Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Br J Cancer ; 130(9): 1453-1462, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38429437

RESUMEN

BACKGROUND: FOXL2 is a transcription factor expressed in ovarian granulosa cells. A somatic variant of FOXL2 (c.402 C > G, p.Cys134Trp) is the hallmark of adult-type granulosa cell tumours. METHODS: We generated KGN cell clones either heterozygous for this variant (MUT) or homozygous for the wild-type (WT) allele by CRISPR/Cas9 editing. They underwent RNA-Seq and bioinformatics analyses to uncover pathways impacted by deregulated genes. Cell morphology and migration were studied. RESULTS: The differentially expressed genes (DEGs) between WT/MUT and WT/WT KGN cells (DEGs-WT/MUT), pointed to several dysregulated pathways, like TGF-beta pathway, cell adhesion and migration. Consistently, WT/MUT cells were rounder than WT/WT cells and displayed a different distribution of stress fibres and paxillin staining. A comparison of the DEGs-WT/MUT with those found when FOXL2 was knocked down (KD) in WT/WT KGN cells showed that most DEGs-WT/MUT cells were not so in the KD experiment, supporting a gain-of-function (GOF) scenario. MUT-FOXL2 also displayed a stronger interaction with SMAD3. CONCLUSIONS: Our work, aiming at better understanding the GOF scenario, shows that the dysregulated genes and pathways are consistent with this idea. Besides, we propose that GOF might result from an enhanced interaction with SMAD3 that could underlie an ectopic capacity of mutated FOXL2 to bind SMAD4.


Asunto(s)
Proteína Forkhead Box L2 , Tumor de Células de la Granulosa , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Humanos , Femenino , Tumor de Células de la Granulosa/genética , Tumor de Células de la Granulosa/patología , Línea Celular Tumoral , Movimiento Celular/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Sistemas CRISPR-Cas , Regulación Neoplásica de la Expresión Génica
2.
Biol Reprod ; 110(5): 985-999, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38376238

RESUMEN

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


Asunto(s)
Factor de Transcripción SOX9 , Procesos de Determinación del Sexo , Testículo , Trombospondinas , Regulación hacia Arriba , Animales , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Masculino , Femenino , Ratones , Trombospondinas/genética , Trombospondinas/metabolismo , Procesos de Determinación del Sexo/genética , Procesos de Determinación del Sexo/fisiología , Testículo/metabolismo , Gónadas/metabolismo , Ovario/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Diferenciación Sexual/genética , Ratones Endogámicos C57BL
3.
Cell Prolif ; 57(5): e13589, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38192172

RESUMEN

Human granulosa cells in different stages are essential for maintaining normal ovarian function, and granulosa cell defect is the main cause of ovarian dysfunction. To address this problem, it is necessary to induce functional granulosa cells at different stages in vitro. In this study, we established a reprogramming method to induce early- and late-stage granulosa cells with different steroidogenic abilities. We used an AMH-fluorescence-reporter system to screen candidate factors for cellular reprogramming and generated human induced granulosa-like cells (hiGC) by overexpressing FOXL2 and NR5A1. AMH-EGFP+ hiGC resembled human cumulus cells in transcriptome profiling and secreted high levels of oestrogen and progesterone, similar to late-stage granulosa cells at antral or preovulatory stage. Moreover, we identified CD55 as a cell surface marker that can be used to isolate early-stage granulosa cells. CD55+ AMH-EGFP- hiGC secreted high levels of oestrogen but low levels of progesterone, and their transcriptome profiles were more similar to early-stage granulosa cells. More importantly, CD55+ hiGC transplantation alleviated polycystic ovary syndrome (PCOS) in a mouse model. Therefore, hiGC provides a cellular model to study the developmental program of human granulosa cells and has potential to treat PCOS.


Asunto(s)
Fibroblastos , Proteína Forkhead Box L2 , Células de la Granulosa , Factor Esteroidogénico 1 , Femenino , Humanos , Proteína Forkhead Box L2/metabolismo , Proteína Forkhead Box L2/genética , Células de la Granulosa/metabolismo , Células de la Granulosa/citología , Animales , Ratones , Fibroblastos/metabolismo , Fibroblastos/citología , Factor Esteroidogénico 1/metabolismo , Factor Esteroidogénico 1/genética , Progesterona/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Reprogramación Celular , Células Cultivadas
4.
Life Sci Alliance ; 6(10)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37532283

RESUMEN

Estradiol and progesterone are the primary sex steroids produced by the ovary. Upon luteinizing hormone surge, estradiol-producing granulosa cells convert into progesterone-producing cells and eventually become large luteal cells of the corpus luteum. Signaling pathways and transcription factors involved in the cessation of estradiol and simultaneous stimulation of progesterone production in granulosa cells are not clearly understood. Here, we decipher that phosphorylated ERK1/2 regulates granulosa cell steroidogenesis by inhibiting estradiol and inducing progesterone production. Down-regulation of transcription factor FOXL2 and up-regulation of SOX9 by ERK underpin its differential steroidogenic function. Interestingly, the incidence of SOX9 is largely uncovered in ovarian cells and is found to regulate FOXL2 along with CYP19A1 and STAR genes, encoding rate-limiting enzymes of steroidogenesis, in cultured granulosa cells. We propose that the novel ERK1/2-SOX9/FOXL2 axis in granulosa cells is a critical regulator of ovarian steroidogenesis and may be considered when addressing pathophysiologies associated with inappropriate steroid production and infertility in humans and animals.


Asunto(s)
Ovario , Progesterona , Femenino , Humanos , Animales , Ovario/metabolismo , Progesterona/metabolismo , Sistema de Señalización de MAP Quinasas , Cuerpo Lúteo/metabolismo , Estradiol , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-36681266

RESUMEN

As the second largest phylum in the zoological kingdom next to arthropods, the mechanism of gonadal differentiation in mollusca is quite complex. Currently, although much has been carried out on gonadal differentiation in the Pacific oyster, there is still unknown information that needs to be further explored. Here, analysis of the Foxl2 and Dmrt1l expression in samples at different development periods of male and female gonads as well as in annual gonad samples revealed that Log10 (Foxl2/Dmrt1l) values were an effective method for sex identification in oysters. In differentiated gonadal tissue, Log10 (Foxl2/Dmrt1l) values greater than 2 were females and less than 1 for males. Subsequent sequential sampling of the same individuals verified that Log10 (Foxl2/Dmrt1l) values greater than 2 for resting gonads would develop as females and less than 1 would develop as males in the future. Relative expression analysis of Foxl2 and Dmrt1l in the annual samples revealed a negative correlation between Log10 (Foxl2) and Log10 (Dmrt1l). Double fluorescence reporter validation results showed that DMRT1L protein was able to bind the Foxl2 promoter and repress its activity with a weak dosage effect. Antagonism between Dmrt1l and Foxl2 is therefore not restricted to vertebrates, and the competing regulatory networks are of great significance in the maintenance of gonadal sex in oysters after sexual differentiation. This study provides novel ideas and insights into the study of early gonadal differentiation in the adult oyster.


Asunto(s)
Gónadas , Ovario , Animales , Femenino , Masculino , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Gónadas/metabolismo , Ovario/metabolismo , Regiones Promotoras Genéticas , Diferenciación Sexual , Ostreidae/genética , Ostreidae/metabolismo , Océano Pacífico
6.
Nucleic Acids Res ; 50(15): 8929-8946, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35920317

RESUMEN

Although both the p53 and forkhead box (FOX) family proteins are key transcription factors associated with cancer progression, their direct relationship is unknown. Here, we found that FOX family proteins bind to the non-canonical homotypic cluster of the p53 promoter region (TP53). Analysis of crystal structures of FOX proteins (FOXL2 and FOXA1) bound to the p53 homotypic cluster indicated that they interact with a 2:1 stoichiometry accommodated by FOX-induced DNA allostery. In particular, FOX proteins exhibited distinct dimerization patterns in recognition of the same p53-DNA; dimer formation of FOXA1 involved protein-protein interaction, but FOXL2 did not. Biochemical and biological functional analyses confirmed the cooperative binding of FOX proteins to the TP53 promoter for the transcriptional activation of TP53. In addition, up-regulation of TP53 was necessary for FOX proteins to exhibit anti-proliferative activity in cancer cells. These analyses reveal the presence of a discrete characteristic within FOX family proteins in which FOX proteins regulate the transcription activity of the p53 tumor suppressor via cooperative binding to the TP53 promoter in alternative dimer configurations.


Asunto(s)
Proteína Forkhead Box L2/metabolismo , Factores de Transcripción Forkhead , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Proteína p53 Supresora de Tumor/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Regiones Promotoras Genéticas , Proteína p53 Supresora de Tumor/metabolismo
7.
Front Endocrinol (Lausanne) ; 13: 863360, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35620392

RESUMEN

The giant wrasse Cheilinus undulatus is a protogynous socially hermaphroditic fish. However, the physiological basis of its sex reversal remains largely unknown. cyp19 is a key gender-related gene encoding P450 aromatase, which converts androgens to estrogens. cyp19 transcription regulation is currently unknown in socially sexually reversible fish. We identified NR5A1 by encoding SF-1, and FOXL2 from giant wrasse cDNA and cyp19a1a and cyp19a1b promoter regions were cloned from genomic DNA to determine the function of both genes in cyp19a1 regulation. Structural analysis showed that SF-1 contained a conserved DNA-binding domain (DBD) and a C-terminal ligand-binding domain (LBD). FOXL2 was comprised of an evolutionarily conserved Forkhead domain. In vitro transfection assays showed that SF-1 could upregulate cyp19a1 promoter activities, but FOXL2 could only enhance cyp19a1b promoter transcriptional activity in the HEK293T cell line. Furthermore, HEK293T and COS-7 cell lines showed that co-transfecting the two transcription factors significantly increased cyp19a1 promoter activity. The -120 to -112 bp (5'-CAAGGGCAC-3') and -890 to -872 bp (5'-AGAGGAGAACAAGGGGAG-3') regions of the cyp19a1a promoter were the core regulatory elements for SF-1 and FOXL2, respectively, to regulate cyp19a1b promoter transcriptional activity. Collectively, these results suggest that both FOXL2 and SF-1 are involved in giant wrasse sex reversal.


Asunto(s)
Aromatasa , Proteínas de Peces , Proteína Forkhead Box L2 , Perciformes , Animales , Aromatasa/genética , Aromatasa/metabolismo , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Células HEK293 , Humanos , Perciformes/genética , Regiones Promotoras Genéticas
8.
Artículo en Inglés | MEDLINE | ID: mdl-34147671

RESUMEN

microRNAs (miRNAs) are important components of non-coding RNAs that participate in diverse life activities by regulating gene expression at the post transcriptional level through base complementary pairing with 3'UTRs of target mRNAs. miR-133b is a member of the miR-133 family, which play important roles in muscle differentiation and tumorigenesis. Recently, miR-133b was reported to affect estrogen synthesis by targeting foxl2 in mouse, while its role in fish reproduction remains to be elucidated. In the present study, we isolated the complete sequence of miR-133b, which was highly expressed in tilapia ovary at 30 and 90 dah (days after hatching) and subsequently decreased at 120 to 150 dah by qPCR. Interestingly, only a few oogonia were remained in the antagomir-133b treated tilapia ovary, while phase I and II oocytes were observed in the ovaries of the control group. Unexpectedly, the expression of foxl2 and cyp19a1a, as well as estradiol levels in serum were increased in the treated group. Furthermore, tagln2, an important factor for oogenesis, was predicted as the target gene of miR-133b, which was confirmed by dual luciferase reporter vector experiments. miR-133b and tagln2 were co-expressed in tilapia ovaries. Taken together, miR-133b may be involved in the early oogenesis of tilapia by regulating tagln2 expression. This study enriches the understanding of miR-133b function during oogenesis and lays a foundation for further study of the regulatory network during oogenesis.


Asunto(s)
Proteínas de Peces/metabolismo , Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , Proteínas de Microfilamentos/metabolismo , Oogénesis , Ovario/metabolismo , Tilapia/metabolismo , Animales , Femenino , Proteínas de Peces/genética , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Perfilación de la Expresión Génica , Proteínas de Microfilamentos/genética , Ovario/citología , Tilapia/genética , Tilapia/crecimiento & desarrollo
9.
Artículo en Inglés | MEDLINE | ID: mdl-34171478

RESUMEN

Transcription factor Foxl2 is an evolutionarily conserved gene playing pivotal roles in regulation of early ovarian differentiation and maintenance in animals. However, the Foxl2 gene has not been thoroughly studied in hermaphroditic scallops. In this study, we cloned and characterized a Foxl2 (designated as AiFoxl2) from the bay scallop Argopecten irradians irradians. The open reading frame of AiFoxl2 was 1122 bp encoding 373 amino acids residues and contained a conserved forkhead box domain. Quantitative real-time PCR showed that AiFoxl2 was mainly expressed in the ovary. Moreover, the highest expression of AiFoxl2 in the ovary was detected at proliferative stage and growing stage, while the lowest level was found at resting stage. During the embryonic and larval development, expression of AiFoxl2 was found first in fertilized eggs, increased significantly at the blastula stage, and reached peak value at the D-larvae stage. When AiFoxl2 was knocked down, testis development-related genes (Dmrt1, Sox7 and Sox9) were up-regulated significantly while the ovary development-related genes (Vg, HSD14, and GATA-1) were down-regulated manifestly. These findings suggested that AiFoxl2 was a female-related gene in A. i. irradians and may be involved in regulation of ovarian development and differentiation.


Asunto(s)
Proteínas de Peces/metabolismo , Proteína Forkhead Box L2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Pectinidae/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de Peces/genética , Proteína Forkhead Box L2/genética , Perfilación de la Expresión Génica , Pectinidae/genética , Pectinidae/crecimiento & desarrollo , Filogenia , Homología de Secuencia , Factores Sexuales
10.
Gene ; 790: 145692, 2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-33961972

RESUMEN

Several evolutionarily conserved classes of transcriptional regulators were involved in diverse sex determination and differentiation pathways across taxa, whereas their roles in most mollusks is still limited. The Pacific oyster Crassostrea gigas, a dioecious bivalve with sex reversal, could be an ideal model for this issue because of its complex sexuality and potential disruption of sex differentiation in triploid individuals. Here, two mRNA splicing isoforms of a DM domain gene CgDsx and two isoforms of a novel sex-related CgBHMG1 (ortholog of BHMG1 in mammals) were identified in C. gigas. Real time PCR showed that two isoforms of CgDsx and one isoform of CgBHMG1 displayed male-specific expression in diploid oysters, opposite with the female-specific CgFoxl2 (a potential factor of female gonadic differentiation). Interestingly, the four sex-specific transcripts in diploid oyster were expressed in triploid oysters with opposite sex, triploid hermaphrodites and individuals at stage I that sex could not be determined. Subsequent in situ hybridization analysis on gonads of diploid oysters revealed predominant expression of CgDsx in spermatogonia of testes, CgBHMG1 in spermatocytes of testes and follicle cells of ovaries, and CgFoxl2 in follicle cells of ovaries and some male germ cells in testes. And aberrant co-expression of the three genes in triploid oysters was localized in gonadal tubules of gonads at stage I, ovarian follicle cells and undetermined gonial cells in nontypical hermaphroditic gonads with rare female materials. From the above, temporal and spatial expression of sex-related genes in diploid and triploid gonads indicated that CgDsx and CgFoxl2 might mainly function in C. gigas sex differentiation, and CgBHMG1 appeared as a factor involved in meiosis. This work will help to illuminate the gene network of sex differentiation in bivalves and provides new sight on this issue from comparison between diploid and triploid individuals.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Diploidia , Proteína Forkhead Box L2/metabolismo , Regulación de la Expresión Génica , Gónadas/metabolismo , Diferenciación Sexual , Triploidía , Animales , Crassostrea , Proteínas de Unión al ADN/genética , Proteína Forkhead Box L2/genética , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Gónadas/crecimiento & desarrollo
11.
FASEB J ; 35(4): e21355, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33749886

RESUMEN

FOXL2 and ESR2 are key transcriptional regulators in ovarian granulosa cells. To explore their transcriptional roles and their interplay, we have depleted Foxl2 and Esr2 in mouse primary granulosa cells to assess their ability to bind their targets and/or to modulate gene expression and cellular functions. We show that FOXL2 is involved in a large number of regulatory actions essential for the maintenance of granulosa cell fate. A parallel ChIP-seq analysis showed that FOXL2 mainly binds to sites located in intergenic regions quite far from its targets. A bioinformatic analysis demonstrated that FOXL2-activated genes were enriched in peaks associated with the H3K27ac mark, whereas FOXL2-repressed genes were not, suggesting that FOXL2 can activate transcription through binding to enhancer sites. We also identified about 500 deregulated genes upon Esr2 silencing, of which one third are also targets of FOXL2. We provide evidence showing that both factors modulate, through a coherent feed-forward loop, a number of common targets. Many of the FOXL2/ESR2 targets are involved in cell motility and, consistently, granulosa cells depleted for either Foxl2 or Esr2 exhibit decreased migration, invasion and adhesion. This effect is paralleled by the depletion of their target Phactr1, involved in actin cytoskeleton dynamics. Our analysis expands the number of direct and indirect transcriptional targets of both FOXL2 and ESR2, which deserve investigation in the context of adult-type granulosa cell tumors whose molecular diagnostic hallmark is the presence of the C134W FOXL2 pathogenic variant.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Proteína Forkhead Box L2/metabolismo , Células de la Granulosa/fisiología , Animales , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Receptor beta de Estrógeno/genética , Femenino , Proteína Forkhead Box L2/genética , Edición Génica , Ratones
12.
Development ; 148(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33741713

RESUMEN

Germline sexual fate has long been believed to be determined by the somatic environment, but this idea is challenged by recent studies of foxl3 mutants in medaka. Here, we demonstrate that the sexual fate of tilapia germline is determined by the antagonistic interaction of dmrt1 and foxl3, which are transcriptionally repressed in male and female germ cells, respectively. Loss of dmrt1 rescued the germ cell sex reversal in foxl3Δ7/Δ7 XX fish, and loss of foxl3 partially rescued germ cell sex reversal but not somatic cell fate in dmrt1Δ5/Δ5 XY fish. Interestingly, germ cells lost sexual plasticity in dmrt1Δ5/Δ5 XY and foxl3Δ7/Δ7 XX single mutants, as aromatase inhibitor (AI) and estrogen treatment failed to rescue the respective phenotypes. However, recovery of germ cell sexual plasticity was observed in dmrt1/foxl3 double mutants. Importantly, mutation of somatic cell-specific foxl2 resulted in testicular development in foxl3Δ7/Δ7 or dmrt1Δ5/Δ5 mutants. Our findings demonstrate that sexual plasticity of germ cells relies on the presence of both dmrt1 and foxl3. The existence of dmrt1 and foxl3 allows environmental factors to influence the sex fate decision in vertebrates.


Asunto(s)
Cíclidos , Proteínas de Peces , Proteína Forkhead Box L2 , Procesos de Determinación del Sexo , Diferenciación Sexual , Factores de Transcripción , Animales , Cíclidos/embriología , Cíclidos/genética , Femenino , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Masculino , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Mol Biol Evol ; 38(5): 1995-2013, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33432361

RESUMEN

Evolutionary fates of duplicated genes have been widely investigated in many polyploid plants and animals, but research is scarce in recurrent polyploids. In this study, we focused on foxl2, a central player in ovary, and elaborated the functional divergence in gibel carp (Carassius gibelio), a recurrent auto-allo-hexaploid fish. First, we identified three divergent foxl2 homeologs (Cgfoxl2a-B, Cgfoxl2b-A, and Cgfoxl2b-B), each of them possessing three highly conserved alleles and revealed their biased retention/loss. Then, their abundant sexual dimorphism and biased expression were uncovered in hypothalamic-pituitary-gonadal axis. Significantly, granulosa cells and three subpopulations of thecal cells were distinguished by cellular localization of CgFoxl2a and CgFoxl2b, and the functional roles and the involved process were traced in folliculogenesis. Finally, we successfully edited multiple foxl2 homeologs and/or alleles by using CRISPR/Cas9. Cgfoxl2a-B deficiency led to ovary development arrest or complete sex reversal, whereas complete disruption of Cgfoxl2b-A and Cgfoxl2b-B resulted in the depletion of germ cells. Taken together, the detailed cellular localization and functional differences indicate that Cgfoxl2a and Cgfoxl2b have subfunctionalized and cooperated to regulate folliculogenesis and gonad differentiation, and Cgfoxl2b has evolved a new function in oogenesis. Therefore, the current study provides a typical case of homeolog/allele diversification, retention/loss, biased expression, and sub-/neofunctionalization in the evolution of duplicated genes driven by polyploidy and subsequent diploidization from the recurrent polyploid fish.


Asunto(s)
Evolución Molecular , Proteína Forkhead Box L2/genética , Duplicación de Gen , Carpa Dorada/genética , Poliploidía , Animales , Femenino , Proteína Forkhead Box L2/metabolismo , Carpa Dorada/crecimiento & desarrollo , Carpa Dorada/metabolismo , Masculino , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Ovario/crecimiento & desarrollo , Ovario/metabolismo
14.
EMBO J ; 39(24): e104719, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33215742

RESUMEN

Recent evidence suggests that animal microRNAs (miRNAs) can target coding sequences (CDSs); however, the pathophysiological importance of such targeting remains unknown. Here, we show that a somatic heterozygous missense mutation (c.402C>G; p.C134W) in FOXL2, a feature shared by virtually all adult-type granulosa cell tumors (AGCTs), introduces a target site for miR-1236, which causes haploinsufficiency of the tumor-suppressor FOXL2. This miR-1236-mediated selective degradation of the variant FOXL2 mRNA is preferentially conducted by a distinct miRNA-loaded RNA-induced silencing complex (miRISC) directed by the Argonaute3 (AGO3) and DHX9 proteins. In both patients and a mouse model of AGCT, abundance of the inversely regulated variant FOXL2 with miR-1236 levels is highly correlated with malignant features of AGCT. Our study provides a molecular basis for understanding the conserved FOXL2 CDS mutation-mediated etiology of AGCT, revealing the existence of a previously unidentified mechanism of miRNA-targeting disease-associated mutations in the CDS by forming a non-canonical miRISC.


Asunto(s)
Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Tumor de Células de la Granulosa/genética , MicroARNs/metabolismo , Mutación , Sistemas de Lectura Abierta , Desequilibrio Alélico , Animales , Apoptosis , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Muerte Celular/fisiología , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Tumor de Células de la Granulosa/patología , Células HEK293 , Humanos , Ratones , Ratones Noqueados , MicroARNs/genética , Mutación Missense , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , ARN Mensajero/metabolismo , Transcriptoma
15.
BMC Mol Cell Biol ; 21(1): 66, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32933467

RESUMEN

BACKGROUND: The increasing incidence of reproductive disorders in humans has been attributed to in utero exposure to estrogenic endocrine disruptors. In particular, exposure of the developing testis to exogenous estrogen can negatively impact male reproductive health. To determine how estrogens impact human gonad function, we treated the human testis-derived cell line NT2/D1 with estrogen and examined its impact on SOX9 and the expression of key markers of granulosa (ovarian) and Sertoli (testicular) cell development. RESULTS: Estrogen successfully activated its cognate receptor (estrogen receptor alpha; ESR1) in NT2/D1 cells. We observed a significant increase in cytoplasmic SOX9 following estrogen treatment. After 48 h of estrogen exposure, mRNA levels of the key Sertoli cell genes SOX9, SRY, AMH, FGF9 and PTGDS were significantly reduced. This was followed by a significant increase in mRNA levels for the key granulosa cell genes FOXL2 and WNT4 after 96 h of estrogen exposure. CONCLUSIONS: These results are consistent with estrogen's effects on marsupial gonads and show that estrogen has a highly conserved impact on gonadal cell fate decisions that has existed in mammals for over 160 million years. This effect of estrogen presents as a potential mechanism contributing to the significant decrease in male fertility and reproductive health reported over recent decades. Given our widespread exposure to estrogenic endocrine disruptors, their effects on SOX9 and Sertoli cell determination could have considerable impact on the adult testis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Estrógenos/metabolismo , Factor de Transcripción SOX9/metabolismo , Testículo/metabolismo , Línea Celular Tumoral , Disruptores Endocrinos/metabolismo , Femenino , Proteína Forkhead Box L2/metabolismo , Gónadas/metabolismo , Células de la Granulosa/metabolismo , Humanos , Masculino , Ovario/metabolismo , ARN Mensajero/metabolismo , Células de Sertoli/metabolismo , Proteína Wnt4/metabolismo
16.
Biol Reprod ; 103(6): 1289-1299, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32940693

RESUMEN

In animals, spermatogonial transplantation in sterile adult males is widely developed; however, despite its utility, ovarian germ cell transplantation is not well developed. We previously showed that the interspecific hybrid offspring of sciaenid was a suitable model for germ cell transplantation studies as they have germ cell-less gonads. However, all these gonads have testis-like characteristics. Here, we tested whether triploidization in hybrid embryos could result in germ cell-less ovary development. Gonadal structure dimorphism and sex-specific gene expression patterns were examined in 6-month-old triploid hybrids (3nHybs). Thirty-one percent of 3nHybs had germ cell-less gonads with an ovarian cavity. cyp19a1a and foxl2, ovarian differentiation-related genes, were expressed in these gonads, whereas dmrt1 and vasa were not expressed, suggesting ovary-like germ cell-less gonad development. Some (26%) 3nHybs had testis-like germ cell-less gonads. Ovarian germ cells collected from homozygous green fluorescent protein (GFP) transgenic blue drum (BD) (Nibea mitsukurii) were transplanted into 6-month-old 3nHybs gonads via the urogenital papilla or oviduct. After 9 months, the recipients were crossed with wild type BD. Among the six 3nHyb recipients that survived, one female and one male produced fertile eggs and motile sperm carrying gfp-specific DNA sequences. Progeny tests revealed that all F1 offspring possessed gfp-specific DNA sequences, suggesting that these recipients produced only donor-derived eggs or sperm. Histological observation confirmed donor-derived gametogenesis in the 3nHyb recipients' gonads. Overall, triploidization reduces male-biased sex differentiation in germ cell-less gonads. We report, for the first time, donor-derived egg production in an animal via direct ovarian germ cell transplantation into a germ cell-less ovary.


Asunto(s)
Peces/genética , Peces/fisiología , Células Germinativas/trasplante , Gónadas/citología , Triploidía , Animales , Animales Modificados Genéticamente , Aromatasa/genética , Aromatasa/metabolismo , Frío , ARN Helicasas DEAD-box , Embrión no Mamífero , Femenino , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Regulación de la Expresión Génica , Masculino , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Biol Reprod ; 103(5): 951-965, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32948877

RESUMEN

The transcription factor forkhead box L2 (FOXL2) regulates sex differentiation and reproductive function. Elevated levels of this transcription factor have been observed in the diseases of the uterus, such as endometriosis. However, the impact of elevated FOXL2 expression on uterine physiology remains unknown. In order to determine the consequences of altered FOXL2 in the female reproductive axis, we generated mice with over-expression of FOXL2 (FOXL2OE) by crossing Foxl2LsL/+ with the Progesterone receptor Pgrcre model. FOXL2OE uterus showed severe morphological abnormality including abnormal epithelial stratification, blunted adenogenesis, increased endometrial fibrosis, and disrupted myometrial morphology. In contrast, increasing FOXL2 levels specifically in uterine epithelium by crossing the Foxl2LsL/+ with the lactoferrin Ltficre mice resulted in the eFOXL2OE mice with uterine epithelial stratification but without defects in endometrial fibrosis and adenogenesis, demonstrating a role of the endometrial stroma in the uterine abnormalities of the FOXL2OE mice. Transcriptomic analysis of 12 weeks old Pgrcre and FOXL2OE uterus at diestrus stage showed multiple signaling pathways related with cellular matrix, wnt/ß-catenin, and altered cell cycle. Furthermore, we found FOXL2OE mice were sterile. The infertility was caused in part by a disruption of the hypophyseal ovarian axis resulting in an anovulatory phenotype. The FOXL2OE mice failed to show decidual responses during artificial decidualization in ovariectomized mice demonstrating the uterine contribution to the infertility phenotype. These data support that aberrantly increased FOXL2 expressions in the female reproductive tract can disrupt ovarian and uterine functions.


Asunto(s)
Proteína Forkhead Box L2/metabolismo , Anomalías Urogenitales/metabolismo , Útero/anomalías , Útero/metabolismo , Animales , Endometrio/metabolismo , Femenino , Proteína Forkhead Box L2/genética , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transducción de Señal/fisiología , Transcriptoma , Anomalías Urogenitales/genética
18.
Biol Reprod ; 103(5): 966-977, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32945847

RESUMEN

Development and functions of the ovary rely on appropriate signaling and communication between various ovarian cell types. FOXL2, a transcription factor that plays a key role at different stages of ovarian development, is associated with primary ovarian insufficiency and ovarian cancer as a result of its loss-of-function or mutations. In this study, we investigated the impact of aberrant, constitutive expression of FOXL2 in somatic cells of the ovary. Overexpression of FOXL2 that started during fetal life resulted in defects in nest breakdown and consequent formation of polyovular follicles. Granulosa cell differentiation was impaired and recruitment and differentiation of steroidogenic theca cells was compromised. As a consequence, adult ovaries overexpressing FOXL2 exhibited defects in compartmentalization of granulosa and theca cells, significant decreased steroidogenesis and lack of ovulation. These findings demonstrate that fine-tuned expression of FOXL2 is required for proper folliculogenesis and fertility.


Asunto(s)
Proteína Forkhead Box L2/metabolismo , Folículo Ovárico/metabolismo , Ovario/metabolismo , Animales , Diferenciación Celular/fisiología , Femenino , Proteína Forkhead Box L2/genética , Células de la Granulosa/metabolismo , Ratones , Mutación , Ovario/crecimiento & desarrollo , Células Tecales/metabolismo
19.
Sci Rep ; 10(1): 13551, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32782329

RESUMEN

Radiotherapy is a well-known cause of premature ovarian failure (POF). Therefore, we investigated the molecular influence of genistein (GEN) on the ovarian reserve of rats exposed to ϒ-radiation. Female Sprague Dawley rats were exposed to a 3.2 Gy γ-radiation to induce POF and/or treated with either GEN (5 mg/kg, i.p.) or Ethinyl estradiol (E2; 0.1 mg/kg, s.c.), once daily for 10 days. GEN was able to conserve primordial follicles stock and population of growing follicles accompanied with reduction in atretic follicles. GEN restored the circulating estradiol and anti-Müllerian hormone levels which were diminished after irradiation. GEN has potent antioxidant activity against radiation-mediated oxidative stress through upregulating endogenous glutathione levels and glutathione peroxidase activity. Mechanistically, GEN inhibited the intrinsic pathway of apoptosis by repressing Bax expression and augmenting Bcl-2 expression resulted in reduced Bax/Bcl-2 ratio with subsequent reduction in cytochrome c and caspase 3 expression. These promising effects of GEN are associated with improving granulosa cells proliferation. On the molecular basis, GEN reversed ovarian apoptosis through up-regulation of ER-ß and FOXL-2 with downregulation of TGF-ß expression, therefore inhibiting transition of primordial follicles to more growing follicles. GEN may constitute a novel therapeutic modality for safeguarding ovarian function of females' cancer survivors.


Asunto(s)
Apoptosis , Receptor beta de Estrógeno/metabolismo , Proteína Forkhead Box L2/metabolismo , Genisteína/farmacología , Ovario/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antioxidantes/farmacología , Receptor beta de Estrógeno/genética , Femenino , Proteína Forkhead Box L2/genética , Regulación de la Expresión Génica , Ovario/patología , Ovario/efectos de la radiación , Fitoestrógenos/farmacología , Radiación Ionizante , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/genética
20.
JCI Insight ; 5(16)2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32814714

RESUMEN

Granulosa cell tumors (GCT) are rare ovarian malignancies. Due to the lack of effective treatment in late relapse, there is a clear unmet need for novel therapies. Forkhead Box L2 (FOXL2) is a protein mainly expressed in granulosa cells (GC) and therefore is a rational therapeutic target. Since we identified tumor infiltrating lymphocytes (TILs) as the main immune population within GCT, TILs from 11 GCT patients were expanded, and their phenotypes were interrogated to determine that T cells acquired late antigen-experienced phenotypes and lower levels of PD1 expression. Importantly, TILs maintained their functionality after ex vivo expansion as they vigorously reacted against autologous tumors (100% of patients) and against FOXL2 peptides (57.1% of patients). To validate the relevance of FOXL2 as a target for immune therapy, we developed a plasmid DNA vaccine (FoxL2-tetanus toxin; FoxL2-TT) by fusing Foxl2 cDNA with the immune-enhancing domain of TT. Mice immunization with FoxL2-TT controlled growth of FOXL2-expressing ovarian (BR5) and breast (4T1) cancers in a T cell-mediated manner. Combination of anti-PD-L1 with FoxL2-TT vaccination further reduced tumor progression and improved mouse survival without affecting the female reproductive system and pregnancy. Together, our results suggest that FOXL2 immune targeting can produce substantial long-term clinical benefits. Our study can serve as a foundation for trials testing immunotherapeutic approaches in patients with ovarian GCT.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Proteína Forkhead Box L2/inmunología , Tumor de Células de la Granulosa/inmunología , Linfocitos Infiltrantes de Tumor/patología , Adulto , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Epítopos , Femenino , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Tumor de Células de la Granulosa/patología , Tumor de Células de la Granulosa/terapia , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos , Persona de Mediana Edad , Embarazo , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Microambiente Tumoral , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA