Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Front Endocrinol (Lausanne) ; 14: 1124143, 2023.
Article in English | MEDLINE | ID: mdl-37576970

ABSTRACT

Background: SOX17 has been identified as a critical factor in specification of human primordial germ cells, but whether SOX17 regulates development of germ cells after sex differentiation is poorly understood. Methods: We collected specimens of gonadal ridge from an embryo (n=1), and ovaries of foetuses (n=23) and adults (n=3). Germ cells were labelled with SOX17, VASA (classic germ cells marker), phosphohistone H3 (PHH3, mitosis marker) and synaptonemal complex protein 3 (SCP3, meiosis marker). Results: SOX17 was detected in both cytoplasm and nucleus of oogonia and oocytes of primordial and primary follicles from 15 to 28 gestational weeks (GW). However, it was exclusively expressed in cytoplasm of oogonia at 7 GW, and in nucleus of oocytes in secondary follicles. Co-expression rates of SOX17 in VASA+ germ cells ranged from 81.29% to 97.81% in foetuses. Co-staining rates of SOX17 and PHH3 or SCP3 were 0%-34% and 0%-57%, respectively. Interestingly, we distinguished a subpopulation of SOX17+VASA- germ cells in fetal ovaries. These cells clustered in the cortex and could be co-stained with the mitosis marker PHH3 but not the meiosis marker SCP3. Conclusions: The dynamic expression of SOX17 was detected in human female germ cells. We discovered a population of SOX17+ VASA- germ cells clustering at the cortex of ovaries. We could not find a relationship between mitosis or meiosis and SOX17 or VASA staining in germ cells. Our findings provide insight into the potential role of SOX17 involving germ cells maturation after specification, although the mechanism is unclear and needs further investigation.


Subject(s)
Germ Cells , Ovary , Humans , Female , Adult , Ovary/metabolism , Oocytes , Oogonia/metabolism , Fetus , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
2.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34878097

ABSTRACT

Gamete formation from germline stem cells (GSCs) is essential for sexual reproduction. However, the regulation of GSC differentiation is incompletely understood. Set2, which deposits H3K36me3 modifications, is required for GSC differentiation during Drosophila oogenesis. We discovered that the H3K36me3 reader Male-specific lethal 3 (Msl3) and histone acetyltransferase complex Ada2a-containing (ATAC) cooperate with Set2 to regulate GSC differentiation in female Drosophila. Msl3, acting independently of the rest of the male-specific lethal complex, promotes transcription of genes, including a germline-enriched ribosomal protein S19 paralog RpS19b. RpS19b upregulation is required for translation of RNA-binding Fox protein 1 (Rbfox1), a known meiotic cell cycle entry factor. Thus, Msl3 regulates GSC differentiation by modulating translation of a key factor that promotes transition to an oocyte fate.


Subject(s)
Drosophila Proteins/metabolism , Nuclear Proteins/metabolism , Oogenesis , Oogonia/metabolism , Transcription Factors/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Meiosis , Nuclear Proteins/genetics , Oogonia/cytology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Transcription Factors/genetics
3.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34495316

ABSTRACT

Emerging evidence suggests that ribosome heterogeneity may have important functional consequences in the translation of specific mRNAs within different cell types and under various conditions. Ribosome heterogeneity comes in many forms, including post-translational modification of ribosome proteins (RPs), absence of specific RPs and inclusion of different RP paralogs. The Drosophila genome encodes two RpS5 paralogs: RpS5a and RpS5b. While RpS5a is ubiquitously expressed, RpS5b exhibits enriched expression in the reproductive system. Deletion of RpS5b results in female sterility marked by developmental arrest of egg chambers at stages 7-8, disruption of vitellogenesis and posterior follicle cell (PFC) hyperplasia. While transgenic rescue experiments suggest functional redundancy between RpS5a and RpS5b, molecular, biochemical and ribo-seq experiments indicate that RpS5b mutants display increased rRNA transcription and RP production, accompanied by increased protein synthesis. Loss of RpS5b results in microtubule-based defects and in mislocalization of Delta and Mindbomb1, leading to failure of Notch pathway activation in PFCs. Together, our results indicate that germ cell-specific expression of RpS5b promotes proper egg chamber development by ensuring the homeostasis of functional ribosomes.


Subject(s)
Infertility/genetics , Oogenesis , Oogonia/metabolism , Ovarian Follicle/metabolism , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mutation , Oogonia/cytology , Ovarian Follicle/cytology , Protein Transport , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Receptors, Notch/metabolism , Signal Transduction
4.
Cells Dev ; 167: 203710, 2021 09.
Article in English | MEDLINE | ID: mdl-34171535

ABSTRACT

P-Element-induced wimpy testis (Piwi) subfamily proteins form complexes that bind to Piwi-interacting RNA. This interaction is crucial for stem cell regulation and formation, maintenance of germline stem cells, and gametogenesis in several metazoans. Planarians are effective models for studying stem cells. In the planarian Dugesia ryukyuensis, DrPiwi-1 is essential for the development of germ cells, but not somatic cells and sexual organs. DrPiwi-2 is indispensable for regeneration. In this study, we aimed to investigate the effects of Piwi on the differentiation of germ cells using monoclonal antibodies against DrPiwi-1 and DrPiwi-2. DrPiwi-1 and DrPiwi-2 co-localized more in immature germ cells than in mature germ cells in the ovary. DrPiwi-1 was found in the cytoplasm of early oogonia as undifferentiated germ cells, whereas DrPiwi-2 was found to localize not only in the nuclei but also in the cytoplasm of early oogonia. In descendant germ cells (oocytes), DrPiwi-2 was not present in the cytoplasm, but was strongly detected in the nucleolus. Moreover, we found that DrPiwi-1 forms a complex with DrPiwi-2. The cause of DrPiwi-1 depletion may be the severe reduction in the DrPiwi-2 level in the cytoplasm of oogonia. These results suggest that the formation of the DrPiwi-1 and DrPiwi-2 complex in the cytoplasm of oogonia is essential for oocyte differentiation. Our findings support the conclusion that DrPiwi-1 forms a complex with DrPiwi-2 in the cytoplasm of undifferentiated germ cells, and it signifies the start of gametogenesis. In contrast, in the testes, Drpiwi-1 was found in undifferentiated germ cells (spermatogonia), whereas DrPiwi-2 was found in descendant germ cells (spermatocytes). The process of germ cell differentiation from adult stem cells in planarians may be regulated in different ways in female and male germ lines by the Piwi family.


Subject(s)
Cell Differentiation , Cytoplasm/metabolism , Oocytes/cytology , Oogonia/metabolism , Planarians/cytology , Planarians/metabolism , Proteins/metabolism , Animals , Female , Male , Ovary/metabolism , Testis/metabolism
5.
Nat Commun ; 12(1): 3492, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108460

ABSTRACT

In the Caenorhabditis elegans germline, thousands of mRNAs are concomitantly expressed with antisense 22G-RNAs, which are loaded into the Argonaute CSR-1. Despite their essential functions for animal fertility and embryonic development, how CSR-1 22G-RNAs are produced remains unknown. Here, we show that CSR-1 slicer activity is primarily involved in triggering the synthesis of small RNAs on the coding sequences of germline mRNAs and post-transcriptionally regulates a fraction of targets. CSR-1-cleaved mRNAs prime the RNA-dependent RNA polymerase, EGO-1, to synthesize 22G-RNAs in phase with translating ribosomes, in contrast to other 22G-RNAs mostly synthesized in germ granules. Moreover, codon optimality and efficient translation antagonize CSR-1 slicing and 22G-RNAs biogenesis. We propose that codon usage differences encoded into mRNA sequences might be a conserved strategy in eukaryotes to regulate small RNA biogenesis and Argonaute targeting.


Subject(s)
Argonaute Proteins/metabolism , Caenorhabditis elegans Proteins/metabolism , Codon Usage , Protein Biosynthesis , RNA, Small Interfering/biosynthesis , Animals , Argonaute Proteins/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Catalysis , Cytosol/metabolism , Mutation , Oogonia/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA-Dependent RNA Polymerase/metabolism , Ribosomes/metabolism
6.
Gene ; 788: 145583, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33753150

ABSTRACT

Macrobrachium nipponense has the characteristics of fast ovarian development cycle, which leads to the coexistence of multiple generations, the reduction of commodity specifications and the low economic benefit. Therefore, the study on the mechanism of ovarian development is of great significance to the development of industry. Cyclin A (CycA)is a key gene regulating ovarian development in vertebrates, but little information was available for its function in crustaceans. In this study, the full-length cDNA of Mn-CycA was obtained from the ovary. The full-length cDNA (2033 bp) with an open reading frame of 1368 bp, encoded a 456-amino acid protein. qRT-PCR revealed tissue-specific expression pattern of Mn-CycA, with abundant expression in the ovary. Results in different developmental stages of ovary indicated that Mn-CycA expression is positively correlated with ovarian maturation. qRT-PCR In different developmental stages, the expression of Mn-CycA mRNA gradually increased during the embryonic stage and decreased significantly on the first day of the hatching stage. At the 25th day of the metamorphosis stage, the expression level of Mn-CycAmRNA in female shrimp was 3.5 times higher than that in male shrimp, which may be related to the proliferation of oogonia and the formation of oocytes. In situ hybridization (ISH) of ovary showed Mn-CycA was examined in all stages and was mainly located in oogonia and oocytes. Compared with the control group, the obvious change of gonad somatic index (GSI) proved that injection of Mn-CycA dsRNA could delay the ovarian development cycle, which provided strong evidence for the involvement of Mn-CycA in ovarian maturation and oogenesis, and expanded a new perspective for studying the fast ovarian development cycle in M. nipponense.


Subject(s)
Cyclin A/genetics , Cyclin A/metabolism , Gene Expression Profiling/methods , Palaemonidae/growth & development , Animals , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Base Sequence , Cloning, Molecular , Female , Gene Expression Regulation, Developmental , Male , Oocytes/growth & development , Oocytes/metabolism , Oogonia/growth & development , Oogonia/metabolism , Open Reading Frames , Organ Specificity , Palaemonidae/genetics , Palaemonidae/metabolism , Phylogeny
7.
Biochem Biophys Res Commun ; 535: 6-11, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33340766

ABSTRACT

No effective cryopreservation technique exists for fish eggs and embryos; thus, the cryopreservation of germ cells (spermatogonia or oogonia) and subsequent generation of eggs and sperm would be an alternative solution for the long-term preservation of piscine genetic resources. Nevertheless, in our previous study using rainbow trout, we showed that recipients transplanted with XY spermatogonia or XX oogonia produced unnatural sex-biased F1 offspring. To overcome these obstacles, we transplanted immature germ cells (XX oogonia or XY spermatogonia; frozen for 33 days) into the body cavities of triploid hatchlings, and the transplanted germ cells possessed a high capacity for differentiating into eggs and sperm in the ovaries and testes of recipients. Approximately 30% of triploid recipients receiving frozen germ cells generated normal salmon that displayed the donor-derived black body color phenotype, although all triploid salmon not receiving transplants were functionally sterile. Furthermore, F1 offspring obtained from insemination of the oogonia-derived eggs and spermatogonia-derived sperm show a normal sex ratio of 1:1 (female:male). Thus, this method presented a critical technique for practical conservation projects for other teleost fish species and masu salmon.


Subject(s)
Cryopreservation/methods , Oncorhynchus/growth & development , Oogonia/cytology , Oogonia/transplantation , Ovum/cytology , Spermatogonia/cytology , Spermatogonia/transplantation , Spermatozoa/cytology , Aging , Animals , Cell Differentiation , Conservation of Natural Resources/methods , Female , Germ Cells , Male , Oncorhynchus/embryology , Oogonia/metabolism , Ovum/metabolism , Sex Ratio , Spermatogonia/metabolism , Spermatozoa/metabolism , Triploidy
8.
Cells ; 9(11)2020 10 31.
Article in English | MEDLINE | ID: mdl-33142835

ABSTRACT

In vertebrates, the carbohydrate polymer polysialic acid (polySia) is especially well known for its essential role during neuronal development, regulating the migration and proliferation of neural precursor cells, for instance. Nevertheless, sialic acid polymers seem to be regulatory elements in other physiological systems, such as the reproductive tract. Interestingly, trout fish eggs have polySia, but we know little of its cellular distribution and role during oogenesis. Therefore, we localized α2,8-linked N-acetylneuraminic acid polymers in the ovaries of Coregonus maraena by immunohistochemistry and found that prevalent clusters of oogonia showed polySia signals on their surfaces. Remarkably, the genome of this salmonid fish contains two st8sia2 genes and one st8sia4 gene, that is, three polysialyltransferases. The expression analysis revealed that for st8sia2-r2, 60 times more mRNA was present than st8sia2-r1 and st8sia4. To compare polysialylation status regarding various polySiaT configurations, we performed a comparable analysis in Sander lucioperca. The genome of this perciform fish contains only one st8sia2 and no st8sia4 gene. Here, too, clusters of oogonia showed polysialylated cell surfaces, and we detected high mRNA values for st8sia2. These results suggest that in teleosts, polySia is involved in the cellular processes of oogonia during oogenesis.


Subject(s)
Ovary , Perches/genetics , Salmonidae/genetics , Sialic Acids/metabolism , Sialyltransferases/genetics , Animals , Female , Histological Techniques , N-Acetylneuraminic Acid/metabolism , Oogenesis , Oogonia/metabolism
9.
PLoS Genet ; 16(11): e1009067, 2020 11.
Article in English | MEDLINE | ID: mdl-33206637

ABSTRACT

Mammalian oogonia proliferate without completing cytokinesis, forming cysts. Within these, oocytes differentiate and initiate meiosis, promoting double-strand break (DSBs) formation, which are repaired by homologous recombination (HR) causing the pairing and synapsis of the homologs. Errors in these processes activate checkpoint mechanisms, leading to apoptosis. At the end of prophase I, in contrast with what is observed in spermatocytes, oocytes accumulate unrepaired DSBs. Simultaneously to the cyst breakdown, there is a massive oocyte death, which has been proposed to be necessary to enable the individualization of the oocytes to form follicles. Based upon all the above-mentioned information, we hypothesize that the apparently inefficient HR occurring in the oocytes may be a requirement to first eliminate most of the oocytes and enable cyst breakdown and follicle formation. To test this idea, we compared perinatal ovaries from control and mutant mice for the effector kinase of the DNA Damage Response (DDR), CHK2. We found that CHK2 is required to eliminate ~50% of the fetal oocyte population. Nevertheless, the number of oocytes and follicles found in Chk2-mutant ovaries three days after birth was equivalent to that of the controls. These data revealed the existence of another mechanism capable of eliminating oocytes. In vitro inhibition of CHK1 rescued the oocyte number in Chk2-/- mice, implying that CHK1 regulates postnatal oocyte death. Moreover, we found that CHK1 and CHK2 functions are required for the timely breakdown of the cyst and to form follicles. Thus, we uncovered a novel CHK1 function in regulating the oocyte population in mice. Based upon these data, we propose that the CHK1- and CHK2-dependent DDR controls the number of oocytes and is required to properly break down oocyte cysts and form follicles in mammals.


Subject(s)
DNA Damage/genetics , Oogonia/metabolism , Ovarian Follicle/metabolism , Animals , Apoptosis/physiology , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Cysts/metabolism , DNA Damage/physiology , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Female , Meiosis/physiology , Meiotic Prophase I/physiology , Mice , Mice, Inbred C57BL , Oocytes/metabolism , Oocytes/physiology , Oogonia/physiology , Ovarian Follicle/physiology , Ovary/metabolism , Progesterone/metabolism
10.
PLoS Genet ; 16(1): e1008529, 2020 01.
Article in English | MEDLINE | ID: mdl-31917788

ABSTRACT

Exposure to diethylhexyl phthalate (DEHP), the most abundant plasticizer used in the production of polyvinyl-containing plastics, has been associated to adverse reproductive health outcomes in both males and females. While the effects of DEHP on reproductive health have been widely investigated, the molecular mechanisms by which exposure to environmentally-relevant levels of DEHP and its metabolites impact the female germline in the context of a multicellular organism have remained elusive. Using the Caenorhabditis elegans germline as a model for studying reprotoxicity, we show that exposure to environmentally-relevant levels of DEHP and its metabolites results in increased meiotic double-strand breaks (DSBs), altered DSB repair progression, activation of p53/CEP-1-dependent germ cell apoptosis, defects in chromosome remodeling at late prophase I, aberrant chromosome morphology in diakinesis oocytes, increased chromosome non-disjunction and defects during early embryogenesis. Exposure to DEHP results in a subset of nuclei held in a DSB permissive state in mid to late pachytene that exhibit defects in crossover (CO) designation/formation. In addition, these nuclei show reduced Polo-like kinase-1/2 (PLK-1/2)-dependent phosphorylation of SYP-4, a synaptonemal complex (SC) protein. Moreover, DEHP exposure leads to germline-specific change in the expression of prmt-5, which encodes for an arginine methyltransferase, and both increased SC length and altered CO designation levels on the X chromosome. Taken together, our data suggest a model by which impairment of a PLK-1/2-dependent negative feedback loop set in place to shut down meiotic DSBs, together with alterations in chromosome structure, contribute to the formation of an excess number of DSBs and altered CO designation levels, leading to genomic instability.


Subject(s)
Crossing Over, Genetic , DNA Breaks, Double-Stranded , Diethylhexyl Phthalate/toxicity , Oogenesis , Oogonia/drug effects , Plasticizers/toxicity , Animals , Apoptosis , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Genomic Instability , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oogonia/cytology , Oogonia/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
G3 (Bethesda) ; 10(1): 165-175, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31685524

ABSTRACT

Immune priming occurs when a past infection experience leads to a more effective immune response upon a secondary exposure to the infection or pathogen. In some instances, parents are able to transmit immune priming to their offspring, creating a subsequent generation with a superior immune capability, through processes that are not yet fully understood. Using a parasitoid wasp, which infects larval stages of Drosophila melanogaster, we describe an example of an intergenerational inheritance of immune priming. This phenomenon is anticipatory in nature and does not rely on parental infection, but rather, when adult fruit flies are cohabitated with a parasitic wasp, they produce offspring that are more capable of mounting a successful immune response against a parasitic macro-infection. This increase in offspring survival correlates with a more rapid induction of lamellocytes, a specialized immune cell. RNA-sequencing of the female germline identifies several differentially expressed genes following wasp exposure, including the peptiodoglycan recognition protein-LB (PGRP-LB). We find that genetic manipulation of maternal PGRP-LB identifies this gene as a key element in this intergenerational phenotype.


Subject(s)
Disease Resistance/genetics , Drosophila melanogaster/genetics , Maternal Inheritance , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/parasitology , Female , Oogonia/metabolism , Wasps/pathogenicity
13.
Genetics ; 213(3): 911-922, 2019 11.
Article in English | MEDLINE | ID: mdl-31484689

ABSTRACT

The conserved RNA helicase Vasa is required for germ cell development in many organisms. In Drosophila melanogaster loss of PIWI-interacting RNA pathway components, including Vasa, causes Chk2-dependent oogenesis arrest. However, whether the arrest is due to Chk2 signaling at a specific stage and whether continuous Chk2 signaling is required for the arrest is unknown. Here, we show that absence of Vasa during the germarial stages causes Chk2-dependent oogenesis arrest. Additionally, we report the age-dependent decline of the ovariole number both in flies lacking Vasa expression only in the germarium and in loss-of-function vasa mutant flies. We show that Chk2 activation exclusively in the germarium is sufficient to interrupt oogenesis and to reduce ovariole number in aging flies. Once induced in the germarium, Chk2-mediated arrest of germ cell development cannot be overcome by restoration of Vasa or by downregulation of Chk2 in the arrested egg chambers. These findings, together with the identity of Vasa-associated proteins identified in this study, demonstrate an essential role of the helicase in the germ cell lineage maintenance and indicate a function of Vasa in germline stem cell homeostasis.


Subject(s)
DEAD-box RNA Helicases/metabolism , Drosophila Proteins/metabolism , Homeostasis , Oogenesis , Animals , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DEAD-box RNA Helicases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Loss of Function Mutation , Oogonia/cytology , Oogonia/metabolism
14.
Gene ; 701: 104-112, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30905810

ABSTRACT

PIWI family member piwil1, which associates with Piwi-interacting RNA (piRNA), is responsible in regulation of germ cell differentiation and maintenance of reproductive stem cells. In this study, we analyzed the piwil1 gene in Paralichthys olivaceus. Bioinformatics analysis and structure prediction showed that piwil1 had the conserved domains: PAZ domain and PIWI domain. Expression analysis during embryonic development implied that piwil1 gene was maternally inherited. The tissue distribution showed a sexually dimorphic gene expression pattern, with higher expression level in testis than ovary. In situ hybridization results demonstrated that piwil1 was predominantly distributed in oogonia, oocytes, sertoli cells and spermatocytes. A CpG island was predicted in the 5'-flanking region of piwil1 gene, and its methylation levels showed significant disparity between males and females, indicating that the sexually dimorphic expression of piwil1 gene might be regulated by methylation. Furthermore, we explored the distinct roles of human chorionic gonadotropin and 17α-methyltestosterone in regulating the expression of piwil1, and found that piwil1 was interacting with the HPG axis hormones. These results indicated that piwil1 might play a crucial role in gonadal development and gametogenesis in Paralichthys olivaceus.


Subject(s)
Argonaute Proteins/biosynthesis , Fish Proteins/biosynthesis , Flounder/growth & development , Gene Expression Regulation , Oogenesis/physiology , Spermatogenesis/physiology , Animals , Argonaute Proteins/genetics , Female , Fish Proteins/genetics , Flounder/genetics , Male , Oocytes/cytology , Oocytes/metabolism , Oogonia/cytology , Oogonia/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism
15.
Sci Rep ; 8(1): 12791, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143724

ABSTRACT

Oocytes, including from mammals, lack centrioles, but neither the mechanism by which mature eggs lose their centrioles nor the exact stage at which centrioles are destroyed during oogenesis is known. To answer questions raised by centriole disappearance during oogenesis, using a transgenic mouse expressing GFP-centrin-2 (GFP CETN2), we traced their presence from e11.5 primordial germ cells (PGCs) through oogenesis and their ultimate dissolution in mature oocytes. We show tightly coupled CETN2 doublets in PGCs, oogonia, and pre-pubertal oocytes. Beginning with follicular recruitment of incompetent germinal vesicle (GV) oocytes, through full oocyte maturation, the CETN2 doublets separate within the pericentriolar material (PCM) and a rise in single CETN2 pairs is identified, mostly at meiotic metaphase-I and -II spindle poles. Partial CETN2 foci dissolution occurs even as other centriole markers, like Cep135, a protein necessary for centriole duplication, are maintained at the PCM. Furthermore, live imaging demonstrates that the link between the two centrioles breaks as meiosis resumes and that centriole association with the PCM is progressively lost. Microtubule inhibition shows that centriole dissolution is uncoupled from microtubule dynamics. Thus, centriole doublets, present in early G2-arrested meiotic prophase oocytes, begin partial reduction during follicular recruitment and meiotic resumption, later than previously thought.


Subject(s)
Centrioles/metabolism , Germ Cells/metabolism , Oocytes/metabolism , Animals , Calcium-Binding Proteins/metabolism , Centrioles/drug effects , Centrosome/drug effects , Centrosome/metabolism , Female , Germ Cells/cytology , Germ Cells/drug effects , Green Fluorescent Proteins/metabolism , Metaphase/drug effects , Mice , Microtubules/drug effects , Microtubules/metabolism , Nocodazole/pharmacology , Oocytes/cytology , Oocytes/drug effects , Oogonia/cytology , Oogonia/drug effects , Oogonia/metabolism , Ovary/embryology , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Spindle Poles/drug effects , Spindle Poles/metabolism , Tubulin/metabolism
16.
G3 (Bethesda) ; 8(8): 2749-2756, 2018 07 31.
Article in English | MEDLINE | ID: mdl-29934375

ABSTRACT

Screening for successful CRISPR/Cas9 editing events remains a time consuming technical bottleneck in the field of Drosophila genome editing. This step can be particularly laborious for events that do not cause a visible phenotype, or those which occur at relatively low frequency. A promising strategy to enrich for desired CRISPR events is to co-select for an independent CRISPR event that produces an easily detectable phenotype. Here, we describe a simple negative co-selection strategy involving CRISPR-editing of a dominant female sterile allele, ovoD1 In this system ("ovoD co-selection"), the only functional germ cells in injected females are those that have been edited at the ovoD1 locus, and thus all offspring of these flies have undergone editing of at least one locus. We demonstrate that ovoD co-selection can be used to enrich for knock-out mutagenesis via nonhomologous end-joining (NHEJ), and for knock-in alleles via homology-directed repair (HDR). Altogether, our results demonstrate that ovoD co-selection reduces the amount of screening necessary to isolate desired CRISPR events in Drosophila.


Subject(s)
CRISPR-Cas Systems , Drosophila/genetics , Gene Targeting/methods , Selection, Genetic , Alleles , Animals , Female , Gene Editing/methods , Oogonia/metabolism , Phenotype
17.
Gene ; 673: 61-69, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-29920365

ABSTRACT

Tudor domain containing protein 1 (tdrd1) is a member of the Tudor family and has shown essential functions during embryogenesis and gametogenesis. In this study, we cloned the full length cDNA of Paralichthys olivaceus tdrd1 (Potdrd1). PoTDRD1 is a multidomain protein with an N-terminal MYND zinc finger domain, followed by four tandem extended Tudor domains. Sequence comparison, genomic structure, phylogenetic analyses and synteny analyses showed that Potdrd1 was homologous to those of other teleosts. In adult individuals, the expression of Potdrd1 was higher in testis than in ovary, demonstrating a sexually dimorphic gene expression pattern. In situ hybridization (ISH) showed that Potdrd1 mRNA was detected in oogonia and oocytes of ovary as well as in spermatogonia and spermatocytes of testis. In juveniles during gonad differentiation its expression level increased rapidly from 30 dph to 100 dph and showed obvious sexual dimorphism that was in accordance with the expression of anti-Mullerian hormone (amh). Potdrd1 mRNA was consistently detected during embryogenesis, and its level was higher from unfertilzed eggs to the blastula stage and subsequently decreased until hatching. When chimeric RNA containing green fluorescent protein (GFP) and 3' untranslated regions (UTR) of Potdrd1 was microinjected into zebrafish fertilized eggs, the green fluorescence could be visualized only in putative PGCs. These results indicated that Potdrd1 is a germline specific and sexually dimorphic factor that potentially functionate in germline development and gametogenesis in Japanese flounder.


Subject(s)
Carrier Proteins/genetics , Fish Proteins/genetics , Flounder/genetics , Germ-Line Mutation , 3' Untranslated Regions , Animals , Carrier Proteins/metabolism , Cloning, Molecular , Female , Fish Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Germ Cells/cytology , Green Fluorescent Proteins/metabolism , In Situ Hybridization , Male , Oocytes/cytology , Oocytes/metabolism , Oogonia/metabolism , Ovary/metabolism , Phylogeny , Protein Domains , Sex Characteristics , Spermatocytes/metabolism , Spermatogonia/metabolism , Testis/metabolism
18.
Biol Reprod ; 98(4): 532-542, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29329412

ABSTRACT

Following proliferation of oogonia in mammals, great numbers of germ cells are discarded, primarily by apoptosis, while the remainder form primordial follicles (the ovarian reserve) that determine fertility and reproductive lifespan. More massive, rapid, and essentially total loss of oocytes, however, occurs when the transcription factor Lhx8 is ablated-though the cause and mechanism of germ cell loss from the Lhx8-/- ovaries has been unknown. We found that Lhx8-/- ovaries maintain the same number of germ cells throughout embryonic development; rapid decrease in the pool of oocytes starts shortly before birth. The loss results from activation of autophagy, which becomes overwhelming within the first postnatal week, with extracellular matrix proteins filling the space previously occupied by follicles to produce a fibrotic ovary. Associated with this process, as early as a few days before birth, Lhx8-/- oocytes failed to repair DNA damage-which normally occurs when meiosis is initiated during embryonic development; and DNA damage repair genes were downregulated throughout the oocyte short lifespan. Based on gene expression analyses and morphological changes, we propose a model in which lineage-restricted failure of DNA repair triggers germ cell autophagy, causing premature depletion of the ovarian reserve in Lhx8-/- mice.


Subject(s)
Autophagy/physiology , DNA Damage/physiology , LIM-Homeodomain Proteins/metabolism , Oocytes/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/physiology , Female , LIM-Homeodomain Proteins/genetics , Meiosis , Mice , Mice, Knockout , Oogenesis/physiology , Oogonia/metabolism , Ovarian Reserve/physiology , Transcription Factors/genetics
19.
Gene ; 648: 68-75, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29331483

ABSTRACT

GATA4 is a well-known transcription factor of the GATA family implicated in regulation of sex determination and gonadal development in mammals. In this study, we cloned the full-length cDNA of Paralichthys olivaceus gata4 (Po-gata4). Phylogenetic, gene structure, and synteny analysis showed that Po-GATA4 is homologous to GATA4 of teleost and tetrapod. Po-gata4 transcripts were detected in Sertoli cells, spermatogonia, oogonia and oocytes, with higher transcript levels overall in the testis than the ovary. The promoter region of P. olivaceus R-spondin1was found to contain a GATA4-binding motif. Results of CBA (cleaved amplified polymorphic sequence-based binding assay) indicated that GATA4 could indeed bind to the promoter sequence of R-spondin1. Moreover, human GATA4 recombinant protein could upregulate R-spondin1 in P. olivaceus ovary cells and FBCs (flounder brain cell line). In FBCs, overexpression of Po-gata4 resulted in elevated transcript levels of R-spondin1. Taken together, our results indicate that Po-GATA4 is involved in gonadal development by regulating R-spondin1 expression.


Subject(s)
Embryo, Nonmammalian/metabolism , Fish Proteins/genetics , Flounder/genetics , GATA4 Transcription Factor/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Female , Fish Proteins/classification , Fish Proteins/metabolism , Flounder/embryology , Flounder/metabolism , GATA4 Transcription Factor/classification , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Male , Oogonia/cytology , Oogonia/metabolism , Phylogeny , Promoter Regions, Genetic/genetics , Protein Binding , Spermatogonia/cytology , Spermatogonia/metabolism
20.
PLoS Genet ; 13(5): e1006790, 2017 May.
Article in English | MEDLINE | ID: mdl-28542174

ABSTRACT

Germline stem cells in the Drosophila ovary are maintained by a somatic niche. The niche is structurally and functionally complex and contains four cell types, the escort, cap, and terminal filament cells and the newly identified transition cell. We find that the large Maf transcription factor Traffic jam (Tj) is essential for determining niche cell fates and architecture, enabling each niche in the ovary to support a normal complement of 2-3 germline stem cells. In particular, we focused on the question of how cap cells form. Cap cells express Tj and are considered the key component of a mature germline stem cell niche. We conclude that Tj controls the specification of cap cells, as the complete loss of Tj function caused the development of additional terminal filament cells at the expense of cap cells, and terminal filament cells developed cap cell characteristics when induced to express Tj. Further, we propose that Tj controls the morphogenetic behavior of cap cells as they adopted the shape and spatial organization of terminal filament cells but otherwise appeared to retain their fate when Tj expression was only partially reduced. Our data indicate that Tj contributes to the establishment of germline stem cells by promoting the cap cell fate, and controls the stem cell-carrying capacity of the niche by regulating niche architecture. Analysis of the interactions between Tj and the Notch (N) pathway indicates that Tj and N have distinct functions in the cap cell specification program. We propose that formation of cap cells depends on the combined activities of Tj and the N pathway, with Tj promoting the cap cell fate by blocking the terminal filament cell fate, and N supporting cap cells by preventing the escort cell fate and/or controlling the number of cap cell precursors.


Subject(s)
Drosophila Proteins/genetics , Maf Transcription Factors, Large/genetics , Ovary/cytology , Proto-Oncogene Proteins/genetics , Stem Cell Niche , Animals , Drosophila/cytology , Drosophila/genetics , Drosophila Proteins/metabolism , Female , Maf Transcription Factors, Large/metabolism , Oogonia/cytology , Oogonia/metabolism , Ovary/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...