Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.908
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Cell ; 185(14): 2576-2590.e12, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35623357

ABSTRACT

Mouse germline cysts, on average, develop into six oocytes supported by 24 nurse cells that transfer cytoplasm and organelles to generate a Balbiani body. We showed that between E14.5 and P5, cysts periodically activate some nurse cells to begin cytoplasmic transfer, which causes them to shrink and turnover within 2 days. Nurse cells die by a programmed cell death (PCD) pathway involving acidification, similar to Drosophila nurse cells, and only infrequently by apoptosis. Prior to initiating transfer, nurse cells co-cluster by scRNA-seq with their pro-oocyte sisters, but during their final 2 days, they cluster separately. The genes promoting oocyte development and nurse cell PCD are upregulated, whereas the genes that repress transfer, such as Tex14, and oocyte factors, such as Nobox and Lhx8, are under-expressed. The transferred nurse cell centrosomes build a cytocentrum that establishes a large microtubule aster in the primordial oocyte that organizes the Balbiani body, defining the earliest oocyte polarity.


Subject(s)
Cell Lineage , Cysts , Oocytes , Animals , Apoptosis , Cell Enlargement , Cysts/genetics , Cysts/metabolism , Cytoplasm/metabolism , Drosophila melanogaster , Female , Gene Expression Regulation, Developmental , Mice , Oocytes/cytology , Oocytes/metabolism , Ovary/cytology , Ovary/embryology , Ovary/metabolism
2.
Nature ; 607(7919): 540-547, 2022 07.
Article in English | MEDLINE | ID: mdl-35794482

ABSTRACT

Gonadal development is a complex process that involves sex determination followed by divergent maturation into either testes or ovaries1. Historically, limited tissue accessibility, a lack of reliable in vitro models and critical differences between humans and mice have hampered our knowledge of human gonadogenesis, despite its importance in gonadal conditions and infertility. Here, we generated a comprehensive map of first- and second-trimester human gonads using a combination of single-cell and spatial transcriptomics, chromatin accessibility assays and fluorescent microscopy. We extracted human-specific regulatory programmes that control the development of germline and somatic cell lineages by profiling equivalent developmental stages in mice. In both species, we define the somatic cell states present at the time of sex specification, including the bipotent early supporting population that, in males, upregulates the testis-determining factor SRY and sPAX8s, a gonadal lineage located at the gonadal-mesonephric interface. In females, we resolve the cellular and molecular events that give rise to the first and second waves of granulosa cells that compartmentalize the developing ovary to modulate germ cell differentiation. In males, we identify human SIGLEC15+ and TREM2+ fetal testicular macrophages, which signal to somatic cells outside and inside the developing testis cords, respectively. This study provides a comprehensive spatiotemporal map of human and mouse gonadal differentiation, which can guide in vitro gonadogenesis.


Subject(s)
Cell Lineage , Germ Cells , Ovary , Sex Differentiation , Single-Cell Analysis , Testis , Animals , Chromatin/genetics , Chromatin/metabolism , Female , Germ Cells/cytology , Germ Cells/metabolism , Granulosa Cells/cytology , Granulosa Cells/metabolism , Humans , Immunoglobulins , Macrophages/metabolism , Male , Membrane Glycoproteins , Membrane Proteins , Mice , Microscopy, Fluorescence , Ovary/cytology , Ovary/embryology , PAX8 Transcription Factor , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Receptors, Immunologic , Sex Differentiation/genetics , Testis/cytology , Testis/embryology , Transcriptome
3.
Mol Cell ; 66(3): 411-419.e4, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28457744

ABSTRACT

Most piRNAs in the Drosophila female germline are transcribed from heterochromatic regions called dual-strand piRNA clusters. Histone 3 lysine 9 trimethylation (H3K9me3) is required for licensing piRNA production by these clusters. However, it is unclear when and how they acquire this permissive heterochromatic state. Here, we show that transient Piwi depletion in Drosophila embryos results in H3K9me3 decrease at piRNA clusters in ovaries. This is accompanied by impaired biogenesis of ovarian piRNAs, accumulation of transposable element transcripts, and female sterility. Conversely, Piwi depletion at later developmental stages does not disturb piRNA cluster licensing. These results indicate that the identity of piRNA clusters is epigenetically acquired in a Piwi-dependent manner during embryonic development, which is reminiscent of the widespread genome reprogramming occurring during early mammalian zygotic development.


Subject(s)
Argonaute Proteins/metabolism , DNA Methylation , DNA Transposable Elements , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epigenetic Repression , Heterochromatin/metabolism , Ovary/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Age Factors , Animals , Argonaute Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Female , Fertility , Gene Expression Regulation, Developmental , Heterochromatin/genetics , Histones/metabolism , Infertility, Female/genetics , Infertility, Female/metabolism , Infertility, Female/physiopathology , Methylation , Morphogenesis , Ovary/embryology , Protein Binding , RNA, Small Interfering/genetics
4.
EMBO J ; 39(21): e104929, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32954504

ABSTRACT

Human germ cells perpetuate human genetic and epigenetic information. However, the underlying mechanism remains elusive, due to a lack of appropriate experimental systems. Here, we show that human primordial germ cell-like cells (hPGCLCs) derived from human-induced pluripotent stem cells (hiPSCs) can be propagated to at least ~106 -fold over a period of 4 months under a defined condition in vitro. During expansion, hPGCLCs maintain an early hPGC-like transcriptome and preserve their genome-wide DNA methylation profiles, most likely due to retention of maintenance DNA methyltransferase activity. These characteristics contrast starkly with those of mouse PGCLCs, which, under an analogous condition, show a limited propagation (up to ~50-fold) and persist only around 1 week, yet undergo cell-autonomous genome-wide DNA demethylation. Importantly, upon aggregation culture with mouse embryonic ovarian somatic cells in xenogeneic-reconstituted ovaries, expanded hPGCLCs initiate genome-wide DNA demethylation and differentiate into oogonia/gonocyte-like cells, demonstrating their germline potential. By creating a paradigm for hPGCLC expansion, our study uncovers critical divergences in expansion potential and the mechanism for epigenetic reprogramming between the human and mouse germ cell lineage.


Subject(s)
Germ Cells/metabolism , Ovary/embryology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Line , DNA Demethylation , DNA Methylation , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Epigenomics , Female , Genome , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice
5.
Development ; 148(4)2021 02 26.
Article in English | MEDLINE | ID: mdl-33526583

ABSTRACT

Basement membranes (BM) are extracellular matrices assembled into complex and highly organized networks essential for organ morphogenesis and function. However, little is known about the tissue origin of BM components and their dynamics in vivo Here, we unravel the assembly and role of the BM main component, Collagen type IV (ColIV), in Drosophila ovarian stalk morphogenesis. Stalks are short strings of cells assembled through cell intercalation that link adjacent follicles and maintain ovarian integrity. We show that stalk ColIV has multiple origins and is assembled following a regulated pattern leading to a unique BM organisation. Absence of ColIV leads to follicle fusion, as observed upon ablation of stalk cells. ColIV and integrins are both required to trigger cell intercalation and maintain mechanically strong cell-cell attachment within the stalk. These results show how the dynamic assembly of a mosaic BM controls complex tissue morphogenesis and integrity.


Subject(s)
Basement Membrane/metabolism , Cell Communication , Drosophila/embryology , Drosophila/metabolism , Ovary/embryology , Ovary/metabolism , Animals , Collagen Type IV/metabolism , Extracellular Matrix/metabolism , Female , Fluorescent Antibody Technique , Morphogenesis , Organogenesis , Pituitary Gland/embryology , Pituitary Gland/metabolism
6.
Development ; 148(16)2021 08 15.
Article in English | MEDLINE | ID: mdl-34387307

ABSTRACT

During early embryogenesis in amniotic vertebrates, the gonads differentiate into either ovaries or testes. The first cell lineage to differentiate gives rise to the supporting cells: Sertoli cells in males and pre-granulosa cells in females. These key cell types direct the differentiation of the other cell types in the gonad, including steroidogenic cells. The gonadal surface epithelium and the interstitial cell populations are less well studied, and little is known about their sexual differentiation programs. Here, we show the requirement of the homeobox transcription factor gene TGIF1 for ovarian development in the chicken embryo. TGIF1 is expressed in the two principal ovarian somatic cell populations: the cortex and the pre-granulosa cells of the medulla. TGIF1 expression is associated with an ovarian phenotype in estrogen-mediated sex reversal experiments. Targeted misexpression and gene knockdown indicate that TGIF1 is required, but not sufficient, for proper ovarian cortex formation. In addition, TGIF1 is identified as the first known regulator of juxtacortical medulla development. These findings provide new insights into chicken ovarian differentiation and development, specifically cortical and juxtacortical medulla formation.


Subject(s)
Chickens/genetics , Genes, Homeobox , Homeodomain Proteins/genetics , Ovary/embryology , Repressor Proteins/genetics , Animals , Cell Differentiation , Cell Lineage/genetics , Chick Embryo , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Gonads/metabolism , Homeodomain Proteins/metabolism , Male , Ovary/cytology , Ovary/metabolism , Repressor Proteins/metabolism , Sertoli Cells/metabolism , Sex Determination Processes/genetics , Sex Differentiation/genetics , Testis/metabolism
7.
Cell Commun Signal ; 22(1): 330, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879537

ABSTRACT

Sex-specific gonadal differentiation is directed by complex signalling promoting development in either male or female direction, while simultaneously inhibiting the opposite pathway. In mice, the WNT/ß-catenin pathway promotes ovarian development and the importance of actively inhibiting this pathway to ensure normal testis development has been recognised. However, the implications of alterations in the tightly regulated WNT/ß-catenin signalling during human fetal gonad development has not yet been examined in detail. Thus, the aim of this study was to examine the consequences of dysregulating the WNT/ß-catenin signalling pathway in the supporting cell lineage during sex-specific human fetal gonad development using an established and extensively validated ex vivo culture model. Inhibition of WNT/ß-catenin signalling in human fetal ovary cultures resulted in only minor effects, including reduced secretion of RSPO1 and reduced cell proliferation although this was not consistently found in all treatment groups. In contrast, promotion of WNT/ß-catenin signalling in testes severely affected development and function. This included disrupted seminiferous cord structures, reduced cell proliferation, reduced expression of SOX9/AMH, reduced secretion of Inhibin B and AMH as well as loss of the germ cell population. Additionally, Leydig cell function was markedly impaired with reduced secretion of testosterone, androstenedione and INSL3. Together, this study suggests that dysregulated WNT/ß-catenin signalling during human fetal gonad development severely impairs testicular development and function. Importantly, our study highlights the notion that sufficient inhibition of the opposite pathway during sex-specific gonadal differentiation is essential to ensure normal development and function also applies to human fetal gonads.


Subject(s)
Testis , Wnt Signaling Pathway , Humans , Male , Testis/metabolism , Testis/embryology , Female , Sex Differentiation/genetics , Fetus/metabolism , Cell Differentiation , Cell Proliferation , beta Catenin/metabolism , Leydig Cells/metabolism , Leydig Cells/cytology , Ovary/metabolism , Ovary/embryology
8.
PLoS Genet ; 17(3): e1009489, 2021 03.
Article in English | MEDLINE | ID: mdl-33780456

ABSTRACT

Here we show that multiple modes of Notch signaling activation specify the complexity of spatial cellular interactions necessary for stem cell niche assembly. In particular, we studied the formation of the germline stem cell niche in Drosophila ovaries, which is a two-step process whereby terminal filaments are formed first. Then, terminal filaments signal to the adjacent cap cell precursors, resulting in Notch signaling activation, which is necessary for the lifelong acquisition of stem cell niche cell fate. The genetic data suggest that in order to initiate the process of stem cell niche assembly, Notch signaling is activated among non-equipotent cells via distant induction, where germline Delta is delivered to somatic cells located several diameters away via cellular projections generated by primordial germ cells. At the same time, to ensure the robustness of niche formation, terminal filament cell fate can also be induced by somatic Delta via cis- or trans-inhibition. This exemplifies a double security mechanism that guarantees that the germline stem cell niche is formed, since it is indispensable for the adjacent germline precursor cells to acquire and maintain stemness necessary for successful reproduction. These findings contribute to our understanding of the formation of stem cell niches in their natural environment, which is important for stem cell biology and regenerative medicine.


Subject(s)
Receptors, Notch/metabolism , Signal Transduction , Stem Cell Niche , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Drosophila , Female , Germ Cells/metabolism , Models, Biological , Mutation , Organogenesis/genetics , Ovary/embryology , Ovary/metabolism , Receptors, Notch/genetics , Stem Cell Niche/genetics
9.
Development ; 147(4)2020 02 25.
Article in English | MEDLINE | ID: mdl-32001442

ABSTRACT

In chickens, the embryonic ovary differentiates into two distinct domains before meiosis: a steroidogenic core (the female medulla), overlain by the germ cell niche (the cortex). The differentiation of the medulla is a cell-autonomous process based on chromosomal sex identity (CASI). In order to address the extent to which cortex differentiation depends on intrinsic or extrinsic factors, we generated models of gonadal intersex by mixing ZW (female) and ZZ (male) cells in gonadal chimeras, or by altering oestrogen levels of ZW and ZZ embryos. We found that CASI does not apply to the embryonic cortex. Both ZW and ZZ cells can form the cortex and this can happen independently of the phenotypic sex of the medulla as long as oestrogen is provided. We also show that the cortex-promoting activity of oestrogen signalling is mediated via estrogen receptor alpha within the left gonad epithelium. However, the presence of a medulla with an 'intersex' or male phenotype may compromise germ cell progression into meiosis, causing cortical germ cells to remain in an immature state in the embryo.


Subject(s)
Estrogens/physiology , Oogenesis , Ovary/embryology , Animals , Chick Embryo , Chickens/genetics , Chromosomes/genetics , Electroporation , Epithelial Cells/cytology , Female , Germ Cells/cytology , Gonads/cytology , Male , Meiosis , Mitosis , Phenotype , Sex Chromosomes , Sex Differentiation/genetics , Signal Transduction
10.
Development ; 147(6)2020 03 30.
Article in English | MEDLINE | ID: mdl-32108023

ABSTRACT

Members of the Iroquois B (IrxB) homeodomain cluster genes, specifically Irx3 and Irx5, are crucial for heart, limb and bone development. Recently, we reported their importance for oocyte and follicle survival within the developing ovary. Irx3 and Irx5 expression begins after sex determination in the ovary but remains absent in the fetal testis. Mutually antagonistic molecular signals ensure ovary versus testis differentiation with canonical Wnt/ß-catenin signals paramount for promoting the ovary pathway. Notably, few direct downstream targets have been identified. We report that Wnt/ß-catenin signaling directly stimulates Irx3 and Irx5 transcription in the developing ovary. Using in silico analysis of ATAC- and ChIP-Seq databases in conjunction with mouse gonad explant transfection assays, we identified TCF/LEF-binding sequences within two distal enhancers of the IrxB locus that promote ß-catenin-responsive ovary expression. Meanwhile, Irx3 and Irx5 transcription is suppressed within the developing testis by the presence of H3K27me3 on these same sites. Thus, we resolved sexually dimorphic regulation of Irx3 and Irx5 via epigenetic and ß-catenin transcriptional control where their ovarian presence promotes oocyte and follicle survival vital for future ovarian health.


Subject(s)
Epigenesis, Genetic/physiology , Gonads/embryology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Gonads/metabolism , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Ovary/embryology , Ovary/metabolism , Sex Characteristics , Sex Differentiation/genetics , Testis/embryology , Testis/metabolism , Transcription Factors/metabolism
11.
Annu Rev Cell Dev Biol ; 25: 457-82, 2009.
Article in English | MEDLINE | ID: mdl-19807280

ABSTRACT

A critical element of successful sexual reproduction is the generation of sexually dimorphic adult reproductive organs, the testis and ovary, which produce functional gametes. Examination of different vertebrate species shows that the adult gonad is remarkably similar in its morphology across different phylogenetic classes. Surprisingly, however, the cellular and molecular programs employed to create similar organs are not evolutionarily conserved. We highlight the mechanisms used by different vertebrate model systems to generate the somatic architecture necessary to support gametogenesis. In addition, we examine the different vertebrate patterns of germ cell migration from their site of origin to colonize the gonad and highlight their roles in sex-specific morphogenesis. We also discuss the plasticity of the adult gonad and consider how different genetic and environmental conditions can induce transitions between testis and ovary morphology.


Subject(s)
Gene Expression Regulation, Developmental , Ovary/embryology , Testis/embryology , Vertebrates/embryology , Animals , Cell Movement , Female , Humans , Male , Morphogenesis , Ovary/metabolism , Sex Differentiation , Testis/metabolism
12.
Cell Mol Life Sci ; 78(2): 695-713, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32367190

ABSTRACT

Meiosis is one of the most finely orchestrated events during gametogenesis with distinct developmental patterns in males and females. However, the molecular mechanisms involved in this process remain not well known. Here, we report detailed transcriptome analyses of cell populations present in the mouse female gonadal ridges (E11.5) and the embryonic ovaries from E12.5 to E14.5 using single-cell RNA sequencing (scRNA seq). These periods correspond with the initiation and progression of meiosis throughout the first stage of prophase I. We identified 13 transcriptionally distinct cell populations and 7 transcriptionally distinct germ cell subclusters that correspond to mitotic (3 clusters) and meiotic (4 clusters) germ cells. By analysing cluster-specific gene expression profiles, we found four cell clusters correspond to different cell stages en route to meiosis and characterized their detailed transcriptome dynamics. Our scRNA seq analysis here represents a new important resource for deciphering the molecular pathways driving female meiosis initiation.


Subject(s)
Gene Expression Profiling/methods , Meiosis , Ovary/cytology , Single-Cell Analysis/methods , Transcriptome , Animals , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Ovary/embryology
13.
Development ; 145(7)2018 04 04.
Article in English | MEDLINE | ID: mdl-29511022

ABSTRACT

Maintenance of cellular identity is essential for tissue development and homeostasis. At the molecular level, cell identity is determined by the coordinated activation and repression of defined sets of genes. The tumor suppressor L(3)mbt has been shown to secure cellular identity in Drosophila larval brains by repressing germline-specific genes. Here, we interrogate the temporal and spatial requirements for L(3)mbt in the Drosophila ovary, and show that it safeguards the integrity of both somatic and germline tissues. l(3)mbt mutant ovaries exhibit multiple developmental defects, which we find to be largely caused by the inappropriate expression of a single gene, nanos, a key regulator of germline fate, in the somatic ovarian cells. In the female germline, we find that L(3)mbt represses testis-specific and neuronal genes. At the molecular level, we show that L(3)mbt function in the ovary is mediated through its co-factor Lint-1 but independently of the dREAM complex. Together, our work uncovers a more complex role for L(3)mbt than previously understood and demonstrates that L(3)mbt secures tissue identity by preventing the simultaneous expression of original identity markers and tissue-specific misexpression signatures.


Subject(s)
Cell Differentiation/genetics , Drosophila Proteins/metabolism , Drosophila/metabolism , Ovary/metabolism , Animals , Drosophila/embryology , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , In Situ Hybridization, Fluorescence , Larva/metabolism , Ovary/cytology , Ovary/embryology , RNA-Binding Proteins/metabolism , Sequence Analysis, RNA
14.
Biol Reprod ; 105(2): 491-502, 2021 08 03.
Article in English | MEDLINE | ID: mdl-33912929

ABSTRACT

Sex determination requires the commitment of bipotential gonads to either a testis or an ovarian fate. Gene deletion of the kinase Map3k4 results in gonadal sex reversal in XY mice, and transgenic re-expression of Map3k4 rescues the sex reversal phenotype. Map3k4 encodes a large, multi-functional protein possessing a kinase domain and several, additional protein-protein interaction domains. Although MAP3K4 plays a critical role in male gonadal sex determination, it is unknown if the kinase activity of MAP3K4 is required. Here, we use mice expressing full-length, kinase-inactive MAP3K4 from the endogenous Map3k4 locus to examine the requirement of MAP3K4 kinase activity in sex determination. Although homozygous kinase-inactivation of MAP3K4 (Map3k4KI/KI) is lethal, a small fraction survive to adulthood. We show Map3k4KI/KI adults exhibit a 4:1 female-biased sex ratio. Many adult Map3k4KI/KI phenotypic females have a Y chromosome. XY Map3k4KI/KI adults with sex reversal display female mating behavior, but do not give rise to offspring. Reproductive organs are overtly female, but there is a broad spectrum of ovarian phenotypes, including ovarian absence, primitive ovaries, reduced ovarian size, and ovaries having follicles in all stages of development. Further, XY Map3k4KI/KI adults are smaller than either male or female Map3k4WT/WT mice. Examination of the critical stage of gonadal sex determination at E11.5 shows that loss of MAP3K4 kinase activity results in the loss of Sry expression in XY Map3k4KI/KI embryos, indicating embryonic male gonadal sex reversal. Together, these findings demonstrate the essential role for kinase activity of MAP3K4 in male gonadal sex determination.


Subject(s)
MAP Kinase Kinase Kinase 4/genetics , Mice/genetics , Ovary/embryology , Sex Determination Processes/genetics , Testis/embryology , Animals , Female , MAP Kinase Kinase Kinase 4/metabolism , Male , Mice/embryology
15.
FASEB J ; 34(11): 14440-14457, 2020 11.
Article in English | MEDLINE | ID: mdl-32892421

ABSTRACT

Nerve growth factor (NGF) is critical for the development and maintenance of the peripheral sympathetic neurons. NGF is also involved in the ovarian sympathetic innervation and in the development and maintenance of folliculogenesis. Women with the endocrine disorder, polycystic ovary syndrome (PCOS), have an increased sympathetic nerve activity and increased ovarian NGF levels. The role of ovarian NGF excess in the PCOS pathophysiology and in the PCOS-related features is unclear. Here, using transgenic mice overexpressesing NGF in the ovarian theca cells (17NF mice), we assessed the female embryonic development, and the reproductive and metabolic profile in adult females. Ovarian NGF excess caused growth restriction in the female fetuses, and a delayed gonocyte and primary oocyte maturation. In adulthood, the 17NF mice displayed irregular estrous cycles and altered ovarian expression of steroidogenic and epigenetic markers. They also exhibited an increased sympathetic output with increased circulating dopamine, and metabolic dysfunction reflected by aberrant adipose tissue morphology and function, impaired glucose metabolism, decreased energy expenditure, and hepatic steatosis. These findings indicate that ovarian NGF excess leads to adverse fetal development and to reproductive and metabolic complications in adulthood, mirroring common features of PCOS. This work provides evidence that NGF excess may be implicated in the PCOS pathophysiology.


Subject(s)
Fetal Development , Nerve Growth Factor/genetics , Ovary/metabolism , Polycystic Ovary Syndrome/genetics , Animals , Cells, Cultured , Dopamine/metabolism , Estrous Cycle , Female , Mice , Nerve Growth Factor/metabolism , Oogenesis , Ovary/embryology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Up-Regulation
16.
PLoS Genet ; 14(2): e1007203, 2018 02.
Article in English | MEDLINE | ID: mdl-29389999

ABSTRACT

Reproduction in sexually dimorphic animals relies on successful gamete production, executed by the germline and aided by somatic support cells. Somatic sex identity in Drosophila is instructed by sex-specific isoforms of the DMRT1 ortholog Doublesex (Dsx). Female-specific expression of Sex-lethal (Sxl) causes alternative splicing of transformer (tra) to the female isoform traF. In turn, TraF alternatively splices dsx to the female isoform dsxF. Loss of the transcriptional repressor Chinmo in male somatic stem cells (CySCs) of the testis causes them to "feminize", resembling female somatic stem cells in the ovary. This somatic sex transformation causes a collapse of germline differentiation and male infertility. We demonstrate this feminization occurs by transcriptional and post-transcriptional regulation of traF. We find that chinmo-deficient CySCs upregulate tra mRNA as well as transcripts encoding tra-splice factors Virilizer (Vir) and Female lethal (2)d (Fl(2)d). traF splicing in chinmo-deficient CySCs leads to the production of DsxF at the expense of the male isoform DsxM, and both TraF and DsxF are required for CySC sex transformation. Surprisingly, CySC feminization upon loss of chinmo does not require Sxl but does require Vir and Fl(2)d. Consistent with this, we show that both Vir and Fl(2)d are required for tra alternative splicing in the female somatic gonad. Our work reveals the need for transcriptional regulation of tra in adult male stem cells and highlights a previously unobserved Sxl-independent mechanism of traF production in vivo. In sum, transcriptional control of the sex determination hierarchy by Chinmo is critical for sex maintenance in sexually dimorphic tissues and is vital in the preservation of fertility.


Subject(s)
Drosophila Proteins/physiology , Nerve Tissue Proteins/physiology , Nuclear Proteins/genetics , Sex Determination Processes/genetics , Sex Differentiation/genetics , Testis/embryology , Alternative Splicing/genetics , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , DNA-Binding Proteins/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental , Male , Nerve Tissue Proteins/genetics , Ovary/embryology , Ovary/metabolism , RNA-Binding Proteins/physiology , Testis/metabolism
17.
Int J Mol Sci ; 22(18)2021 Sep 18.
Article in English | MEDLINE | ID: mdl-34576272

ABSTRACT

Follicle stimulating hormone (FSH) is produced by the pituitary gland in a coordinated hypothalamic-pituitary-gonadal (HPG) axis event, plays important roles in reproduction and germ cell development during different phases of reproductive development (fetal, neonatal, puberty, and adult life), and is consequently essential for fertility. FSH is a heterodimeric glycoprotein hormone of two dissociable subunits, α and ß. The FSH ß-subunit (FSHß) function starts upon coupling to its specific receptor: follicle-stimulating hormone receptor (FSHR). FSHRs are localized mainly on the surface of target cells on the testis and ovary (granulosa and Sertoli cells) and have recently been found in testicular stem cells and extra-gonadal tissue. Several reproduction disorders are associated with absent or low FSH secretion, with mutation of the FSH ß-subunit or the FSH receptor, and/or its signaling pathways. However, the influence of FSH on germ cells is still poorly understood; some studies have suggested that this hormone also plays a determinant role in the self-renewal of germinative cells and acts to increase undifferentiated spermatogonia proliferation. In addition, in vitro, together with other factors, it assists the process of differentiation of primordial germ cells (PGCLCs) into gametes (oocyte-like and SSCLCs). In this review, we describe relevant research on the influence of FSH on spermatogenesis and folliculogenesis, mainly in the germ cell of humans and other species. The possible roles of FSH in germ cell generation in vitro are also presented.


Subject(s)
Follicle Stimulating Hormone/metabolism , Granulosa Cells/metabolism , Ovary/metabolism , Sertoli Cells/metabolism , Animals , Dimerization , Female , Fertility , Follicle Stimulating Hormone, beta Subunit/metabolism , Germ Cells/metabolism , Gonadotropins/metabolism , Humans , Male , Mice , Ovary/embryology , Ovary/growth & development , Pituitary Gland/embryology , Pituitary Gland/growth & development , Pituitary Gland/metabolism , Rats , Receptors, FSH/metabolism , Reproduction , Sexual Maturation , Spermatogenesis/genetics , Spermatogonia/cytology
18.
Dev Biol ; 452(2): 127-133, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31071312

ABSTRACT

Understanding how cell fate decisions are regulated is a central question in stem cell biology. Recent studies have demonstrated that intracellular pH (pHi) dynamics contribute to this process. Indeed, the pHi of cells within a tissue is not simply a consequence of chemical reactions in the cytoplasm and other cellular activity, but is actively maintained at a specific setpoint in each cell type. We found previously that the pHi of cells in the follicle stem cell (FSC) lineage in the Drosophila ovary increases progressively during differentiation from an average of 6.8 in the FSCs, to 7.0 in newly produced daughter cells, to 7.3 in more differentiated cells. Two major regulators of pHi in this lineage are Drosophila sodium-proton exchanger 2 (dNhe2) and a previously uncharacterized gene, CG8177, that is homologous to mammalian anion exchanger 2 (AE2). Based on this homology, we named the gene anion exchanger 2 (ae2). Here, we generated null alleles of ae2 and found that homozygous mutant flies are viable but have severe defects in ovary development and adult oogenesis. Specifically, we find that ae2 null flies have smaller ovaries, reduced fertility, and impaired follicle formation. In addition, we find that the follicle formation defect can be suppressed by a decrease in dNhe2 copy number and enhanced by the overexpression of dNhe2, suggesting that this phenotype is due to the dysregulation of pHi. These findings support the emerging idea that pHi dynamics regulate cell fate decisions and our studies provide new genetic tools to investigate the mechanisms by which this occurs.


Subject(s)
Antiporters/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Oogenesis , Ovary/embryology , Ovary/metabolism , Animals , Drosophila melanogaster/genetics , Epistasis, Genetic , Female , Fertility , Mutation/genetics , Organ Size , Ovarian Follicle/embryology , RNA Interference , Sequence Homology, Amino Acid
19.
Hum Mol Genet ; 27(1): 190-198, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29145650

ABSTRACT

CREB-binding protein (CBP, CREBBP, KAT3A) and its closely related paralogue p300 (EP300, KAT3B), together termed p300/CBP, are histone/lysine acetyl-transferases that control gene expression by modifying chromatin-associated proteins. Here, we report roles for both of these chromatin-modifying enzymes in mouse sex determination, the process by which the embryonic gonad develops into a testis or an ovary. By targeting gene ablation to embryonic gonadal somatic cells using an inducible Cre line, we show that gonads lacking either gene exhibit major abnormalities of XY gonad development at 14.5 dpc, including partial sex reversal. Embryos lacking three out of four functional copies of p300/Cbp exhibit complete XY gonadal sex reversal and have greatly reduced expression of the key testis-determining genes Sry and Sox9. An analysis of histone acetylation at the Sry promoter in mutant gonads at 11.5 dpc shows a reduction in levels of the positive histone mark H3K27Ac. Our data suggest a role for CBP/p300 in testis determination mediated by control of histone acetylation at the Sry locus and reveal a novel element in the epigenetic control of Sry and mammalian sex determination. They also suggest possible novel causes of human disorders of sex development (DSD).


Subject(s)
CREB-Binding Protein/deficiency , Disorders of Sex Development/metabolism , E1A-Associated p300 Protein/deficiency , Histones/metabolism , Sex Determination Processes/physiology , Sex-Determining Region Y Protein/genetics , Testis/embryology , Acetylation , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Disorders of Sex Development/genetics , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Female , Male , Mice , Ovary/embryology , Ovary/metabolism , Promoter Regions, Genetic , Sex-Determining Region Y Protein/metabolism , Testis/metabolism
20.
Biochem Biophys Res Commun ; 523(4): 823-828, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31948756

ABSTRACT

Although studies have focused on extracellular matrix (ECM) remodeling during the formation and functioning of adult ovaries, there is no comprehensive understanding of the mechanisms controlling preantral follicle development in fetal bovine ovaries during gestation. Thus, to gain insights into ECM remodeling during initial ovarian development, we used fetal ovaries to quantify the fractal dimension (FD), total collagen, and relative mRNA abundance of genes related to ECM remodeling (COL1A1, COL1A2, COL4A1, MMP2, MMP9, MMP14, TIMP1, and TIMP2). For this, pairs of fetal ovaries were obtained from cows in a local abattoir at days 60, 90, 120, and 150 of gestation; one of each pair was submitted to RNA extraction for target transcript analysis, and the other was used for total collagen and FD evaluation. From day 120 total collagen appeared to occupy a greater area in the fetal ovary. The fractal analysis with picrosirius red staining shows higher at day 150 when compared with that on day 60. On the contrary, we found an inverse pattern when we used the hematoxylin and eosin staining approach. Concerning target gene expression, the relative abundances of COL1A1, COL4A1, MMP2, MMP14, TIMP1, and TIMP2 mRNA were higher on day 150 when compared with that on day 60. We conclude that fractal analysis reflects the morphological changes occurring during structural organization of the fetal ovary and that the expression of genes related to ECM remodeling is modulated throughout gestation in bovine fetal ovaries.


Subject(s)
Extracellular Matrix/metabolism , Fetus/embryology , Fractals , Ovary/embryology , Ovary/metabolism , Animals , Cattle , Female , Gene Expression Regulation, Developmental , Models, Biological , Phenotype , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL