Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Nat Commun ; 15(1): 5248, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898112

ABSTRACT

Reproductive success relies on proper establishment and maintenance of biological sex. In many animals, including mammals, the primary gonad is initially ovary biased. We previously showed the RNA binding protein (RNAbp), Rbpms2, is required for ovary fate in zebrafish. Here, we identified Rbpms2 targets in oocytes (Rbpms2-bound oocyte RNAs; rboRNAs). We identify Rbpms2 as a translational regulator of rboRNAs, which include testis factors and ribosome biogenesis factors. Further, genetic analyses indicate that Rbpms2 promotes nucleolar amplification via the mTorc1 signaling pathway, specifically through the mTorc1-activating Gap activity towards Rags 2 (Gator2) component, Missing oocyte (Mios). Cumulatively, our findings indicate that early gonocytes are in a dual poised, bipotential state in which Rbpms2 acts as a binary fate-switch. Specifically, Rbpms2 represses testis factors and promotes oocyte factors to promote oocyte progression through an essential Gator2-mediated checkpoint, thereby integrating regulation of sexual differentiation factors and nutritional availability pathways in zebrafish oogenesis.


Subject(s)
Oocytes , Oogenesis , RNA-Binding Proteins , Zebrafish Proteins , Zebrafish , Animals , Female , Male , Gene Expression Regulation, Developmental , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Nutrients/metabolism , Oocytes/metabolism , Oogenesis/genetics , Ovary/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , Testis/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism
2.
bioRxiv ; 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38328218

ABSTRACT

Reproductive success relies on proper establishment and maintenance of biological sex. In many animals, including mammals, the primary gonad is initially ovary in character. We previously showed the RNA binding protein (RNAbp), Rbpms2, is required for ovary fate in zebrafish. Here, we identified Rbpms2 targets in oocytes (Rbpms2-bound oocyte RNAs; rboRNAs). We identify Rbpms2 as a translational regulator of rboRNAs, which include testis factors and ribosome biogenesis factors. Further, genetic analyses indicate that Rbpms2 promotes nucleolar amplification via the mTorc1 signaling pathway, specifically through the mTorc1-activating Gap activity towards Rags 2 (Gator2) component, Missing oocyte (Mios). Cumulatively, our findings indicate that early gonocytes are in a dual poised, bipotential state in which Rbpms2 acts as a binary fate-switch. Specifically, Rbpms2 represses testis factors and promotes oocyte factors to promote oocyte progression through an essential Gator2-mediated checkpoint, thereby integrating regulation of sexual differentiation factors and nutritional availability pathways in zebrafish oogenesis.

3.
Environ Toxicol Chem ; 41(11): 2708-2720, 2022 11.
Article in English | MEDLINE | ID: mdl-35920346

ABSTRACT

Metformin, along with its biotransformation product guanylurea, is commonly observed in municipal wastewaters and subsequent surface waters. Previous studies in fish have identified metformin as a potential endocrine-active compound, but there are inconsistencies with regard to its effects. To further investigate the potential reproductive toxicity of metformin and guanylurea to fish, a series of experiments was performed with adult fathead minnows (Pimephales promelas). First, explants of fathead minnow ovary tissue were exposed to 0.001-100 µM metformin or guanylurea to investigate whether the compounds could directly perturb steroidogenesis. Second, spawning pairs of fathead minnows were exposed to metformin (0.41, 4.1, and 41 µg/L) or guanylurea (1.0, 10, and 100 µg/L) for 23 days to assess impacts on reproduction. Lastly, male fathead minnows were exposed to 41 µg/L metformin, 100 µg/L guanylurea, or a mixture of both compounds, with samples collected over a 96-h time course to investigate potential impacts to the hepatic transcriptome or metabolome. Neither metformin nor guanylurea affected steroid production by ovary tissue exposed ex vivo. In the 23 days of exposure, neither compound significantly impacted transcription of endocrine-related genes in male liver or gonad, circulating steroid concentrations in either sex, or fecundity of spawning pairs. In the 96-h time course, 100 µg guanylurea/L elicited more differentially expressed genes than 41 µg metformin/L and showed the greatest impacts at 96 h. Hepatic transcriptome and metabolome changes were chemical- and time-dependent, with the largest impact on the metabolome observed at 23 days of exposure to 100 µg guanylurea/L. Overall, metformin and guanylurea did not elicit effects consistent with reproductive toxicity in adult fathead minnows at environmentally relevant concentrations. Environ Toxicol Chem 2022;41:2708-2720. © 2022 SETAC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.


Subject(s)
Cyprinidae , Metformin , Water Pollutants, Chemical , Animals , Female , Male , Metformin/toxicity , Wastewater , Water Pollutants, Chemical/analysis , Reproduction
4.
Development ; 147(18)2020 09 28.
Article in English | MEDLINE | ID: mdl-32895289

ABSTRACT

Sex determination and differentiation is a complex process regulated by multiple factors, including factors from the germline or surrounding somatic tissue. In zebrafish, sex-determination involves establishment of a bipotential ovary that undergoes sex-specific differentiation and maintenance to form the functional adult gonad. However, the relationships among these factors are not fully understood. Here, we identify potential Rbpms2 targets and apply genetic epistasis experiments to decipher the genetic hierarchy of regulators of sex-specific differentiation. We provide genetic evidence that the crucial female factor rbpms2 is epistatic to the male factor dmrt1 in terms of adult sex. Moreover, the role of Rbpms2 in promoting female fates extends beyond repression of Dmrt1, as Rbpms2 is essential for female differentiation even in the absence of Dmrt1. In contrast, female fates can be restored in mutants lacking both cyp19a1a and dmrt1, and prolonged in bmp15 mutants in the absence of dmrt1. Taken together, this work indicates that cyp19a1a-mediated suppression of dmrt1 establishes a bipotential ovary and initiates female fate acquisition. Then, after female fate specification, Cyp19a1a regulates subsequent oocyte maturation and sustains female fates independently of Dmrt1 repression.


Subject(s)
Aromatase/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Female , Germ Cells/physiology , Male , Ovary/physiology , Sex Determination Processes/genetics , Sex Determination Processes/physiology , Sex Differentiation/genetics , Sex Differentiation/physiology , Zebrafish/physiology
5.
Gastroenterology ; 156(6): 1788-1804.e13, 2019 05.
Article in English | MEDLINE | ID: mdl-30641053

ABSTRACT

BACKGROUND & AIMS: Patients with cirrhosis are at high risk for hepatocellular carcinoma (HCC) and often have increased serum levels of estrogen. It is not clear how estrogen promotes hepatic growth. We investigated the effects of estrogen on hepatocyte proliferation during zebrafish development, liver regeneration, and carcinogenesis. We also studied human hepatocytes and liver tissues. METHODS: Zebrafish were exposed to selective modifiers of estrogen signaling at larval and adult stages. Liver growth was assessed by gene expression, fluorescent imaging, and histologic analyses. We monitored liver regeneration after hepatocyte ablation and HCC development after administration of chemical carcinogens (dimethylbenzanthrazene). Proliferation of human hepatocytes was measured in a coculture system. We measured levels of G-protein-coupled estrogen receptor (GPER1) in HCC and nontumor liver tissues from 68 patients by immunohistochemistry. RESULTS: Exposure to 17ß-estradiol (E2) increased proliferation of hepatocytes and liver volume and mass in larval and adult zebrafish. Chemical genetic and epistasis experiments showed that GPER1 mediates the effects of E2 via the phosphoinositide 3-kinase-protein kinase B-mechanistic target of rapamycin pathway: gper1-knockout and mtor-knockout zebrafish did not increase liver growth in response to E2. HCC samples from patients had increased levels of GPER1 compared with nontumor tissue samples; estrogen promoted proliferation of human primary hepatocytes. Estrogen accelerated hepatocarcinogenesis specifically in male zebrafish. Chemical inhibition or genetic loss of GPER1 significantly reduced tumor development in the zebrafish. CONCLUSIONS: In an analysis of zebrafish and human liver cells and tissues, we found GPER1 to be a hepatic estrogen sensor that regulates liver growth during development, regeneration, and tumorigenesis. Inhibitors of GPER1 might be developed for liver cancer prevention or treatment. TRANSCRIPT PROFILING: The accession number in the Gene Expression Omnibus is GSE92544.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Liver Neoplasms/metabolism , Liver/growth & development , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Zebrafish Proteins/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Hepatocytes , Humans , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/pathology , Liver Regeneration , Male , Organ Size/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Receptors, G-Protein-Coupled/genetics , Sex Factors , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Zebrafish , Zebrafish Proteins/genetics
6.
Endocrinology ; 159(10): 3515-3523, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30169775

ABSTRACT

Estrogens regulate vertebrate development and function through binding to nuclear estrogen receptors α and ß (ERα and ERß) and the G protein-coupled estrogen receptor (GPER). Studies in mutant animal models demonstrated that ERα and ERß are required for normal ovary development and function. However, the degree to which GPER signaling contributes to ovary development and function is less well understood. Previous studies using cultured fish oocytes found that estradiol inhibits oocyte maturation in a GPER-dependent manner, but whether GPER regulates oocyte maturation in vivo is not known. To test the hypothesis that GPER regulates oocyte maturation in vivo, we assayed ovary development and function in gper mutant zebrafish. We found that homozygous mutant gper embryos developed into male and female adults with normal sex ratios. Adult mutant fish exhibited normal secondary sex characteristics and fertility. Additionally, mutant ovaries were histologically normal. We observed no differences in the number of immature versus mature oocytes in mutant versus wild-type ovaries from both young and aged adults. Furthermore, expression of genes associated with sex determination and ovary function was normal in gper mutant ovaries compared with wild type. Our findings suggest that GPER is not required for sex determination, ovary development, or fertility in zebrafish.


Subject(s)
Ovary/metabolism , Receptors, G-Protein-Coupled/genetics , Sex Determination Analysis/methods , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Female , Gene Expression Regulation, Developmental , Genotype , Male , Mutation , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Ovary/embryology , Ovary/growth & development , Receptors, G-Protein-Coupled/metabolism , Sex Ratio , Zebrafish/metabolism , Zebrafish Proteins/metabolism
7.
Gen Comp Endocrinol ; 261: 190-197, 2018 05 15.
Article in English | MEDLINE | ID: mdl-28450143

ABSTRACT

In 2005, two groups independently discovered that the G protein-coupled receptor GPR30 binds estradiol in cultured cells and, in response, initiates intracellular signaling cascades Revankar et al. (2005), Thomas et al. (2005). GPR30 is now referred to as GPER, the G-protein coupled estrogen receptor Prossnitz and Arterburn (2015). While studies in animal models are illuminating GPER function, there is controversy as to whether GPER acts as an autonomous estrogen receptor in vivo, or whether GPER interacts with nuclear estrogen receptor signaling pathways in response to estrogens. Here, we review the evidence that GPER acts as an autonomous estrogen receptor in vivo and discuss experimental approaches to test this hypothesis directly. We propose that the degree to which GPER influences nuclear estrogen receptor signaling likely depends on cell type, developmental stage and pathology.


Subject(s)
Receptor Cross-Talk/physiology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Nucleus/metabolism , Estradiol/metabolism , Estrogens/metabolism , Humans , Signal Transduction/physiology
8.
PLoS Genet ; 13(10): e1007069, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29065151

ABSTRACT

Estrogens act by binding to estrogen receptors alpha and beta (ERα, ERß), ligand-dependent transcription factors that play crucial roles in sex differentiation, tumor growth and cardiovascular physiology. Estrogens also activate the G protein-coupled estrogen receptor (GPER), however the function of GPER in vivo is less well understood. Here we find that GPER is required for normal heart rate in zebrafish embryos. Acute exposure to estrogens increased heart rate in wildtype and in ERα and ERß mutant embryos but not in GPER mutants. GPER mutant embryos exhibited reduced basal heart rate, while heart rate was normal in ERα and ERß mutants. We detected gper transcript in discrete regions of the brain and pituitary but not in the heart, suggesting that GPER acts centrally to regulate heart rate. In the pituitary, we observed gper expression in cells that regulate levels of thyroid hormone triiodothyronine (T3), a hormone known to increase heart rate. Compared to wild type, GPER mutants had reduced levels of T3 and estrogens, suggesting pituitary abnormalities. Exposure to exogenous T3, but not estradiol, rescued the reduced heart rate phenotype in gper mutant embryos, demonstrating that T3 acts downstream of GPER to regulate heart rate. Using genetic and mass spectrometry approaches, we find that GPER regulates maternal estrogen levels, which are required for normal embryonic heart rate. Our results demonstrate that estradiol plays a previously unappreciated role in the acute modulation of heart rate during zebrafish embryonic development and suggest that GPER regulates embryonic heart rate by altering maternal estrogen levels and embryonic T3 levels.


Subject(s)
Embryo, Nonmammalian/physiology , Estradiol/administration & dosage , Heart Rate/physiology , Receptors, G-Protein-Coupled/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Animals , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Estrogens/analysis , Gene Expression Regulation, Developmental/drug effects , Mutation , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Triiodothyronine/analysis , Zebrafish Proteins/genetics
9.
J Vis Exp ; (87)2014 May 17.
Article in English | MEDLINE | ID: mdl-24894681

ABSTRACT

Zebrafish embryos are a powerful tool for large-scale screening of small molecules. Transgenic zebrafish that express fluorescent reporter proteins are frequently used to identify chemicals that modulate gene expression. Chemical screens that assay fluorescence in live zebrafish often rely on expensive, specialized equipment for high content screening. We describe a procedure using a standard epifluorescence microscope with a motorized stage to automatically image zebrafish embryos and detect tissue-specific fluorescence. Using transgenic zebrafish that report estrogen receptor activity via expression of GFP, we developed a semi-automated procedure to screen for estrogen receptor ligands that activate the reporter in a tissue-specific manner. In this video we describe procedures for arraying zebrafish embryos at 24-48 hours post fertilization (hpf) in a 96-well plate and adding small molecules that bind estrogen receptors. At 72-96 hpf, images of each well from the entire plate are automatically collected and manually inspected for tissue-specific fluorescence. This protocol demonstrates the ability to detect estrogens that activate receptors in heart valves but not in liver.


Subject(s)
Green Fluorescent Proteins/chemistry , Microscopy, Fluorescence/methods , Zebrafish/embryology , Animals , Animals, Genetically Modified , Automation , Embryo, Nonmammalian , Female , Ligands , Male , Receptors, Estrogen/chemistry , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL