have ambiguous external genitalia. The streak gonads have an increased malignant potential and thus, these patients should be carefully screened and followed up for gonadoblastoma.


Assuntos
Transtornos do Desenvolvimento Sexual/fisiopatologia , Disgenesia Gonadal Mista/fisiopatologia , Aberrações dos Cromossomos Sexuais , Diferenciação Sexual/fisiologia , Cariótipo Anormal , Adolescente , Deleção Cromossômica , Criptorquidismo/genética , Feminino , Humanos , Hipospadia/fisiopatologia , Lactente , Masculino , Mosaicismo , Linha Primitiva/embriologia , Testículo/anormalidades , Útero/anormalidades
11.
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
12.
J Endocrinol ; 247(1): R27-R44, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32755997

RESUMO

The spotted hyaena (Crocuta crocuta) is a unique species, even amongst the Hyaenidae. Extreme clitoral development in female spotted hyaenas challenges aspects of the accepted framework of sexual differentiation and reproductive function. They lack a vulva and instead urinate, copulate and give birth through a single, long urogenital canal that traverses a clitoris superficially resembling a penis. Recent and historical evidence is reviewed to describe our changing understanding of the biology of this species. Expanding upon observations from hyaenas in nature, much has been learned from studies utilising the captive colony at the University of California, Berkeley. The steroid environment of pregnancy is shaped by placental androgen and oestrogen secretion and a late gestational increase in sex hormone binding globulin, the regulated expression and steroid-binding characteristics of which are unique within the Hyaenidae. While initial external genital development is largely free of androgenic influence, the increase in testosterone concentrations in late gestation influences foetal development. Specifically, anti-androgen (AA) treatment of pregnant females reduced the developmental influence of androgens on their foetuses, resulting in reduced androstenedione concentrations in young females and easier birth through a 'feminised' clitoris, but precluded intromission and mating by 'feminised' male offspring, and altered social interactions. Insight into the costs and benefits of androgen exposure on spotted hyaena reproductive development, endocrinology and behaviour emphasises the delicate balance that sustains reproductive success, forces a re-evaluation of how we define masculine vs feminine sexual characteristics, and motivates reflection about the representative value of model species.


Assuntos
Genitália Feminina , Genitália Masculina , Hormônios Esteroides Gonadais/fisiologia , Hyaenidae , Reprodução/fisiologia , Diferenciação Sexual/fisiologia , Androgênios/fisiologia , Animais , Estrogênios/fisiologia , Feminino , Genitália Feminina/anatomia & histologia , Genitália Feminina/embriologia , Genitália Feminina/crescimento & desenvolvimento , Genitália Masculina/anatomia & histologia , Genitália Masculina/embriologia , Genitália Masculina/crescimento & desenvolvimento , Hyaenidae/anatomia & histologia , Hyaenidae/embriologia , Hyaenidae/fisiologia , Masculino , Gravidez , Globulina de Ligação a Hormônio Sexual/fisiologia , Comportamento Sexual Animal/fisiologia
13.
Horm Behav ; 121: 104728, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32119880

RESUMO

Fish present a wide variety of sex determination systems ranging from strict genetic control (genetic sex determination, GSD) to strict environmental control (environmental sex determination, ESD). Temperature is the most frequent environmental factor influencing sex determination. Nile tilapia (Oreochromis niloticus) is characterized by GSD with male heterogamety (XY/XX), which can be overridden by exposure to high masculinizing temperatures. Sex reversed Nile tilapia (XX males; neomales) have been described in the wild and seem undistinguishable from XY males, but little is known about their physiology. The consideration of climate change urges the need to understand the possible physiological and behavioral consequences of such a sex reversal. The present study compared XX females, XY males and XX neomales for testis maturation, circulating sex -steroid concentrations as well as the size and number of neurons expressing arginine-vasotocin [AVT] and gonadotropin releasing hormone [GnRH] which are involved in sociosexual pathways. The results revealed that temperature-induced sex reversal does not affect testis maturation nor circulating sex steroid concentrations. Neomales show dramatically fewer GnRH1-immunoreactive (-ir) neurons than males and females, despite the observed normal testis physiology. Neomales also present fewer AVT-ir neurons in the magnocellular preoptic area than females and bigger AVT-ir neurons in the parvocellular POA (pPOA) compared to both males and females. The absence of consequences of sex reversal on testis development and secretions despite the reduced numbers of GnRH1 neurons suggests the existence of compensatory mechanisms in the hypothalamic-pituitary-gonadal axis, while the larger pPOA AVT neurons might predict a more submissive behavior in neomales.


Assuntos
Encéfalo/metabolismo , Ciclídeos/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Diferenciação Sexual/fisiologia , Temperatura , Vasotocina/metabolismo , Animais , Ciclídeos/metabolismo , Feminino , Regulação da Expressão Gênica , Hormônios Esteroides Gonadais/sangue , Masculino , Neurônios/metabolismo , Área Pré-Óptica/metabolismo , Testículo/crescimento & desenvolvimento
14.
Biol Reprod ; 102(3): 758-769, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-31837141

RESUMO

Foxh1, a member of fox gene family, was first characterized as a transcriptional partner in the formation of the Smad protein complex. Recent studies have shown foxh1 is highly expressed in the cytoplasm of oocytes in both tilapia and mouse. However, its function in oogenesis remains unexplored. In the present study, foxh1-/- tilapia was created by CRISPR/Cas9. At 180 dah (days after hatching), the foxh1-/- XX fish showed oogenesis arrest and a significantly lower GSI. The transition of oocytes from phase II to phase III and follicle cells from one to two layers was blocked, resulting in infertility of the mutant. Transcriptomic analysis revealed that expression of genes involved in estrogen synthesis and oocyte growth were altered in the foxh1-/- ovaries. Loss of foxh1 resulted in significantly decreased Cyp19a1a and increased Cyp11b2 expression, consistent with significantly lower concentrations of serum estradiol-17ß (E2) and higher concentrations of 11-ketotestosterone (11-KT). Moreover, administration of E2 rescued the phenotypes of foxh1-/- XX fish, as indicated by the appearance of phase III and IV oocytes and absence of Cyp11b2 expression. Taken together, these results suggest that foxh1 functions in the oocytes to regulate oogenesis by promoting cyp19a1a expression, and therefore estrogen production. Disruption of foxh1 may block the estrogen synthesis and oocyte growth, leading to the arrest of oogenesis and thus infertility in tilapia.


Assuntos
Fatores de Transcrição Forkhead/genética , Infertilidade Feminina/genética , Mutação , Oogênese/genética , Animais , Animais Geneticamente Modificados , Ciclídeos , Estradiol/sangue , Feminino , Diferenciação Sexual/fisiologia , Testosterona/análogos & derivados , Testosterona/sangue
15.
Endocrinology ; 161(1)2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31758175

RESUMO

Gonadotropin-releasing hormone (Gnrh) plays important roles in reproduction by stimulating luteinizing hormone release, and subsequently ovulation and sperm release, ultimately controlling reproduction in many species. Here we report on a new role for this decapeptide. Surprisingly, Gnrh3-null zebrafish generated by CRISPR/Cas9 exhibited a male-biased sex ratio. After the dome stage, the number of primordial germ cells (PGCs) in gnrh3-/- fish was lower than that in wild-type, an effect that was partially rescued by gnrh3 overexpression. A terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) analysis revealed no detectable apoptosis of PGCs in gnrh3-/- embryos. Proliferating PGCs could be detected in wild-type embryos, while there was no detectable signal in gnrh3-/- embryos. Compared with wild type, the phosphorylation of AKT was not significantly different in gnrh3-/- embryos, but the phosphorylation of ERK1/2 decreased significantly. Treatment with a Gnrh analog (Alarelin) induced ERK1/2 phosphorylation and increased PGC numbers in both wild-type and gnrh3-/- embryos, and this was blocked by the MEK inhibitor PD0325901. The relative expression of sox9a, amh, and cyp11b were significantly upregulated, while cyp19a1a was significantly downregulated at 18 days post-fertilization in gnrh3-/- zebrafish. Taken together, these results indicate that Gnrh3 plays an important role in early sex differentiation by regulating the proliferation of PGCs through a MAPK-dependent path.


Assuntos
Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , Diferenciação Sexual/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Benzamidas/farmacologia , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Embrião não Mamífero/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácido Pirrolidonocarboxílico/metabolismo , Razão de Masculinidade , Proteínas de Peixe-Zebra/genética
16.
Endocrinology ; 161(1)2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-31742329

RESUMO

Many neural sex differences are differences in the number of neurons of a particular phenotype. For example, male rodents have more calbindin-expressing neurons in the medial preoptic area (mPOA) and bed nucleus of the stria terminalis (BNST), and females have more neurons expressing estrogen receptor alpha (ERα) and kisspeptin in the ventromedial nucleus of the hypothalamus (VMH) and the anteroventral periventricular nucleus (AVPV), respectively. These sex differences depend on neonatal exposure to testosterone, but the underlying molecular mechanisms are unknown. DNA methylation is important for cell phenotype differentiation throughout the developing organism. We hypothesized that testosterone causes sex differences in neurochemical phenotype via changes in DNA methylation, and tested this by inhibiting DNA methylation neonatally in male and female mice, and in females given a masculinizing dose of testosterone. Neonatal testosterone treatment masculinized calbindin, ERα and kisspeptin cell number of females at weaning. Inhibiting DNA methylation with zebularine increased calbindin cell number only in control females, thus eliminating sex differences in calbindin in the mPOA and BNST. Zebularine also reduced the sex difference in ERα cell number in the VMH, in this case by increasing ERα neuron number in males and testosterone-treated females. In contrast, the neonatal inhibition of DNA methylation had no effect on kisspeptin cell number. We conclude that testosterone normally increases the number of calbindin cells and reduces ERα cells in males through orchestrated changes in DNA methylation, contributing to, or causing, the sex differences in both cell types.


Assuntos
Encéfalo/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Diferenciação Sexual/efeitos dos fármacos , Testosterona/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/metabolismo , Química Encefálica/efeitos dos fármacos , Calbindinas/metabolismo , Citidina/administração & dosagem , Citidina/análogos & derivados , Citidina/farmacologia , Receptor alfa de Estrogênio/metabolismo , Feminino , Kisspeptinas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Diferenciação Sexual/fisiologia , Fatores Sexuais , Testosterona/administração & dosagem
17.
Sci Rep ; 9(1): 18446, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31804598

RESUMO

Insulin family peptides are known to be key regulators of growth and metabolism in insects and vertebrates. Insects have two types of insulin family peptides: insulin-like peptides and insulin-like growth factor (IGF)-like peptides (IGFLPs). We recently demonstrated that an IGFLP in the silkmoth, Bombyx mori (BIGFLP) promotes the growth of the genital imaginal disc ex vivo. However, the role of BIGFLP in the regulation of insect growth remains unclear because no in vivo study has been performed. Therefore, we analysed the functions of BIGFLP in vivo by constructing BIGFLP knock-out (KO) B. mori using the clustered regularly interspaced palindromic repeats (CRISPR) and CRISPR-associated protein 9 (CRISPR-Cas9) system. The KO moths exhibited decreased body weights and size of the appendages compared wild-type (wt) moths. Interestingly, KO females also had drastically lower ovary weights and number of eggs than wt females. However, mutant ovaries that were transplanted into wt host pupae reached a similar weight to wt ovaries that were transplanted into the wt hosts, suggesting that IGFLP in the haemolymph promotes ovarian development. These findings show that BIGFLP regulates the growth and development of adult organs, particularly the ovaries, in B. mori.


Assuntos
Bombyx/fisiologia , Proteínas de Insetos/metabolismo , Ovário/crescimento & desenvolvimento , Peptídeos/metabolismo , Somatomedinas/metabolismo , Animais , Feminino , Técnicas de Silenciamento de Genes , Hemolinfa/metabolismo , Discos Imaginais/crescimento & desenvolvimento , Proteínas de Insetos/genética , Diferenciação Sexual/fisiologia , Somatomedinas/genética
18.
Gen Comp Endocrinol ; 277: 56-65, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30878349

RESUMO

Unlike its paralog Foxl2, which is well known for its role in ovarian development in vertebrates, the function of Foxl3 is still unclear. Foxl3 is an ancient duplicated copy of Foxl2. It is present as a single copy in ray-finned fish. But, due to repeated losses, it is absent in most tetrapods. Our transcriptomic data, however, show that two Foxl3s (Foxl3a and its paralog Foxl3b) are present in Japanese eel. Foxl3a is predominantly expressed in the pituitary, and Foxl3b is predominantly expressed in the gills. Both Foxl3s show a sex-dimorphic expression, being higher expression in testes than in ovaries. Moreover, Foxl3a and Foxl3b were exclusively expressed during gonadal differentiation in control eels (100% male). Conversely, Foxl3a and Foxl3b significantly decreased after gonadal differentiation in E2-treated eels (100% female). Furthermore, in accordance the difference in adhesive ability between somatic cells and germline cells in testes, Foxl3s showed a high expression in suspension cells (putative germline cells) and low expression in adhesive cells (putative somatic cells). In situ hybridization further showed that Foxl3a and Foxl3b were expressed in the testicular germline cells. In addition, Foxl3s expression was not changed by sex steroids in in vitro testes culture. Taken together, our results suggest that the teleost-specific Foxl3 paralog was repeatedly lost in most fish after the third round of whole genome duplication. The two germline-expressed Foxl3s had higher expression levels in males than in females during gonadal differentiation in Japanese eel. These results demonstrated that Foxl3s might play an important role in germline sexual fate determination from ancient fish to modern fish.


Assuntos
Anguilla/genética , Anguilla/fisiologia , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Gônadas/fisiologia , Diferenciação Sexual/fisiologia , Sequência de Aminoácidos , Animais , Tamanho Corporal/efeitos dos fármacos , Estradiol/farmacologia , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células Germinativas/efeitos dos fármacos , Gônadas/efeitos dos fármacos , Masculino , Filogenia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Diferenciação Sexual/efeitos dos fármacos , Diferenciação Sexual/genética , Esteroides/farmacologia , Testículo/citologia , Testículo/efeitos dos fármacos , Testículo/metabolismo
19.
Gen Comp Endocrinol ; 275: 82-93, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30738863

RESUMO

This study evaluated the impact of continuous light (LL) within the photolabile period on advanced puberty in juvenile male European sea bass. The exposure to an LL regime for 1 month, from August 15 to September 15 (LLa/s), was compared to a constant simulated natural photoperiod (NP) and constant continuous light conditions year-round (LLy). Somatic growth, hormone plasma levels, rates of testicular maturation and spermiation, as well as the mRNA levels of some reproductive genes were analyzed. Our results demonstrated that both LLa/s and LLy treatments, which include LL exposure during the photolabile period, were highly effective in inhibiting the gametogenesis process that affects testicular development, and clearly reduced the early sexual maturation of males. Exposure to an LL photoperiod affected body weight and length of juvenile fish during early gametogenesis and throughout the first year of life. Interestingly, LL induced bi-weekly changes in some reproductive factors affecting Gnrh1 and Gnrh2 content in the brain, and also reduced pituitary fshß expression and plasmatic levels of 11-KT, E2, Fsh throughout early gametogenesis. We suggest that low levels of E2 in early September in the LL groups, which would be concomitant with the reduced number of spermatogonial mitoses in these groups, might indicate a putative role for estrogens in spermatogonial proliferation during the early gonadal development of this species. Furthermore, a significant decrease in amh expression was observed, coinciding with low plasma levels of 11-KT under LL regimes, which is consistent with the idea that this growth factor may be crucial for the progress of spermatogenesis in male sea bass.


Assuntos
Bass/crescimento & desenvolvimento , Iluminação , Fotoperíodo , Reprodução/fisiologia , Maturidade Sexual/fisiologia , Animais , Bass/sangue , Subunidade beta do Hormônio Folículoestimulante/sangue , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/sangue , Masculino , Precursores de Proteínas/sangue , Diferenciação Sexual/fisiologia , Espermatogênese/fisiologia , Fatores de Tempo
20.
Insect Biochem Mol Biol ; 104: 30-38, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30529581

RESUMO

The Masculinizer (Masc) gene encodes a novel lepidopteran-specific protein that controls both masculinization and dosage compensation in the silkworm Bombyx mori. The Masc protein possesses two CCCH-type zinc finger domains (ZFs), a nuclear localization signal, and an 11-amino-acid region that is highly conserved among lepidopteran insects. Using a cell-based assay system, we revealed that two cysteine residues localized in the conserved region, but not ZFs, are required for masculinization. In addition, nuclear localization of the Masc protein is not associated with masculinizing activity. Because dosage compensation is considered to occur in the nucleus, we inferred that the two ZFs play a role in the establishment of dosage compensation. To investigate this hypothesis at the organism level, we utilized the CRISPR/Cas9 system and established three B. mori strains whose Masc is partially deleted at different regions. The strain lacking the 210 C-terminal amino acids of the Masc protein showed male-specific embryonic lethality due to its low abundance and/or instability. The male embryos of this strain expressed the female-type splice variants of B. mori doublesex and did not express the male-type mRNA of B. mori IGF-II mRNA-binding protein. Furthermore, mRNA levels of Z-linked genes were abnormally enhanced only in male embryos. In contrast, the strain lacking both ZFs grew normally and did not show any defective phenotypes including sexual differentiation and the expression of Z-linked genes, demonstrating that the two CCCH-type ZFs, which are conserved in lepidopteran Masc homologs, are dispensable for masculinization and dosage compensation.


Assuntos
Mecanismo Genético de Compensação de Dose , Processos de Determinação Sexual/fisiologia , Diferenciação Sexual/fisiologia , Animais , Bombyx/genética , Bombyx/metabolismo , Feminino , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Masculino , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA