Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 12: 1410177, 2024.
Article in English | MEDLINE | ID: mdl-38911025

ABSTRACT

Mammalian germ cells are derived from primordial germ cells (PGCs) and ensure species continuity through generations. Unlike irreversible committed mature germ cells, migratory PGCs exhibit a latent pluripotency characterized by the ability to derive embryonic germ cells (EGCs) and form teratoma. Here, we show that inhibition of p38 mitogen-activated protein kinase (MAPK) by chemical compounds in mouse migratory PGCs enables derivation of chemically induced Embryonic Germ-like Cells (cEGLCs) that do not require conventional growth factors like LIF and FGF2/Activin-A, and possess unique naïve pluripotent-like characteristics with epiblast features and chimera formation potential. Furthermore, cEGLCs are regulated by a unique PI3K-Akt signaling pathway, distinct from conventional naïve pluripotent stem cells described previously. Consistent with this notion, we show by performing ex vivo analysis that inhibition of p38 MAPK in organ culture supports the survival and proliferation of PGCs and also potentially reprograms PGCs to acquire indefinite proliferative capabilities, marking these cells as putative teratoma-producing cells. These findings highlight the utility of our ex vivo model in mimicking in vivo teratoma formation, thereby providing valuable insights into the cellular mechanisms underlying tumorigenesis. Taken together, our research underscores a key role of p38 MAPK in germ cell development, maintaining proper cell fate by preventing unscheduled pluripotency and teratoma formation with a balance between proliferation and differentiation.

2.
EMBO Rep ; 24(11): e56845, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37842859

ABSTRACT

Fate determination of primordial germ cells (PGCs) is regulated in a multi-layered manner, involving signaling pathways, epigenetic mechanisms, and transcriptional control. Chemical modification of macromolecules, including epigenetics, is expected to be closely related with metabolic mechanisms but the detailed molecular machinery linking these two layers remains poorly understood. Here, we show that the hexosamine biosynthetic pathway controls PGC fate determination via O-linked ß-N-acetylglucosamine (O-GlcNAc) modification. Consistent with this model, reduction of carbohydrate metabolism via a maternal ketogenic diet that decreases O-GlcNAcylation levels causes repression of PGC formation in vivo. Moreover, maternal ketogenic diet intake until mid-gestation affects the number of ovarian germ cells in newborn pups. Taken together, we show that nutritional and metabolic mechanisms play a previously unappreciated role in PGC fate determination.


Subject(s)
Acetylglucosamine , Signal Transduction , Infant, Newborn , Humans , Signal Transduction/physiology , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Gene Expression Regulation , Epigenesis, Genetic , Germ Cells/metabolism , Protein Processing, Post-Translational
4.
Elife ; 102021 07 28.
Article in English | MEDLINE | ID: mdl-34319233

ABSTRACT

Exposure to environmental factors during fetal development may lead to epigenomic modifications in fetal germ cells, altering gene expression and promoting diseases in successive generations. In mouse, maternal exposure to di(2-ethylhexyl) phthalate (DEHP) is known to induce defects in spermatogenesis in successive generations, but the mechanism(s) of impaired spermatogenesis are unclear. Here, we showed that maternal DEHP exposure results in DNA hypermethylation of promoters of spermatogenesis-related genes in fetal testicular germ cells in F1 mice, and hypermethylation of Hist1h2ba, Sycp1, and Taf7l, which are crucial for spermatogenesis, persisted from fetal testicular cells to adult spermatogonia, resulting in the downregulation of expression of these genes. Forced methylation of these gene promoters silenced expression of these loci in a reporter assay. These results suggested that maternal DEHP exposure-induced hypermethylation of Hist1h2ba, Sycp1, and Taf7l results in downregulation of these genes in spermatogonia and subsequent defects in spermatogenesis, at least in the F1 generation.


Subject(s)
Diethylhexyl Phthalate/pharmacology , Maternal Exposure/adverse effects , Mutation , Phthalic Acids/adverse effects , Prenatal Exposure Delayed Effects/genetics , Spermatogenesis/drug effects , Spermatogenesis/genetics , Animals , DNA Methylation , Down-Regulation , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phthalic Acids/chemistry , Plasticizers/adverse effects , Pregnancy , Spermatogonia/drug effects , Testis/cytology , Testis/drug effects
5.
Biol Reprod ; 105(1): 64-75, 2021 07 02.
Article in English | MEDLINE | ID: mdl-33824958

ABSTRACT

Fetal ovarian germ cells show characteristic energy metabolism status, such as enhanced mitochondrial metabolism as well as glycolysis, but their roles in early folliculogenesis are unclear. We show here that inhibition of pyruvate uptake to mitochondria by UK5099 in organ cultures of fetal mouse ovaries resulted in repressed early folliculogenesis without affecting energy production, survival of oocytes, or meiosis. In addition, the abnormal folliculogenesis by UK5099 was partially rescued by α-ketoglutarate and succinate, intermediate metabolites in the TCA cycle, suggesting the importance of those metabolites. The expression of TGFß-related genes Gdf9 and Bmp15 in ovarian germ cells, which are crucial for folliculogenesis, was downregulated by UK5099, and the addition of recombinant GDF9 partially rescued the abnormal folliculogenesis induced by UK5099. We also found that early folliculogenesis was similarly repressed, as in the culture, in the ovaries of a germ cell-specific knockout of Mpc2, which encodes a mitochondria pyruvate carrier that is targeted by UK5099. These results suggest that insufficient Gdf9 expression induced by abnormal pyruvate metabolism in oocytes results in early follicular dysgenesis, which is a possible cause of defective folliculogenesis in humans.


Subject(s)
Acrylates/pharmacology , Bone Morphogenetic Protein 15/genetics , Growth Differentiation Factor 9/genetics , Oocytes/drug effects , Ovarian Follicle/growth & development , Pyruvic Acid/metabolism , Animals , Biological Transport , Bone Morphogenetic Protein 15/metabolism , Citric Acid Cycle , Female , Gene Expression Regulation , Growth Differentiation Factor 9/metabolism , Mice , Mitochondria/metabolism , Oocytes/metabolism
6.
Biol Reprod ; 103(4): 717-735, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32627815

ABSTRACT

Regulatory mechanisms of germline differentiation have generally been explained via the function of signaling pathways, transcription factors, and epigenetic regulation; however, little is known regarding proteomic and metabolomic regulation and their contribution to germ cell development. Here, we conducted integrated proteomic and metabolomic analyses of fetal germ cells in mice on embryonic day (E)13.5 and E18.5 and demonstrate sex- and developmental stage-dependent changes in these processes. In male germ cells, RNA processing, translation, oxidative phosphorylation, and nucleotide synthesis are dominant in E13.5 and then decline until E18.5, which corresponds to the prolonged cell division and more enhanced hyper-transcription/translation in male primordial germ cells and their subsequent repression. Tricarboxylic acid cycle and one-carbon pathway are consistently upregulated in fetal male germ cells, suggesting their involvement in epigenetic changes preceding in males. Increased protein stability and oxidative phosphorylation during female germ cell differentiation suggests an upregulation of aerobic energy metabolism, which likely contributes to the proteostasis required for oocyte maturation in subsequent stages. The features elucidated in this study shed light on the unrevealed mechanisms of germ cell development.


Subject(s)
Cell Differentiation/physiology , Embryonic Germ Cells/physiology , Metabolomics , Proteomics , Animals , DNA/genetics , DNA/metabolism , DNA Methylation , Embryo, Mammalian/physiology , Female , Gene Expression Regulation, Developmental , Genome-Wide Association Study , Male , Mice , Mice, Transgenic , Sex Differentiation , Sex Factors , Transcription Factors
8.
PLoS One ; 14(2): e0211739, 2019.
Article in English | MEDLINE | ID: mdl-30707741

ABSTRACT

MicroRNAs (miRNAs) play a critical role in multiple aspects of biology. Dicer, an RNase III endonuclease, is essential for the biogenesis of miRNAs, and the germ cell-specific Dicer1 knockout mouse shows severe defects in gametogenesis. How miRNAs regulate germ cell development is still not fully understood. In this study, we identified germ cell-specific miRNAs (miR-741-3p, miR-871-3p, miR-880-3p) by analyzing published RNA-seq data of mouse. These miRNA genes are contiguously located on the X chromosome near other miRNA genes. We named them X chromosome-linked miRNAs (XmiRs). To elucidate the functions of XmiRs, we generated knockout mice of these miRNA genes using the CRISPR/Cas9-mediated genome editing system. Although no histological abnormalities were observed in testes of F0 mice in which each miRNA gene was disrupted, a deletion covering miR-871 and miR-880 or covering all XmiRs (ΔXmiRs) resulted in arrested spermatogenesis in meiosis in a few seminiferous tubules, indicating their redundant functions in spermatogenesis. Among candidate targets of XmiRs, we found increased expression of a gene encoding a WNT receptor, FZD4, in ΔXmiRs testis compared with that in wildtype testis. miR-871-3p and miR-880-3p repressed the expression of Fzd4 via the 3'-untranslated region of its mRNA. In addition, downstream genes of the WNT/ß-catenin pathway were upregulated in ΔXmiRs testis. We also found that miR-871, miR-880, and Fzd4 were expressed in spermatogonia, spermatocytes and spermatids, and overexpression of miR-871 and miR-880 in germ stem cells in culture repressed their increase in number and Fzd4 expression. Previous studies indicated that the WNT/ß-catenin pathway enhances and represses proliferation and differentiation of spermatogonia, respectively, and our results consistently showed that stable ß-catenin enhanced GSC number. In addition, stable ß-catenin partially rescued reduced GSC number by overexpression of miR-871 and miR-880. The results together suggest that miR-871 and miR-880 cooperatively regulate the WNT/ß-catenin pathway during testicular germ cell development.


Subject(s)
MicroRNAs/genetics , Spermatogenesis/genetics , X Chromosome/genetics , Animals , CRISPR-Cas Systems , Cell Differentiation/genetics , Cell Proliferation , Germ Cells , Male , Meiosis , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/classification , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogonia/metabolism , Testis/metabolism
9.
Dev Biol ; 446(1): 43-55, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30529251

ABSTRACT

Teratomas are tumors consisting of components of the three germ layers that differentiate from pluripotent stem cells derived from germ cells. In the normal mouse testis, teratomas rarely form, but a deficiency in Dead-end1 (Dnd1) in mice with a 129/Sv genetic background greatly enhances teratoma formation. Thus, DND1 is crucial for suppression of teratoma development from germ cells. In the Dnd1 mutant testis, nascent teratoma cells emerge at E15.5. To understand the nature of early teratoma cells, we established cell lines in the presence of serum and leukemia inhibitory factor (LIF) from teratoma-forming cells in neonatal Dnd1 mutant testis. These cells, which we designated cultured Dnd1 mutant germ cells (CDGCs), were morphologically similar to embryonic stem cells (ESCs) and could be maintained in the naïve pluripotent condition. In addition, the cells expressed pluripotency genes including Oct4, Nanog, and Sox2; differentiated into cells of the three germ layers in culture; and contributed to chimeric mice. The expression levels of pluripotency genes and global transcriptomes in CDGCs as well as these cells' adaption to culture conditions for primed pluripotency suggested that their pluripotent status is intermediate between naïve and primed pluripotency. In addition, the teratoma-forming cells in the neonatal testis from which CDGCs were derived also showed gene expression profiles intermediate between naïve and primed pluripotency. The results suggested that germ cells in embryonic testes of Dnd1 mutants acquire the intermediate pluripotent status during the course of conversion into teratoma cells.


Subject(s)
Cell Differentiation/genetics , Mouse Embryonic Stem Cells/metabolism , Neoplasm Proteins/genetics , Pluripotent Stem Cells/metabolism , Animals , Animals, Newborn , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Developmental , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplasm Proteins/deficiency , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Teratoma/genetics , Teratoma/metabolism , Teratoma/pathology , Testis/cytology , Testis/embryology , Testis/metabolism
10.
Development ; 145(23)2018 12 03.
Article in English | MEDLINE | ID: mdl-30446626

ABSTRACT

In mouse embryos, primordial germ cells (PGCs) are fate-determined from epiblast cells. Signaling pathways involved in PGC formation have been identified, but their epigenetic mechanisms remain poorly understood. Here, we show that the histone methyltransferase SETDB1 is an epigenetic regulator of PGC fate determination. Setdb1-deficient embryos exhibit drastic reduction of nascent PGCs. Dppa2, Otx2 and Utf1 are de-repressed whereas mesoderm development-related genes, including BMP4 signaling-related genes, are downregulated by Setdb1 knockdown during PGC-like cell (PGCLC) induction. In addition, binding of SETDB1 is observed at the flanking regions of Dppa2, Otx2 and Utf1 in cell aggregates containing PGCLCs, and trimethylation of lysine 9 of histone H3 is reduced by Setdb1 knockdown at those regions. Furthermore, DPPA2, OTX2 and UTF1 binding is increased in genes encoding BMP4 signaling-related proteins, including SMAD1. Finally, overexpression of Dppa2, Otx2 and Utf1 in cell aggregates containing PGCLCs results in the repression of BMP4 signaling-related genes and PGC determinant genes. We propose that the localization of SETDB1 to Dppa2, Otx2 and Utf1, and subsequent repression of their expression, are crucial for PGC determination by ensuring BMP4 signaling.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Cell Lineage , Germ Cells/cytology , Germ Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Signal Transduction , Animals , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Mesoderm/embryology , Mesoderm/metabolism , Mice , Transcription Factors/metabolism , Up-Regulation/genetics
11.
Cell Rep ; 24(10): 2682-2693.e6, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30184502

ABSTRACT

Primordial germ cells (PGCs) are fate determined from pluripotent epiblasts. Signaling pathways and transcriptional regulators involved in PGC formation have been identified, but detailed molecular mechanisms of PGC fate determination remains poorly understood. Using RNAi screening, we identified histone deacetylase 3 (HDAC3) as a regulator of PGC formation. Hdac3 deficiency resulted in decreased nascent PGCs in vitro and in vivo, and somatic developmental genes were de-repressed by Hdac3 knockdown during PGC induction. We also demonstrated BLIMP1-dependent enrichment of HDAC3 and deacetylation of H3 and H4 histones in the somatic developmental genes in epiblast-like cells. In addition, the HDAC3/BLIMP1-targeted somatic gene products were enriched in PGC determinant genes; overexpression of these gene products in PGC-like cells in culture resulted in repression of PGC determinant genes. We propose that selective recruitment of HDAC3 to somatic genes by BLIMP1 and subsequent repression of these somatic genes are crucial for PGC fate determination.


Subject(s)
Germ Cells/metabolism , Histone Deacetylases/metabolism , Positive Regulatory Domain I-Binding Factor 1/metabolism , Acetylation , Animals , Gene Expression Regulation, Developmental , Histone Deacetylases/genetics , Histones/metabolism , Mice , Positive Regulatory Domain I-Binding Factor 1/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...