Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
BMC Genomics ; 25(1): 500, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773374

ABSTRACT

BACKGROUND: The ricefield eel Monopterus albus undergoes a natural sex change from female to male during its life cycle, and previous studies have shown the potential mechanisms of this transition at the transcriptional and protein levels. However, the changes in protein levels have not been fully explored, especially in the intersexual stage. RESULTS: In the present study, the protein expression patterns in the gonadal tissues from five different periods, the ovary (OV), early intersexual stage gonad (IE), middle intersexual stage gonad (IM), late intersexual stage gonad (IL), and testis (TE), were determined by untargeted proteomics sequencing. A total of 5125 proteins and 394 differentially expressed proteins (DEPs) were detected in the gonadal tissues. Of the 394 DEPs, there were 136 between the OV and IE groups, 20 between the IM and IE groups, 179 between the IL and IM groups, and 59 between the TE and IL groups. Three candidate proteins, insulin-like growth factor 2 mRNA-binding protein 3 isoform X1 (Igf2bp3), triosephosphate isomerase (Tpi), and Cu-Zn superoxide dismutase isoform X1 [(Cu-Zn) Sod1], were validated by western blotting to verify the reliability of the data. Furthermore, metal metabolite-related proteins were enriched in the IL vs. IM groups and TE vs. IL groups, which had close relationships with sex change, including Cu2+-, Ca2+-, Zn2+- and Fe2+/Fe3+-related proteins. Analysis of the combined transcriptome data revealed consistent protein/mRNA expression trends for two metal metabolite-related proteins/genes [LOC109953912 and calcium Binding Protein 39 Like (cab39l)]. Notably, we detected significantly higher levels of Cu2+ during the sex change process, suggesting that Cu2+ is a male-related metal metabolite that may have an important function in male reproductive development. CONCLUSIONS: In summary, we analyzed the protein profiles of ricefield eel gonadal tissues in five sexual stages (OV, IE, IM, IL, and TE) and verified the plausibility of the data. After preforming the functional enrichment of metal metabolite-related DEPs, we detected the contents of the metal metabolites Zn2+, Cu2+, Ca2+, and Fe2+/Fe3+ at these five stages and screened for (Cu-Zn) Sod1 and Mmp-9 as possible key proteins in the sex reversal process.


Subject(s)
Metals , Animals , Male , Female , Metals/metabolism , Eels/metabolism , Eels/genetics , Proteomics , Fish Proteins/metabolism , Fish Proteins/genetics , Smegmamorpha/metabolism , Smegmamorpha/genetics , Hermaphroditic Organisms/metabolism , Hermaphroditic Organisms/genetics , Gene Expression Profiling , Testis/metabolism
2.
Sci Rep ; 11(1): 22881, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819550

ABSTRACT

The stunning sexual transformation commonly triggered by age, size or social context in some fishes is one of the best examples of phenotypic plasticity thus far described. To date our understanding of this process is dominated by studies on a handful of subtropical and tropical teleosts, often in wild settings. Here we have established the protogynous New Zealand spotty wrasse, Notolabrus celidotus, as a temperate model for the experimental investigation of sex change. Captive fish were induced to change sex using aromatase inhibition or manipulation of social groups. Complete female-to-male transition occurred over 60 days in both cases and time-series sampling was used to quantify changes in hormone production, gene expression and gonadal cellular anatomy. Early-stage decreases in plasma 17ß-estradiol (E2) concentrations or gonadal aromatase (cyp19a1a) expression were not detected in spotty wrasse, despite these being commonly associated with the onset of sex change in subtropical and tropical protogynous (female-to-male) hermaphrodites. In contrast, expression of the masculinising factor amh (anti-Müllerian hormone) increased during early sex change, implying a potential role as a proximate trigger for masculinisation. Collectively, these data provide a foundation for the spotty wrasse as a temperate teleost model to study sex change and cell fate in vertebrates.


Subject(s)
Fishes/physiology , Hermaphroditic Organisms/physiology , Sex Determination Processes , Animals , Anti-Mullerian Hormone/genetics , Anti-Mullerian Hormone/metabolism , Aromatase Inhibitors/pharmacology , Estradiol/blood , Female , Fish Proteins/genetics , Fish Proteins/metabolism , Fishes/blood , Fishes/genetics , Gene Expression Regulation , Gonads/physiology , Hermaphroditic Organisms/drug effects , Hermaphroditic Organisms/genetics , Hermaphroditic Organisms/metabolism , Male , Models, Animal , Phenotype , Sex Characteristics , Sex Determination Processes/drug effects , Social Behavior , Testosterone/analogs & derivatives , Testosterone/blood
3.
BMC Plant Biol ; 21(1): 468, 2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34645403

ABSTRACT

BACKGROUND: The fragrant flower plant Osmanthus fragrans has an extremely rare androdioecious breeding system displaying the occurrence of males and hermaphrodites in a single population, which occupies a crucial intermediate stage in the evolutionary transition between hermaphroditism and dioecy. However, the molecular mechanism of androdioecy plant is very limited and still largely unknown. RESULTS: Here, we used SWATH-MS-based quantitative approach to study the proteome changes between male and hermaphroditic O. fragrans pistils. A total of 428 proteins of diverse functions were determined to show significant abundance changes including 210 up-regulated and 218 down-regulated proteins in male compared to hermaphroditic pistils. Functional categorization revealed that the differentially expressed proteins (DEPs) primarily distributed in the carbohydrate metabolism, secondary metabolism as well as signaling cascades. Further experimental analysis showed the substantial carbohydrates accumulation associated with promoted net photosynthetic rate and water use efficiency were observed in purplish red pedicel of hermaphroditic flower compared with green pedicel of male flower, implicating glucose metabolism serves as nutritional modulator for the differentiation of male and hermaphroditic flower. Meanwhile, the entire upregulation of secondary metabolism including flavonoids, isoprenoids and lignins seem to protect and maintain the male function in male flowers, well explaining important feature of androdioecy that aborted pistil of a male flower still has a male function. Furthermore, nine selected DEPs were validated via gene expression analysis, suggesting an extra layer of post-transcriptional regulation occurs during O. fragrans floral development. CONCLUSION: Taken together, our findings represent the first SWATH-MS-based proteomic report in androdioecy plant O. fragrans, which reveal carbohydrate metabolism, secondary metabolism and post-transcriptional regulation contributing to the androdioecy breeding system and ultimately extend our understanding on genetic basis as well as the industrialization development of O. fragrans.


Subject(s)
Carbohydrate Metabolism/genetics , Flowers/growth & development , Flowers/genetics , Oleaceae/growth & development , Oleaceae/genetics , Oleaceae/metabolism , Reproduction/genetics , Reproduction/physiology , Biological Evolution , China , Gene Expression Regulation, Plant , Genetic Variation , Hermaphroditic Organisms/genetics , Hermaphroditic Organisms/growth & development , Hermaphroditic Organisms/metabolism , Phenotype , Proteomics
4.
Dev Biol ; 478: 122-132, 2021 10.
Article in English | MEDLINE | ID: mdl-34224682

ABSTRACT

Sexual systems are surprisingly diverse, considering the ubiquity of sexual reproduction. Sequential hermaphroditism, the ability of an individual to change sex, has emerged multiple times independently across the animal kingdom. In molluscs, repeated shifts between ancestrally separate sexes and hermaphroditism are generally found at the level of family and above, suggesting recruitment of deeply conserved mechanisms. Despite this, molecular mechanisms of sexual development are poorly known. In molluscs with separate sexes, endocrine disrupting toxins bind the retinoid X receptor (RXR), activating ectopic male development in females, suggesting the retinoid pathway as a candidate controlling sexual transitions in sequential hermaphrodites. We therefore tested the role of retinoic acid signaling in sequentially hermaphroditic Crepidula snails, which develop first into males, then change sex, maturing into females. We show that retinoid agonists induce precocious penis growth in juveniles and superimposition of male development in females. Combining RXR antagonists with retinoid agonists significantly reduces penis length in induced juveniles, while similar treatments using retinoic acid receptor (RAR) antagonists increase penis length. Transcripts of both receptors are expressed in the induced penis. Our findings therefore show that retinoid signaling can initiate molluscan male genital development, and regulate penis length. Further, we show that retinoids induce ectopic male development in multiple Crepidula species. Species-specific influence of conspecific induction of sexual transitions correlates with responsiveness to retinoids. We propose that retinoid signaling plays a conserved role in molluscan male development, and that shifts in the timing of retinoid signaling may have been important for the origins of sequential hermaphroditism within molluscs.


Subject(s)
Hermaphroditic Organisms/growth & development , Retinoids/metabolism , Snails/growth & development , Snails/metabolism , Animals , Cytochrome P450 Family 26/genetics , Female , Hermaphroditic Organisms/genetics , Hermaphroditic Organisms/metabolism , Male , Penis/growth & development , Penis/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Signal Transduction , Snails/anatomy & histology , Snails/genetics , Species Specificity , Tretinoin/metabolism , Trialkyltin Compounds/pharmacology
5.
Dev Biol ; 470: 10-20, 2021 02.
Article in English | MEDLINE | ID: mdl-33160939

ABSTRACT

VAMP/synaptobrevin-associated protein B (VAP-B) is a type II ER membrane protein, but its N-terminal MSP domain (MSPd) can be cleaved and secreted. Mutations preventing the cleavage and secretion of MSPd have been implicated in cases of human neurodegenerative diseases. The site of VAP cleavage and the tissues capable in releasing the processed MSPd are not understood. In this study, we analyze the C. elegans VAP-B homolog, VPR-1, for its processing and secretion from the intestine. We show that intestine-specific expression of an N-terminally FLAG-tagged VPR-1 rescues underdeveloped gonad and sterility defects in vpr-1 null hermaphrodites. Immunofluorescence studies reveal that the tagged intestinal expressed VPR-1 is present at the distal gonad. Mass spectrometry analysis of a smaller product of the N-terminally tagged VPR-1 identifies a specific cleavage site at Leu156. Mutation of the leucine results in loss of gonadal MSPd signal and reduced activity of the mutant VPR-1. Thus, we report for the first time the cleavage site of VPR-1 and provide direct evidence that intestinally expressed VPR-1 can be released and signal in the distal gonad. These results establish the foundation for further exploration of VAP cleavage, MSPd secretion, and non-cell-autonomous signaling in development and diseases.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Helminth Proteins/metabolism , Membrane Proteins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Endoplasmic Reticulum/metabolism , Genes, Helminth , Gonads/chemistry , Gonads/growth & development , Gonads/metabolism , Helminth Proteins/chemistry , Hermaphroditic Organisms/genetics , Hermaphroditic Organisms/metabolism , Hermaphroditic Organisms/physiology , Infertility , Intestines/cytology , Intestines/physiology , Leucine/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Phenotype , Point Mutation , Protein Domains , Protein Processing, Post-Translational
6.
Biomolecules ; 10(12)2020 12 10.
Article in English | MEDLINE | ID: mdl-33321846

ABSTRACT

In Caenorhabditis elegans, gap junctions couple cells of the somatic gonad with the germline to support germ cell proliferation and gametogenesis. A strong loss-of-function mutation (T239I) affects the second extracellular loop (EL2) of the somatic INX-8 hemichannel subunit. These mutant hemichannels form non-functional gap junctions with germline-expressed innexins. We conducted a genetic screen for suppressor mutations that restore germ cell proliferation in the T239I mutant background and isolated seven intragenic mutations, located in diverse domains of INX-8 but not the EL domains. These second-site mutations compensate for the original channel defect to varying degrees, from nearly complete wild-type rescue, to partial rescue of germline proliferation. One suppressor mutation (E350K) supports the innexin cryo-EM structural model that the channel pore opening is surrounded by a cytoplasmic dome. Two suppressor mutations (S9L and I36N) may form leaky channels that support germline proliferation but cause the demise of somatic sheath cells. Phenotypic analyses of three of the suppressors reveal an equivalency in the rescue of germline proliferation and comparable delays in gametogenesis but a graded rescue of fertility. The mutations described here may be useful for elucidating the biochemical pathways that produce the active biomolecules transiting through soma-germline gap junctions.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Connexins/genetics , Gametogenesis/genetics , Hermaphroditic Organisms/genetics , Mutation , Amino Acid Sequence , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Cell Proliferation , Connexins/chemistry , Connexins/metabolism , Fertility/genetics , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Gonads/cytology , Gonads/metabolism , Hermaphroditic Organisms/cytology , Hermaphroditic Organisms/metabolism , Male , Oocytes/cytology , Oocytes/metabolism , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Sequence Alignment , Spermatozoa/cytology , Spermatozoa/metabolism , Structural Homology, Protein
7.
Biol Reprod ; 103(4): 817-827, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32582944

ABSTRACT

To date, the molecular mechanisms of the unique gonadal development mode known as protandric simultaneous hermaphroditism (PSH) are unclear in crustaceans. In this study, cDNA of a gonad-inhibiting hormone (Lv-GIH1) was isolated from the PSH peppermint shrimp Lysmata vittata, and its expression was exclusively found in the eyestalk ganglion. Real-time quantitative polymerase chain reaction (qRT-PCR) revealed that the expression of Lv-GIH1 increased during gonadal development of the functional male stages but decreased significantly at subsequent simultaneous hermaphroditism stage. Further in vitro experiment showed that recombinant GIH1 protein (rGIH1) effectively inhibited Vg expression in the cultured hepatopancreas tissues while the short-term injection of GIH1-dsRNA resulted in reduced expression of Lv-GIH1 and upregulated expression of Vg in the hepatopancreas. Moreover, long-term rGIH1 injection led to significantly reduced expression of Lv-Vg, Lv-VgR, and Lv-CFSH1, subdued growth of oocytes, and feathery setae as a secondary sexual characteristic in females. Interestingly, while germ cells in testicular part were suppressed by rGIH1 injection, the expression of Lv-IAGs showed no significant difference; and long-term GIH1-dsRNA injection results were contrary to those of rGIH1 injection. Taken together, the results of this study indicate that Lv-GIH1 is involved in gonadal development and might also participate in controlling secondary sexual characteristic development in L. vittata by inhibiting Lv-CFSH1 expression.


Subject(s)
Decapoda/physiology , Gene Expression Regulation/physiology , Hermaphroditic Organisms/metabolism , Invertebrate Hormones/metabolism , Animals , Cloning, Molecular , Decapoda/growth & development , Gene Knockdown Techniques , Gonads/growth & development , Hepatopancreas/drug effects , Hepatopancreas/metabolism , Invertebrate Hormones/pharmacology , Phylogeny , RNA/genetics , RNA/metabolism , Sex Differentiation
8.
Dev Biol ; 446(2): 193-205, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30599151

ABSTRACT

Proper germ cell sex determination in Caenorhabditis nematodes requires a network of RNA-binding proteins (RBPs) and their target mRNAs. In some species, changes in this network enabled limited XX spermatogenesis, and thus self-fertility. In C. elegans, one of these selfing species, the global sex-determining gene tra-2 is regulated in germ cells by a conserved RBP, GLD-1, via the 3' untranslated region (3'UTR) of its transcript. A C. elegans-specific GLD-1 cofactor, FOG-2, is also required for hermaphrodite sperm fate, but how it modifies GLD-1 function is unknown. Germline feminization in gld-1 and fog-2 null mutants has been interpreted as due to cell-autonomous elevation of TRA-2 translation. Consistent with the proposed role of FOG-2 in translational control, the abundance of nearly all GLD-1 target mRNAs (including tra-2) is unchanged in fog-2 mutants. Epitope tagging reveals abundant TRA-2 expression in somatic tissues, but an undetectably low level in wild-type germ cells. Loss of gld-1 function elevates germline TRA-2 expression to detectable levels, but loss of fog-2 function does not. A simple quantitative model of tra-2 activity constrained by these results can successfully sort genotypes into normal or feminized groups. Surprisingly, fog-2 and gld-1 activity enable the sperm fate even when GLD-1 cannot bind to the tra-2 3' UTR. This suggests the GLD-1-FOG-2 complex regulates uncharacterized sites within tra-2, or other mRNA targets. Finally, we quantify the RNA-binding capacities of dominant missense alleles of GLD-1 that act genetically as "hyper-repressors" of tra-2 activity. These variants bind RNA more weakly in vitro than does wild-type GLD-1. These results indicate that gld-1 and fog-2 regulate germline sex via multiple interactions, and that our understanding of the control and evolution of germ cell sex determination in the C. elegans hermaphrodite is far from complete.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Hermaphroditic Organisms/genetics , Transcription Factors/genetics , 3' Untranslated Regions/genetics , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , Female , Gene Expression Profiling , Hermaphroditic Organisms/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Genetic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism
9.
Dev Cell ; 47(6): 801-813.e6, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30416013

ABSTRACT

To advance our understanding of the genetic programs that drive cell and tissue specialization, it is necessary to obtain a comprehensive overview of gene expression patterns. Here, we have used spatial transcriptomics to generate high-resolution, anteroposterior gene expression maps of C. elegans males and hermaphrodites. To explore these maps, we have developed computational methods for discovering region- and tissue-specific genes. We have found extensive sex-specific gene expression differences in the germline and sperm and discovered genes that are specifically expressed in the male reproductive tract. These include a group of uncharacterized genes that encode small secreted proteins that are required for male fertility. We conclude that spatial gene expression maps provide a powerful resource for identifying tissue-specific gene functions in C. elegans. Importantly, we found that expression maps from different animals can be precisely aligned, enabling transcriptome-wide comparisons of gene expression patterns.


Subject(s)
Gene Expression Profiling/methods , Sex Characteristics , Sex Determination Processes/genetics , Transcriptome/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Differentiation , Disorders of Sex Development/genetics , Female , Gene Expression Regulation, Developmental/genetics , Germ Cells/metabolism , Gonads/metabolism , Hermaphroditic Organisms/metabolism , Male , Meiosis , Nuclear Proteins/metabolism , Ovary/metabolism , RNA, Messenger/genetics , Spatio-Temporal Analysis , Spermatozoa/metabolism , Transcription Factors/metabolism
10.
Int J Mol Sci ; 19(9)2018 Sep 10.
Article in English | MEDLINE | ID: mdl-30201859

ABSTRACT

One of the most studied phosphoinositides is phosphatidylinositol 4,5-bisphosphate (PIP2), which localizes to the plasma membrane, nuclear speckles, small foci in the nucleoplasm, and to the nucleolus in mammalian cells. Here, we show that PIP2 also localizes to the nucleus in prophase I, during the gametogenesis of C. elegans hermaphrodite. The depletion of PIP2 by type I PIP kinase (PPK-1) kinase RNA interference results in an altered chromosome structure and leads to various defects during meiotic progression. We observed a decreased brood size and aneuploidy in progeny, defects in synapsis, and crossover formation. The altered chromosome structure is reflected in the increased transcription activity of a tightly regulated process in prophase I. To elucidate the involvement of PIP2 in the processes during the C. elegans development, we identified the PIP2-binding partners, leucine-rich repeat (LRR-1) protein and proteasome subunit beta 4 (PBS-4), pointing to its involvement in the ubiquitin⁻proteasome pathway.


Subject(s)
Caenorhabditis elegans/growth & development , Cell Nucleus/metabolism , Gametogenesis , Phosphatidylinositol 4,5-Diphosphate/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Chromosomes/chemistry , Gene Expression Regulation, Developmental , Hermaphroditic Organisms/genetics , Hermaphroditic Organisms/growth & development , Hermaphroditic Organisms/metabolism , Leucine-Rich Repeat Proteins , Meiotic Prophase I , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , RNA Interference
11.
BMC Evol Biol ; 18(1): 81, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29848299

ABSTRACT

BACKGROUND: Along with sperm, in many taxa ejaculates also contain large numbers of seminal fluid proteins (SFPs). SFPs and sperm are transferred to the mating partner, where they are thought to play key roles in mediating post-mating sexual selection. They modulate the partner's behavior and physiology in ways that influence the reproductive success of both partners, thus potentially leading to sexual conflict. Despite the presumed general functional and evolutionary significance of SFPs, their identification and characterization has to date focused on just a few animal groups, predominantly insects and mammals. Moreover, until now seminal fluid profiling has mainly focused on species with separate sexes. Here we report a comprehensive screen for putative SFPs in the simultaneously hermaphroditic flatworm Macrostomum lignano. RESULTS: Based on existing transcriptomic data, we selected 150 transcripts known to be (a) predominantly expressed in the tail region of the worms, where the seminal fluid-producing prostate gland cells are located, and (b) differentially expressed in social environments differing in sperm competition level, strongly implying that they represent a phenotypically plastic aspect of male reproductive allocation in this species. For these SFP candidates, we then performed whole-mount in situ hybridization (ISH) experiments to characterize tissue-specific expression. In total, we identified 98 transcripts that exhibited prostate-specific expression, 76 of which we found to be expressed exclusively in the prostate gland cells; additional sites of expression for the remaining 22 included the testis or other gland cells. Bioinformatics analyses of the prostate-limited candidates revealed that at least 64 are predicted to be secretory proteins, making these especially strong candidates to be SFPs that are transferred during copulation. CONCLUSIONS: Our study represents a first comprehensive analysis using a combination of transcriptomic and ISH screen data to identify SFPs based on transcript expression in seminal fluid-producing tissues. We thereby extend the range of taxa for which seminal fluid has been characterized to a flatworm species with a sequenced genome and for which several methods such as antibody staining, transgenesis and RNA interference have been established. Our data provide a basis for testing the functional and evolutionary significance of SFPs.


Subject(s)
Hermaphroditic Organisms/metabolism , In Situ Hybridization/methods , Platyhelminths/metabolism , Seminal Plasma Proteins/metabolism , Animals , Female , Gene Expression Regulation , Gene Ontology , Hermaphroditic Organisms/genetics , Insect Proteins/genetics , Male , Organ Specificity , Phenotype , Platyhelminths/genetics , Prostate/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproduction , Spermatozoa/metabolism
12.
Mol Reprod Dev ; 85(6): 532-542, 2018 06.
Article in English | MEDLINE | ID: mdl-29693773

ABSTRACT

In Caenorhabditis briggsae hermaphrodites, spermatogenesis begins in the L4 larval stage and persists into early adulthood. Oogenesis begins after spermatogenesis; the sperm-to-oocyte transition is irreversible. The timing of this transition is believed to have evolved in response to selection to maximize the intrinsic growth rate. Sperm-to-oocyte transitions occurred early in Cbr-met-2 and Cbr-fem-3 mutants. These early transitions resulted in reduced brood sizes, but had little or no impact on the intrinsic growth rate. In Cbr-met-2; Cbr-fem-3 doubly mutant hermaphrodites, the transition to oogenesis occurred even earlier and brood size was further reduced, indicating that Cbr-met-2 and Cbr-fem-3 regulate the sperm-to-oocyte transition through separate pathways. Mutations in Cbr-met-2 also resulted in an increase in the frequency of males in mutant populations. These increased male frequencies were not caused by increased rates of X nondisjunction during oogenesis in mutant hermaphrodites. Rather, increases in the rates of outcrossing in mutant populations likely were an indirect effect of reduced brood sizes derived from self-fertilization. Based on these observations, it is possible that the timing of the sperm-to-oocyte transition in C. briggsae evolved in response to sexual selection on hermaphrodites to limit rates of outcrossing. Mutations in the orthologous Caenorhabditis elegans gene, Cel-met-2, did not impact the timing of the sperm-to-oocyte transition, consistent with the independent evolution of hermaphroditic reproduction in these species. Although brood sizes were reduced in Cel-met-2 mutant strains, increased male frequencies were not observed. Cbr- and Cel-met-2 mutations also differed in terms of germline mortality, observed in C. elegans, but not in C. briggsae.


Subject(s)
Caenorhabditis/metabolism , Helminth Proteins/metabolism , Hermaphroditic Organisms/metabolism , Oocytes/metabolism , Oogenesis/physiology , Spermatogenesis/physiology , Spermatozoa/metabolism , Animals , Caenorhabditis/cytology , Caenorhabditis/genetics , Helminth Proteins/genetics , Hermaphroditic Organisms/cytology , Hermaphroditic Organisms/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/cytology , Spermatozoa/cytology
13.
Nat Commun ; 9(1): 834, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483514

ABSTRACT

The formation of haploid gametes from diploid germ cells requires the regulated two-step release of sister chromatid cohesion (SCC) during the meiotic divisions. Here, we show that phosphorylation of cohesin subunit REC-8 by Aurora B promotes SCC release at anaphase I onset in C. elegans oocytes. Aurora B loading to chromatin displaying Haspin-mediated H3 T3 phosphorylation induces spatially restricted REC-8 phosphorylation, preventing full SCC release during anaphase I. H3 T3 phosphorylation is locally antagonized by protein phosphatase 1, which is recruited to chromosomes by HTP-1/2 and LAB-1. Mutating the N terminus of HTP-1 causes ectopic H3 T3 phosphorylation, triggering precocious SCC release without impairing earlier HTP-1 roles in homolog pairing and recombination. CDK-1 exerts temporal regulation of Aurora B recruitment, coupling REC-8 phosphorylation to oocyte maturation. Our findings elucidate a complex regulatory network that uses chromosome axis components, H3 T3 phosphorylation, and cell cycle regulators to ensure accurate chromosome segregation during oogenesis.


Subject(s)
Aurora Kinase B/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Hermaphroditic Organisms/genetics , Oocytes/metabolism , Anaphase , Animals , Aurora Kinase B/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromatids/metabolism , Chromatids/ultrastructure , Chromatin/metabolism , Chromatin/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Gene Expression Regulation , Hermaphroditic Organisms/cytology , Hermaphroditic Organisms/metabolism , Histones/genetics , Histones/metabolism , Oocytes/cytology , Oogenesis/genetics , Phosphorylation , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cohesins
14.
Mol Reprod Dev ; 84(11): C1, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29144576

ABSTRACT

Ctenophores, also known as comb jellies, are non-bilaterian invertebrates and nearly all are self-fertile hermaphrodites. Mnemiopsis leidyi is a particularly useful model for the study of cellular, tissue, and organ patterning in ctenophores due to their extreme transparency, as seen in these adults. The locomotory ctene rows, highlighted by iridescence, overlie the germ line, from which gametes and embryos are readily available in large numbers. In this issue, Davidson et al. characterize transcript expression and timing of the maternal-to-zygotic transition and accompanying zygotic genome activation during early embryogenesis in this ctenophore.


Subject(s)
Ctenophora/embryology , Gene Expression Regulation, Developmental/physiology , Genome , Germ Cells/metabolism , Hermaphroditic Organisms/metabolism , Zygote/metabolism , Animals , Ctenophora/cytology , Germ Cells/cytology , Hermaphroditic Organisms/cytology
15.
Dev Biol ; 429(1): 306-320, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28648844

ABSTRACT

RNA-binding proteins (RBPs) are essential regulators of gene expression that act through a variety of mechanisms to ensure the proper post-transcriptional regulation of their target RNAs. RBPs in multiple species have been identified as playing crucial roles during development and as having important functions in various adult organ systems, including the heart, nervous, muscle, and reproductive systems. ETR-1, a highly conserved ELAV-Type RNA-binding protein belonging to the CELF/Bruno protein family, has been previously reported to be involved in C. elegans muscle development. Animals depleted of ETR-1 have been previously characterized as arresting at the two-fold stage of embryogenesis. In this study, we show that ETR-1 is expressed in the hermaphrodite somatic gonad and germ line, and that reduction of ETR-1 via RNA interference (RNAi) results in reduced hermaphrodite fecundity. Detailed characterization of this fertility defect indicates that ETR-1 is required in both the somatic tissue and the germ line to ensure wild-type reproductive levels. Additionally, the ability of ETR-1 depletion to suppress the published WEE-1.3-depletion infertility phenotype is dependent on ETR-1 being reduced in the soma. Within the germline of etr-1(RNAi) hermaphrodite animals, we observe a decrease in average oocyte size and an increase in the number of germline apoptotic cell corpses as evident by an increased number of CED-1::GFP and acridine orange positive apoptotic germ cells. Transmission Electron Microscopy (TEM) studies confirm the significant increase in apoptotic cells in ETR-1-depleted animals, and reveal a failure of the somatic gonadal sheath cells to properly engulf dying germ cells in etr-1(RNAi) animals. Through investigation of an established engulfment pathway in C. elegans, we demonstrate that co-depletion of CED-1 and ETR-1 suppresses both the reduced fecundity and the increase in the number of apoptotic cell corpses observed in etr-1(RNAi) animals. Combined, this data identifies a novel role for ETR-1 in hermaphrodite gametogenesis and in the process of engulfment of germline apoptotic cell corpses.


Subject(s)
Apoptosis , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Germ Cells/metabolism , Phagocytosis , RNA-Binding Proteins/metabolism , Animals , Caenorhabditis elegans/ultrastructure , Cell Size , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Fertility , Gene Deletion , Germ Cells/cytology , Germ Cells/ultrastructure , Gonads/metabolism , Hermaphroditic Organisms/metabolism , Mitosis , Oocytes/cytology , Ovulation , Phenotype , RNA Interference , Reproduction
16.
Nucleic Acids Res ; 45(15): 9093-9107, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28645154

ABSTRACT

Caenorhabditis elegans contains 25 Argonautes, of which, ALG-1 and ALG-2 are known to primarily interact with miRNAs. ALG-5 belongs to the AGO subfamily of Argonautes that includes ALG-1 and ALG-2, but its role in small RNA pathways is unknown. We analyzed by high-throughput sequencing the small RNAs associated with ALG-5, ALG-1 and ALG-2, as well as changes in mRNA expression in alg-5, alg-1 and alg-2 mutants. We show that ALG-5 defines a distinct branch of the miRNA pathway affecting the expression of genes involved in immunity, defense, and development. In contrast to ALG-1 and ALG-2, which associate with most miRNAs and have general roles throughout development, ALG-5 interacts with only a small subset of miRNAs and is specifically expressed in the germline where it localizes alongside the piRNA and siRNA machinery at P granules. alg-5 is required for optimal fertility and mutations in alg-5 lead to a precocious transition from spermatogenesis to oogenesis. Our results provide a near-comprehensive analysis of miRNA-Argonaute interactions in C. elegans and reveal a new role for miRNAs in the germline.


Subject(s)
Argonaute Proteins/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental , Germ Cells/metabolism , RNA, Helminth/genetics , RNA-Binding Proteins/genetics , Animals , Argonaute Proteins/metabolism , Caenorhabditis elegans/classification , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Germ Cells/growth & development , Hermaphroditic Organisms/genetics , Hermaphroditic Organisms/growth & development , Hermaphroditic Organisms/metabolism , High-Throughput Nucleotide Sequencing , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Oogenesis/genetics , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Helminth/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Spermatogenesis/genetics
17.
Comp Biochem Physiol B Biochem Mol Biol ; 208-209: 47-57, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28400332

ABSTRACT

Dead end (dnd), vertebrate-specific germ cell marker, had been demonstrated to be essential for primordial germ cell (PGC) migration and survival, and the link between PGC number and sex change had been revealed in some teleost species, but little is known about dnd in hermaphroditic vertebrates. In the present study, a protogynous hermaphroditic orange-spotted grouper (Epinephelus coioides) dnd homologue (Ecdnd) was identified and characterized. Quantitative real-time PCR and in situ hybridization analysis revealed a dynamic and sexually dimorphic expression pattern in PGCs and germ cells of gonads. During sex changing, the Ecdnd transcript sharply increased in early transitional gonad, reached the highest level at late transitional gonad stage, and decreased after testis maturation. Visualization of zebrafish PGCs by injecting with RFP-Ecdnd-3'UTR RNA and GFP-zfnanos3-3'UTR RNA confirmed importance of Ecdnd 3'UTR for the PGC distribution. In addition, knockdown of EcDnd by using antisense morpholinos (MO) caused the ablation of PGCs in orange-spotted grouper. Therefore, the current data indicate that Ecdnd is essential for PGCs survival and may serve as a useful germ cell marker during gametogenesis in hermaphroditic grouper.


Subject(s)
Fish Proteins/metabolism , Germ Cells/cytology , Gonads/metabolism , Hermaphroditic Organisms/metabolism , Sex Characteristics , Sex Differentiation , Zebrafish/metabolism , Amino Acid Sequence , Animals , Female , Fish Proteins/genetics , Gene Expression Regulation , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Gonads/embryology , Hermaphroditic Organisms/genetics , Hermaphroditic Organisms/growth & development , In Situ Hybridization , Male , Phylogeny , Sequence Homology, Amino Acid , Zebrafish/genetics
18.
Sci Rep ; 7: 45175, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28338057

ABSTRACT

Many metazoans start germ cell development during embryogenesis, while some metazoans possessing pluripotent stem cells undergo postembryonic germ cell development. The latter reproduce asexually but develop germ cells from pluripotent stem cells or dormant primordial germ cells when they reproduce sexually. Sexual induction of the planarian Dugesia ryukyuensis is an important model for postembryonic germ cell development. In this experimental system, hermaphroditic reproductive organs are differentiated in presumptive gonadal regions by the administration of a crude extract from sexual planarians to asexual ones. However, the substances involved in the first event during postembryonic germ cell development, i.e., ovarian development, remain unknown. Here, we aimed to identify a bioactive compound associated with postembryonic ovarian development. Bioassay-guided fractionation identified ʟ-tryptophan (Trp) on the basis of electrospray ionization-mass spectrometry, circular dichroism, and nuclear magnetic resonance spectroscopy. Originally masked by a large amount of ʟ-Trp, ᴅ-Trp was detected by reverse-phase high-performance liquid chromatography. The ovary-inducing activity of ᴅ-Trp was 500 times more potent than that of ʟ-Trp. This is the first report describing a role for an intrinsic ᴅ-amino acid in postembryonic germ cell development. Our findings provide a novel insight into the mechanisms of germ cell development regulated by low-molecular weight bioactive compounds.


Subject(s)
Oogenesis , Ovary/metabolism , Planarians/metabolism , Tryptophan/metabolism , Animals , Female , Hermaphroditic Organisms/growth & development , Hermaphroditic Organisms/metabolism , Male , Ovary/cytology , Ovary/diagnostic imaging , Planarians/growth & development
19.
Int J Mol Sci ; 18(4)2017 Mar 23.
Article in English | MEDLINE | ID: mdl-28333083

ABSTRACT

Multiple nanos genes have been characterized in several fishes, but the functional implications of their various expression patterns remain unclear. In this study, we identified and characterized four nanos genes from a hermaphroditic fish orange-spotted grouper, Epinephelus coioides. Ecnanos1a and Ecnanos1b show divergent expression patterns, and the dynamic expression change of Ecnanos1a in pituitaries during sex change is associated with testis differentiation and spermatogenesis. Ecnanos2 and Ecnanos3 might be germline stem cells (GSCs) and primordial germ cells (PGCs)-specific markers, respectively. Significantly, Ecnanos3 3'-untranslated region (UTR) is necessary for PGC specific expression, where a non-canonical "GCACGTTT" sequence is required for miR-430-mediated repression of Ecnanos3 RNA. Furthermore, grouper Dead end (Dnd) can relieve miR-430 repression in PGCs by associating with a 23 bp U-rich region (URR) in Ecnanos3 3'-UTR. The current study revealed the functional association of multiple nanos genes with PGC formation and germ cell development in orange-spotted grouper, and opened up new possibilities for developing biotechnologies through utilizing the associations between Ecnanos3 and PGCs or between Ecnanos2 and GSCs in the hermaphroditic fish.


Subject(s)
Fish Proteins/genetics , Gene Expression Regulation, Developmental , Perciformes/genetics , RNA-Binding Proteins/genetics , 3' Untranslated Regions , Animals , Cell Differentiation , Fish Proteins/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Hermaphroditic Organisms/genetics , Hermaphroditic Organisms/growth & development , Hermaphroditic Organisms/metabolism , Male , MicroRNAs/genetics , Perciformes/growth & development , Perciformes/metabolism , RNA-Binding Proteins/metabolism , Testis/metabolism
20.
Biol Reprod ; 94(6): 132, 2016 06.
Article in English | MEDLINE | ID: mdl-27103447

ABSTRACT

In most hermaphroditic fish, the sexual phase of the gonad responds to external stimuli so that only one sex remains functional while the other sex becomes dormant. However, protandrous black porgy are male during their first two reproductive cycles. Estradiol (E2)-induced female growth results in a transient and immature female, and the sexual phase reverts from female to male after E2 is withdrawn. Conversely, excising the testis results in a precocious female when performed during the second reproductive cycle. We used these characteristics to study epigenetic modifications of cyp19a1a promoter in black porgy. Our results showed that higher levels of gonadotropins receptors were observed in testis than in ovary during the alteration of sexual phase from induced femaleness to maleness, and hCG treatment did not stimulate ovarian gene expression in male (1-yr-old maleness) and female phase (testis excision-induced femaleness) fish. The cyp19a1a promoter exhibited tissue- and lineage-specific methylation patterns. The follicle cells in the ovary had a hypomethylated (0%-20%) cyp19a1a promoter region. In the ovary, the first sign of female phase decision was decreased methylation levels and increased numbers of hypomethylated clones of cyp19a1a promoter during the natural sex change process. Similar methylation patterns were observed in the testis-removed ovary 1 mo after surgery, with no histological difference between the sham and the testis-removed fish. Conversely, there was no increase in methylation levels of cyp19a1a promoter in E2-fed fish. These results suggest that in the digonic gonad of black porgy, the testis is the primary tissue that affects epigenetics of the ovary.


Subject(s)
Epigenesis, Genetic , Hermaphroditic Organisms/metabolism , Ovary/metabolism , Perciformes/metabolism , Testis/metabolism , Animals , DNA Methylation , Female , Gonadotropins/metabolism , Hermaphroditic Organisms/genetics , Male , Perciformes/genetics , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...