Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
EMBO J ; 42(23): e113955, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37850882

ABSTRACT

Recent studies have reported the differentiation of pluripotent cells into oocytes in vitro. However, the developmental competence of in vitro-generated oocytes remains low. Here, we perform a comprehensive comparison of mouse germ cell development inĀ vitro over all culture steps versus in vivo with the goal to understand mechanisms underlying poor oocyte quality. We show that the in vitro differentiation of primordial germ cells to growing oocytes and subsequent follicle growth is critical for competence for preimplantation development. Systematic transcriptome analysis of single oocytes that were subjected to different culture steps identifies genes that are normally upregulated during oocyte growth to be susceptible for misregulation during in vitro oogenesis. Many misregulated genes are Polycomb targets. Deregulation of Polycomb repression is therefore a key cause and the earliest defect known in in vitro oocyte differentiation. Conversely, structurally normal in vitro-derived oocytes fail at zygotic genome activation and show abnormal acquisition of 5-hydroxymethylcytosine on maternal chromosomes. Our data identify epigenetic regulation at an early stage of oogenesis limiting developmental competence and suggest opportunities for future improvements.


Subject(s)
Epigenesis, Genetic , Oocytes , Female , Animals , Mice , Ovarian Follicle , Oogenesis/genetics , Germ Cells
2.
EMBO J ; 41(12): e109457, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35603814

ABSTRACT

The mammalian germline is characterized by extensive epigenetic reprogramming during its development into functional eggs and sperm. Specifically, the epigenome requires resetting before parental marks can be established and transmitted to the next generation. In the female germline, X-chromosome inactivation and reactivation are among the most prominent epigenetic reprogramming events, yet very little is known about their kinetics and biological function. Here, we investigate X-inactivation and reactivation dynamics using a tailor-made in vitro system of primordial germ cell-like cell (PGCLC) differentiation from mouse embryonic stem cells. We find that X-inactivation in PGCLCs in vitro and in germ cell-competent epiblast cells in vivo is moderate compared to somatic cells, and frequently characterized by escaping genes. X-inactivation is followed by step-wise X-reactivation, which is mostly completed during meiotic prophase I. Furthermore, we find that PGCLCs which fail to undergo X-inactivation or reactivate too rapidly display impaired meiotic potential. Thus, our data reveal fine-tuned X-chromosome remodelling as a critical feature of female germ cell development towards meiosis and oogenesis.


Subject(s)
Germ Cells , Meiosis , Animals , Cell Differentiation , Chromosomes , Mammals/genetics , Meiosis/genetics , Mice , X Chromosome Inactivation/genetics
3.
EMBO J ; 36(21): 3100-3119, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28928204

ABSTRACT

The mechanism for sex determination in mammalian germ cells remains unclear. Here, we reconstitute the female sex determination in mouse germ cells inĀ vitro under a defined condition without the use of gonadal somatic cells. We show that retinoic acid (RA) and its key effector, STRA8, are not sufficient to induce the female germ-cell fate. In contrast, bone morphogenetic protein (BMP) and RA synergistically induce primordial germ cells (PGCs)/PGC-like cells (PGCLCs) derived from embryonic stem cells (ESCs) into fetal primary oocytes. The induction is characterized by entry into the meiotic prophase, occurs synchronously and recapitulates cytological and transcriptome progression inĀ vivo faithfully. Importantly, the female germ-cell induction necessitates a proper cellular competence-most typically, DNA demethylation of relevant genes-which is observed in appropriately propagated PGCs/PGCLCs, but not in PGCs/PGCLCs immediately after induction. This provides an explanation for the differential function of BMP signaling between PGC specification and female germ-cell induction. Our findings represent a framework for a comprehensive delineation of the sex-determination pathway in mammalian germ cells, including humans.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Bone Morphogenetic Proteins/pharmacology , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/drug effects , Oocytes/drug effects , Tretinoin/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Female , Fetus , Gene Expression Profiling , Genes, Reporter , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred ICR , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Prophase , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sex Determination Processes , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Nat Rev Genet ; 13(7): 493-504, 2012 Jun 18.
Article in English | MEDLINE | ID: mdl-22705668

ABSTRACT

Trisomic and monosomic (aneuploid) embryos account for at least 10% of human pregnancies and, for women nearing the end of their reproductive lifespan, the incidence may exceed 50%. The errors that lead to aneuploidy almost always occur in the oocyte but, despite intensive investigation, the underlying molecular basis has remained elusive. Recent studies of humans and model organisms have shed new light on the complexity of meiotic defects, providing evidence that the age-related increase in errors in the human female is not attributable to a single factor but to an interplay between unique features of oogenesis and a host of endogenous and exogenous factors.


Subject(s)
Aneuploidy , Chromosome Segregation/physiology , Endocrine Disruptors/adverse effects , M Phase Cell Cycle Checkpoints/physiology , Meiosis/physiology , Oogenesis/physiology , Reproductive Techniques, Assisted , Age Factors , Female , Humans , Meiosis/genetics , Oogenesis/genetics , Sex Factors
5.
PLoS Genet ; 11(7): e1005355, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26176933

ABSTRACT

The origin of most ovarian tumors is undefined. Here, we report development of a novel mouse model in which conditional inactivation of the tumor suppressor gene Rb1 in oocytes leads to the formation of ovarian teratomas (OTs). While parthenogenetically activated ooctyes are a known source of OT in some mutant mouse models, enhanced parthenogenetic propensity in vitro was not observed for Rb1-deficient oocytes. Further analyses revealed that follicle recruitment and growth is disrupted in ovaries of mice with conditional inactivation of Rb1, leading to abnormal accumulation of secondary/preantral follicles. These findings underpin the concept that miscues between the germ cell and somatic compartments cause premature oocyte activation and the formation of OTs. Furthermore, these results suggest that defects in folliculogenesis and a permissive genetic background are sufficient to drive OT development, even in the absence of enhanced parthenogenetic activation. Thus, we have discovered a novel role of Rb1 in regulating the entry of primordial oocytes into the pool of growing follicles and signaling between the oocyte and granulosa cells during the protracted process of oocyte growth. Our findings, coupled with data from studies of other OT models, suggest that defects in the coordinated regulation between growth of the oocyte and somatic components in follicles are an underlying cause of OT formation.


Subject(s)
Oocytes/growth & development , Oogenesis , Ovarian Follicle/growth & development , Retinoblastoma Protein/genetics , Animals , Disease Models, Animal , Female , Humans , Meiosis/genetics , Mice , Oocytes/pathology , Ovarian Follicle/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Retinoblastoma Protein/biosynthesis , Signal Transduction , Teratoma/genetics , Teratoma/pathology
6.
Biol Reprod ; 94(1): 7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26586842

ABSTRACT

Primordial germ cells (PGCs) are the foundation of totipotency and vital for reproduction and heredity. PGCs in mice arise from the epiblast around Embryonic Day (E) 7.0, migrate through the hindgut endoderm, and colonize and proliferate in the embryonic gonads until around E13.5 prior to their differentiation either into prospermatogonia or oogonia. PRDM1, a transcriptional repressor, plays an essential role in PGC specification that includes robustly repressing a somatic mesodermal program. Using an inducible conditional knockout system, we show here that PRDM1 is critically required throughout PGC development. When Prdm1 was deleted in migrating PGCs at E9.5 or E10.5, or in male gonadal PGCs at E11.5, PGCs were eliminated by apoptosis from around E10.5, E11.5, or E13.5, respectively. When Prdm1 was deleted in female gonadal PGCs at E11.5, PGCs progressed into the first meiotic prophase in an apparently normal fashion, but the oogonia exhibited an aberrant pachytene phenotype, undergoing abrupt apoptosis from around E16.5. The escape of a fraction of PGCs (Ć¢ĀˆĀ¼10%) from the Prdm1 deletion was sufficient to recover fairly normal germ cell pools, both in male and female adults. The key targets of PRDM1 in migrating and/or gonadal PGCs, including genes for development, apoptosis, and prospermatogonial differentiation, showed only a modest overlap with those upon PGC specification, and were enriched with histone H3 lysine 27 trimethylation (H3K27me3). Our findings provide critical insight into the mechanism for maintaining the transcriptional integrity of PGCs.


Subject(s)
Germ Cells/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Histones/metabolism , Male , Meiosis , Mice , Mice, Knockout , Oogonia/metabolism , Ovary/embryology , Ovary/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Pregnancy , Testis/embryology , Testis/metabolism
7.
PLoS Genet ; 9(2): e1003241, 2013.
Article in English | MEDLINE | ID: mdl-23408896

ABSTRACT

Based on studies in mice and humans, cohesin loss from chromosomes during the period of protracted meiotic arrest appears to play a major role in chromosome segregation errors during female meiosis. In mice, mutations in meiosis-specific cohesin genes cause meiotic disturbances and infertility. However, the more clinically relevant situation, heterozygosity for mutations in these genes, has not been evaluated. We report here evidence from the mouse that partial loss of gene function for either Smc1b or Rec8 causes perturbations in the formation of the synaptonemal complex (SC) and affects both synapsis and recombination between homologs during meiotic prophase. Importantly, these defects increase the frequency of chromosomally abnormal eggs in the adult female. These findings have important implications for humans: they suggest that women who carry mutations or variants that affect cohesin function have an elevated risk of aneuploid pregnancies and may even be at increased risk of transmitting structural chromosome abnormalities.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation/genetics , Chromosomes , Meiosis/genetics , Animals , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Pairing/genetics , Chromosomes/genetics , Chromosomes/ultrastructure , Female , Gene Dosage , Humans , Mice , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Recombination, Genetic , Synaptonemal Complex/genetics , Cohesins
8.
Life Sci Alliance ; 6(11)2023 11.
Article in English | MEDLINE | ID: mdl-37722727

ABSTRACT

High-quality, straightforward single-cell RNA sequencing (RNA-seq) with spatial resolution remains challenging. Here, we developed DRaqL (direct RNA recovery and quenching for laser capture microdissection), an experimental approach for efficient cell lysis of tissue sections, directly applicable to cDNA amplification. Single-cell RNA-seq combined with DRaqL allowed transcriptomic profiling from alcohol-fixed sections with efficiency comparable with that of profiling from freshly dissociated cells, together with effective exon-exon junction profiling. The combination of DRaqL with protease treatment enabled robust and efficient single-cell transcriptome analysis from formalin-fixed tissue sections. Applying this method to mouse ovarian sections, we were able to predict the transcriptome of oocytes by their size and identified an anomaly in the size-transcriptome relationship relevant to growth retardation of oocytes, in addition to detecting oocyte-specific splice isoforms. Furthermore, we identified differentially expressed genes in granulosa cells in association with their proximity to the oocytes, suggesting distinct epigenetic regulations and cell-cycle activities governing the germ-soma relationship. Thus, DRaqL is a versatile, efficient approach for high-quality single-cell RNA-seq from tissue sections, thereby revealing histological heterogeneity in folliculogenic transcriptome.


Subject(s)
Ovary , Transcriptome , Female , Animals , Mice , Transcriptome/genetics , Gene Expression Profiling , Oocytes , Cell Cycle
9.
Nat Commun ; 12(1): 5041, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34413299

ABSTRACT

In vivo reprogramming provokes a wide range of cell fate conversion. Here, we discover that in vivo induction of higher levels of OSKM in mouse somatic cells leads to increased expression of primordial germ cell (PGC)-related genes and provokes genome-wide erasure of genomic imprinting, which takes place exclusively in PGCs. Moreover, the in vivo OSKM reprogramming resultsĀ in development of cancer that resembles human germ cell tumors. Like a subgroup of germ cell tumors, propagated tumor cells can differentiate into trophoblasts. Moreover, these tumor cells giveĀ rise to induced pluripotent stem cells (iPSCs) with expanded differentiation potential into trophoblasts. Remarkably, the tumor-derived iPSCs are able to contribute to non-neoplastic somatic cells in adult mice. Mechanistically, DMRT1, which is expressed in PGCs, drives the reprogramming and propagation of the tumor cells in vivo. Furthermore, the DMRT1-related epigenetic landscape is associated with trophoblast competence of the reprogrammed cells and provides a therapeutic target for germ cell tumors. These results reveal an unappreciated route for somatic cell reprogramming and underscore the impact of reprogramming in development of germ cell tumors.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms/pathology , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Cellular Reprogramming/physiology , Epigenesis, Genetic , Female , Genomic Imprinting , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred ICR , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Transcription Factors/genetics
11.
Science ; 367(6482)2020 03 06.
Article in English | MEDLINE | ID: mdl-32054698

ABSTRACT

Sex determination of germ cells is vital to creating the sexual dichotomy of germ cell development, thereby ensuring sexual reproduction. However, the underlying mechanisms remain unclear. Here, we show that ZGLP1, a conserved transcriptional regulator with GATA-like zinc fingers, determines the oogenic fate in mice. ZGLP1 acts downstream of bone morphogenetic protein, but not retinoic acid (RA), and is essential for the oogenic program and meiotic entry. ZGLP1 overexpression induces differentiation of in vitro primordial germ cell-like cells (PGCLCs) into fetal oocytes by activating the oogenic programs repressed by Polycomb activities, whereas RA signaling contributes to oogenic program maturation and PGC program repression. Our findings elucidate the mechanism for mammalian oogenic fate determination, providing a foundation for promoting in vitro gametogenesis and reproductive medicine.


Subject(s)
Gene Expression Regulation, Developmental , Oocytes/physiology , Oogenesis/genetics , Repressor Proteins/physiology , Sex Differentiation/genetics , Transcription Factors/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Bone Morphogenetic Proteins/metabolism , Female , Fetus/cytology , Male , Meiosis/genetics , Mice , Mice, Knockout , Oocytes/cytology , Polycomb-Group Proteins/metabolism , Repressor Proteins/genetics , Sex Determination Processes , Signal Transduction , Transcription Factors/genetics , Transcriptome , Tretinoin/physiology
12.
Article in English | MEDLINE | ID: mdl-29208639

ABSTRACT

Meiosis is a fundamental process that underpins sexual reproduction. In mammals, the execution of meiosis is tightly integrated within the complex processes of oogenesis and spermatogenesis, and elucidation of the molecular mechanisms regulating meiotic initiation remains challenging. We have recently developed in vitro culture strategies to induce mouse pluripotent stem cells into germ cells, which successfully contribute to both oogenesis and spermatogenesis and to fertile offspring. The culture strategies faithfully recapitulate transcriptional and epigenetic dynamics as well as signaling principles for germ cell specification, proliferation, and female sex determination/meiotic induction, providing a valuable platform for studies to illuminate the molecular mechanisms underlying such critical processes. Here, we review mammalian gametogenesis with a focus on the implementation of meiosis and, based on our recent studies, discuss new insights into the mechanisms for meiotic initiation and germ cell sex determination in mice.

13.
Cell Stem Cell ; 21(4): 517-532.e5, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28985527

ABSTRACT

Germline specification underlies human reproduction and evolution, but it has proven difficult to study in humans since it occurs shortly after blastocyst implantation. This process can be modeled with human induced pluripotent stem cells (hiPSCs) by differentiating them into primordial germ cell-like cells (hPGCLCs) through an incipient mesoderm-like cell (iMeLC) state. Here, we elucidate the key transcription factors and their interactions with important signaling pathways in driving hPGCLC differentiation from iPSCs. Germline competence of iMeLCs is dictated by the duration and dosage of WNT signaling, which induces expression of EOMES to activate SOX17, a key driver of hPGCLC specification. Upon hPGCLC induction, BMP signaling activates TFAP2C in a SOX17-independent manner. SOX17 and TFAP2C then cooperatively instate an hPGCLC transcriptional program, including BLIMP1 expression. This specification program diverges from its mouse counterpart regarding key transcription factors and their hierarchies, and it provides a foundation for further study of human germ cell development.


Subject(s)
Biological Evolution , Cell Lineage , Germ Cells/cytology , Pluripotent Stem Cells/cytology , Signal Transduction/genetics , Transcription, Genetic , Animals , Cell Lineage/genetics , Embryo Implantation/genetics , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Models, Biological , Pluripotent Stem Cells/metabolism , Primates , Transcription Factors/metabolism , Transcriptome/genetics , Wnt Signaling Pathway/genetics
14.
Genetics ; 196(2): 385-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24318536

ABSTRACT

Increasing age in a woman is a well-documented risk factor for meiotic errors, but the effect of paternal age is less clear. Although it is generally agreed that spermatogenesis declines with age, the mechanisms that account for this remain unclear. Because meiosis involves a complex and tightly regulated series of processes that include DNA replication, DNA repair, and cell cycle regulation, we postulated that the effects of age might be evident as an increase in the frequency of meiotic errors. Accordingly, we analyzed spermatogenesis in male mice of different ages, examining meiotic chromosome dynamics in spermatocytes at prophase, at metaphase I, and at metaphase II. Our analyses demonstrate that recombination levels are reduced in the first wave of spermatogenesis in juvenile mice but increase in older males. We also observed age-dependent increases in XY chromosome pairing failure at pachytene and in the frequency of prematurely separated autosomal homologs at metaphase I. However, we found no evidence of an age-related increase in aneuploidy at metaphase II, indicating that cells harboring meiotic errors are eliminated by cycle checkpoint mechanisms, regardless of paternal age. Taken together, our data suggest that advancing paternal age affects pairing, synapsis, and recombination between homologous chromosomes--and likely results in reduced sperm counts due to germ cell loss--but is not an important contributor to aneuploidy.


Subject(s)
Chromosomes, Mammalian , Meiosis , Paternal Age , Age Factors , Animals , Chromosome Pairing , DNA Breaks, Double-Stranded , Male , Metaphase , Mice , Recombination, Genetic , Sex Chromosomes , Spermatocytes/metabolism , Spermatogenesis/genetics
15.
Curr Biol ; 23(24): R1105-8, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24355789

ABSTRACT

The spindle assembly checkpoint prevents aneuploidy by ensuring that chromosomes are properly distributed during cell division. A new study shows that the integrity of the checkpoint response depends on centromeric cohesin in mammalian oocytes.


Subject(s)
Cell Cycle Proteins/physiology , Chromosomal Proteins, Non-Histone/physiology , Kinetochores/ultrastructure , M Phase Cell Cycle Checkpoints , Meiosis , Oocytes/cytology , Animals , Female , Cohesins
16.
Curr Biol ; 21(8): 651-7, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21497085

ABSTRACT

Segregation of homologs at the first meiotic division (MI) is facilitated by crossovers and by a physical constraint imposed on sister kinetochores that facilitates monopolar attachment to the MI spindle. Recombination failure or premature separation of homologs results in univalent chromosomes at MI, and univalents constrained to form monopolar attachments should be inherently unstable and trigger the spindle assembly checkpoint (SAC). Although univalents trigger cell-cycle arrest in the male, this is not the case in mammalian oocytes. Because the spindle assembly portion of the SAC appears to function normally, two hypotheses have been proposed to explain the lack of response to univalents: (1) reduced stringency of the oocyte SAC to aberrant chromosome behavior, and (2) the ability of univalents to satisfy the SAC by forming bipolar attachments. The present study of Mlh1 mutant mice demonstrates that metaphase alignment is not a prerequisite for anaphase onset and provides strong evidence that MI spindle stabilization and anaphase onset require stable bipolar attachment of a critical mass--but not all--of chromosomes. We postulate that subtle differences in SAC-mediated control make the human oocyte inherently error prone and contribute to the age-related increase in aneuploidy.


Subject(s)
Chromosome Segregation , Chromosomes, Mammalian/metabolism , Meiosis , Oocytes/cytology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aneuploidy , Animals , Cell Cycle , Cellular Senescence , Chromosomes, Mammalian/genetics , Female , Kinetochores/metabolism , Mice , Mice, Inbred C3H , MutL Protein Homolog 1 , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/metabolism , Sex Characteristics , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL