Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52.170
Filter
Add more filters

Publication year range
1.
Cell ; 187(5): 1109-1126.e21, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38382525

ABSTRACT

Oocytes are among the longest-lived cells in the body and need to preserve their cytoplasm to support proper embryonic development. Protein aggregation is a major threat for intracellular homeostasis in long-lived cells. How oocytes cope with protein aggregation during their extended life is unknown. Here, we find that mouse oocytes accumulate protein aggregates in specialized compartments that we named endolysosomal vesicular assemblies (ELVAs). Combining live-cell imaging, electron microscopy, and proteomics, we found that ELVAs are non-membrane-bound compartments composed of endolysosomes, autophagosomes, and proteasomes held together by a protein matrix formed by RUFY1. Functional assays revealed that in immature oocytes, ELVAs sequester aggregated proteins, including TDP-43, and degrade them upon oocyte maturation. Inhibiting degradative activity in ELVAs leads to the accumulation of protein aggregates in the embryo and is detrimental for embryo survival. Thus, ELVAs represent a strategy to safeguard protein homeostasis in long-lived cells.


Subject(s)
Cytoplasmic Vesicles , Oocytes , Protein Aggregates , Animals , Female , Mice , Autophagosomes , Cytoplasmic Vesicles/metabolism , Lysosomes/metabolism , Oocytes/cytology , Oocytes/metabolism , Proteasome Endopeptidase Complex , Proteolysis
2.
Cell ; 187(8): 1889-1906.e24, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38503281

ABSTRACT

Nucleoli are multicomponent condensates defined by coexisting sub-phases. We identified distinct intrinsically disordered regions (IDRs), including acidic (D/E) tracts and K-blocks interspersed by E-rich regions, as defining features of nucleolar proteins. We show that the localization preferences of nucleolar proteins are determined by their IDRs and the types of RNA or DNA binding domains they encompass. In vitro reconstitutions and studies in cells showed how condensation, which combines binding and complex coacervation of nucleolar components, contributes to nucleolar organization. D/E tracts of nucleolar proteins contribute to lowering the pH of co-condensates formed with nucleolar RNAs in vitro. In cells, this sets up a pH gradient between nucleoli and the nucleoplasm. By contrast, juxta-nucleolar bodies, which have different macromolecular compositions, featuring protein IDRs with very different charge profiles, have pH values that are equivalent to or higher than the nucleoplasm. Our findings show that distinct compositional specificities generate distinct physicochemical properties for condensates.


Subject(s)
Cell Nucleolus , Nuclear Proteins , Proton-Motive Force , Cell Nucleolus/chemistry , Cell Nucleus/chemistry , Nuclear Proteins/chemistry , RNA/metabolism , Phase Separation , Intrinsically Disordered Proteins/chemistry , Animals , Xenopus laevis , Oocytes/chemistry , Oocytes/cytology
3.
Cell ; 186(24): 5308-5327.e25, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37922900

ABSTRACT

Mammalian oocytes are filled with poorly understood structures called cytoplasmic lattices. First discovered in the 1960s and speculated to correspond to mammalian yolk, ribosomal arrays, or intermediate filaments, their function has remained enigmatic to date. Here, we show that cytoplasmic lattices are sites where oocytes store essential proteins for early embryonic development. Using super-resolution light microscopy and cryoelectron tomography, we show that cytoplasmic lattices are composed of filaments with a high surface area, which contain PADI6 and subcortical maternal complex proteins. The lattices associate with many proteins critical for embryonic development, including proteins that control epigenetic reprogramming of the preimplantation embryo. Loss of cytoplasmic lattices by knocking out PADI6 or the subcortical maternal complex prevents the accumulation of these proteins and results in early embryonic arrest. Our work suggests that cytoplasmic lattices enrich maternally provided proteins to prevent their premature degradation and cellular activity, thereby enabling early mammalian development.


Subject(s)
Oocytes , Proteins , Pregnancy , Animals , Female , Oocytes/metabolism , Proteins/metabolism , Embryo, Mammalian/metabolism , Cytoskeleton , Ribosomes , Embryonic Development , Mammals
4.
Cell ; 185(14): 2576-2590.e12, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35623357

ABSTRACT

Mouse germline cysts, on average, develop into six oocytes supported by 24 nurse cells that transfer cytoplasm and organelles to generate a Balbiani body. We showed that between E14.5 and P5, cysts periodically activate some nurse cells to begin cytoplasmic transfer, which causes them to shrink and turnover within 2 days. Nurse cells die by a programmed cell death (PCD) pathway involving acidification, similar to Drosophila nurse cells, and only infrequently by apoptosis. Prior to initiating transfer, nurse cells co-cluster by scRNA-seq with their pro-oocyte sisters, but during their final 2 days, they cluster separately. The genes promoting oocyte development and nurse cell PCD are upregulated, whereas the genes that repress transfer, such as Tex14, and oocyte factors, such as Nobox and Lhx8, are under-expressed. The transferred nurse cell centrosomes build a cytocentrum that establishes a large microtubule aster in the primordial oocyte that organizes the Balbiani body, defining the earliest oocyte polarity.


Subject(s)
Cell Lineage , Cysts , Oocytes , Animals , Apoptosis , Cell Enlargement , Cysts/genetics , Cysts/metabolism , Cytoplasm/metabolism , Drosophila melanogaster , Female , Gene Expression Regulation, Developmental , Mice , Oocytes/cytology , Oocytes/metabolism , Ovary/cytology , Ovary/embryology , Ovary/metabolism
5.
Cell ; 185(8): 1308-1324.e23, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35325593

ABSTRACT

Asymmetric localization of oskar ribonucleoprotein (RNP) granules to the oocyte posterior is crucial for abdominal patterning and germline formation in the Drosophila embryo. We show that oskar RNP granules in the oocyte are condensates with solid-like physical properties. Using purified oskar RNA and scaffold proteins Bruno and Hrp48, we confirm in vitro that oskar granules undergo a liquid-to-solid phase transition. Whereas the liquid phase allows RNA incorporation, the solid phase precludes incorporation of additional RNA while allowing RNA-dependent partitioning of client proteins. Genetic modification of scaffold granule proteins or tethering the intrinsically disordered region of human fused in sarcoma (FUS) to oskar mRNA allowed modulation of granule material properties in vivo. The resulting liquid-like properties impaired oskar localization and translation with severe consequences on embryonic development. Our study reflects how physiological phase transitions shape RNA-protein condensates to regulate the localization and expression of a maternal RNA that instructs germline formation.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Embryo, Nonmammalian/metabolism , Animals , Cytoplasmic Ribonucleoprotein Granules , Drosophila/embryology , Drosophila Proteins/genetics , Embryonic Development , Oocytes/metabolism , RNA/metabolism
6.
Cell ; 184(19): 4904-4918.e11, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34433012

ABSTRACT

Selfish centromere DNA sequences bias their transmission to the egg in female meiosis. Evolutionary theory suggests that centromere proteins evolve to suppress costs of this "centromere drive." In hybrid mouse models with genetically different maternal and paternal centromeres, selfish centromere DNA exploits a kinetochore pathway to recruit microtubule-destabilizing proteins that act as drive effectors. We show that such functional differences are suppressed by a parallel pathway for effector recruitment by heterochromatin, which is similar between centromeres in this system. Disrupting the kinetochore pathway with a divergent allele of CENP-C reduces functional differences between centromeres, whereas disrupting heterochromatin by CENP-B deletion amplifies the differences. Molecular evolution analyses using Murinae genomes identify adaptive evolution in proteins in both pathways. We propose that centromere proteins have recurrently evolved to minimize the kinetochore pathway, which is exploited by selfish DNA, relative to the heterochromatin pathway that equalizes centromeres, while maintaining essential functions.


Subject(s)
Centromere Protein B/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Alleles , Amino Acid Sequence , Animals , Biological Evolution , CRISPR-Cas Systems/genetics , Centromere Protein A/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomes, Mammalian/metabolism , Female , Heterochromatin/metabolism , Kinetochores/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Oocytes/metabolism , Protein Domains
7.
Cell ; 184(11): 2860-2877.e22, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33964210

ABSTRACT

Most human embryos are aneuploid. Aneuploidy frequently arises during the early mitotic divisions of the embryo, but its origin remains elusive. Human zygotes that cluster their nucleoli at the pronuclear interface are thought to be more likely to develop into healthy euploid embryos. Here, we show that the parental genomes cluster with nucleoli in each pronucleus within human and bovine zygotes, and clustering is required for the reliable unification of the parental genomes after fertilization. During migration of intact pronuclei, the parental genomes polarize toward each other in a process driven by centrosomes, dynein, microtubules, and nuclear pore complexes. The maternal and paternal chromosomes eventually cluster at the pronuclear interface, in direct proximity to each other, yet separated. Parental genome clustering ensures the rapid unification of the parental genomes on nuclear envelope breakdown. However, clustering often fails, leading to chromosome segregation errors and micronuclei, incompatible with healthy embryo development.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Development/genetics , Aneuploidy , Animals , Cattle , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Centrosome/metabolism , Chromosome Segregation/physiology , Chromosomes/metabolism , Fertilization/genetics , Humans , Male , Microtubules/metabolism , Mitosis , Oocytes/metabolism , Spermatozoa/metabolism , Zygote/metabolism
8.
Cell ; 184(13): 3528-3541.e12, 2021 06 24.
Article in English | MEDLINE | ID: mdl-33984278

ABSTRACT

Nucleotide-binding, leucine-rich repeat receptors (NLRs) are major immune receptors in plants and animals. Upon activation, the Arabidopsis NLR protein ZAR1 forms a pentameric resistosome in vitro and triggers immune responses and cell death in plants. In this study, we employed single-molecule imaging to show that the activated ZAR1 protein can form pentameric complexes in the plasma membrane. The ZAR1 resistosome displayed ion channel activity in Xenopus oocytes in a manner dependent on a conserved acidic residue Glu11 situated in the channel pore. Pre-assembled ZAR1 resistosome was readily incorporated into planar lipid-bilayers and displayed calcium-permeable cation-selective channel activity. Furthermore, we show that activation of ZAR1 in the plant cell led to Glu11-dependent Ca2+ influx, perturbation of subcellular structures, production of reactive oxygen species, and cell death. The results thus support that the ZAR1 resistosome acts as a calcium-permeable cation channel to trigger immunity and cell death.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/immunology , Arabidopsis/metabolism , Calcium/metabolism , Carrier Proteins/metabolism , Disease Resistance/immunology , Plant Immunity , Signal Transduction , Animals , Cell Death , Cell Membrane/metabolism , Cell Membrane Permeability , Glutamic Acid/metabolism , Lipid Bilayers/metabolism , Oocytes/metabolism , Plant Cells/metabolism , Protein Multimerization , Protoplasts/metabolism , Reactive Oxygen Species/metabolism , Single Molecule Imaging , Vacuoles/metabolism , Xenopus
9.
Annu Rev Biochem ; 89: 135-158, 2020 06 20.
Article in English | MEDLINE | ID: mdl-31815535

ABSTRACT

DNA methylation at the 5-position of cytosine (5mC) plays vital roles in mammalian development. DNA methylation is catalyzed by DNA methyltransferases (DNMTs), and the two DNMT families, DNMT3 and DNMT1, are responsible for methylation establishment and maintenance, respectively. Since their discovery, biochemical and structural studies have revealed the key mechanisms underlying how DNMTs catalyze de novo and maintenance DNA methylation. In particular, recent development of low-input genomic and epigenomic technologies has deepened our understanding of DNA methylation regulation in germ lines and early stage embryos. In this review, we first describe the methylation machinery including the DNMTs and their essential cofactors. We then discuss how DNMTs are recruited to or excluded from certain genomic elements. Lastly, we summarize recent understanding of the regulation of DNA methylation dynamics in mammalian germ lines and early embryos with a focus on both mice and humans.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA/genetics , Gene Expression Regulation, Developmental , Genome , Animals , Coenzymes/chemistry , Coenzymes/metabolism , CpG Islands , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Embryo, Mammalian , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Oocytes/cytology , Oocytes/enzymology , Oocytes/growth & development , Signal Transduction , Spermatozoa/cytology , Spermatozoa/enzymology , Spermatozoa/growth & development
10.
Annu Rev Biochem ; 89: 695-715, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569527

ABSTRACT

The zona pellucida (ZP) is an extracellular matrix that surrounds all mammalian oocytes, eggs, and early embryos and plays vital roles during oogenesis, fertilization, and preimplantation development. The ZP is composed of three or four glycosylated proteins, ZP1-4, that are synthesized, processed, secreted, and assembled into long, cross-linked fibrils by growing oocytes. ZP proteins have an immunoglobulin-like three-dimensional structure and a ZP domain that consists of two subdomains, ZP-N and ZP-C, with ZP-N of ZP2 and ZP3 required for fibril assembly. A ZP2-ZP3 dimer is located periodically along ZP fibrils that are cross-linked by ZP1, a protein with a proline-rich N terminus. Fibrils in the inner and outer regions of the ZP are oriented perpendicular and parallel to the oolemma, respectively, giving the ZP a multilayered appearance. Upon fertilization of eggs, modification of ZP2 and ZP3 results in changes in the ZP's physical and biological properties that have important consequences. Certain structural features of ZP proteins suggest that they may be amyloid-like proteins.


Subject(s)
Amyloidogenic Proteins/chemistry , Zona Pellucida Glycoproteins/chemistry , Zygote/metabolism , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/metabolism , Animals , Embryo, Mammalian/metabolism , Embryo, Mammalian/ultrastructure , Female , Gene Expression Regulation, Developmental , Humans , Oocytes/growth & development , Oocytes/metabolism , Oocytes/ultrastructure , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Zona Pellucida/metabolism , Zona Pellucida/ultrastructure , Zona Pellucida Glycoproteins/genetics , Zona Pellucida Glycoproteins/metabolism , Zygote/growth & development , Zygote/ultrastructure
11.
Annu Rev Biochem ; 89: 255-282, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32259458

ABSTRACT

Facultative heterochromatin (fHC) concerns the developmentally regulated heterochromatinization of different regions of the genome and, in the case of the mammalian X chromosome and imprinted loci, of only one allele of a homologous pair. The formation of fHC participates in the timely repression of genes, by resisting strong trans activators. In this review, we discuss the molecular mechanisms underlying the establishment and maintenance of fHC in mammals using a mouse model. We focus on X-chromosome inactivation (XCI) as a paradigm for fHC but also relate it to genomic imprinting and homeobox (Hox) gene cluster repression. A vital role for noncoding transcription and/or transcripts emerges as the general principle of triggering XCI and canonical imprinting. However, other types of fHC are established through an unknown mechanism, independent of noncoding transcription (Hox clusters and noncanonical imprinting). We also extensively discuss polycomb-group repressive complexes (PRCs), which frequently play a vital role in fHC maintenance.


Subject(s)
Gene Expression Regulation, Developmental , Genomic Imprinting , Heterochromatin/metabolism , Polycomb-Group Proteins/genetics , X Chromosome Inactivation , X Chromosome/metabolism , Animals , Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Female , Gene Silencing , Heterochromatin/chemistry , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Male , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Polycomb-Group Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spermatozoa/cytology , Spermatozoa/growth & development , Spermatozoa/metabolism , X Chromosome/chemistry
12.
Nat Rev Mol Cell Biol ; 24(1): 27-44, 2023 01.
Article in English | MEDLINE | ID: mdl-36068367

ABSTRACT

During fertilization, the egg and the sperm are supposed to contribute precisely one copy of each chromosome to the embryo. However, human eggs frequently contain an incorrect number of chromosomes - a condition termed aneuploidy, which is much more prevalent in eggs than in either sperm or in most somatic cells. In turn, aneuploidy in eggs is a leading cause of infertility, miscarriage and congenital syndromes. Aneuploidy arises as a consequence of aberrant meiosis during egg development from its progenitor cell, the oocyte. In human oocytes, chromosomes often segregate incorrectly. Chromosome segregation errors increase in women from their mid-thirties, leading to even higher levels of aneuploidy in eggs from women of advanced maternal age, ultimately causing age-related infertility. Here, we cover the two main areas that contribute to aneuploidy: (1) factors that influence the fidelity of chromosome segregation in eggs of women from all ages and (2) factors that change in response to reproductive ageing. Recent discoveries reveal new error-causing pathways and present a framework for therapeutic strategies to extend the span of female fertility.


Subject(s)
Infertility , Semen , Animals , Female , Male , Humans , Oocytes/metabolism , Aneuploidy , Meiosis , Aging/genetics , Chromosome Segregation/genetics , Infertility/metabolism , Mammals
13.
Cell ; 180(6): 1212-1227.e14, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32169215

ABSTRACT

The paternal genome undergoes a massive exchange of histone with protamine for compaction into sperm during spermiogenesis. Upon fertilization, this process is potently reversed, which is essential for parental genome reprogramming and subsequent activation; however, it remains poorly understood how this fundamental process is initiated and regulated. Here, we report that the previously characterized splicing kinase SRPK1 initiates this life-beginning event by catalyzing site-specific phosphorylation of protamine, thereby triggering protamine-to-histone exchange in the fertilized oocyte. Interestingly, protamine undergoes a DNA-dependent phase transition to gel-like condensates and SRPK1-mediated phosphorylation likely helps open up such structures to enhance protamine dismissal by nucleoplasmin (NPM2) and enable the recruitment of HIRA for H3.3 deposition. Remarkably, genome-wide assay for transposase-accessible chromatin sequencing (ATAC-seq) analysis reveals that selective chromatin accessibility in both sperm and MII oocytes is largely erased in early pronuclei in a protamine phosphorylation-dependent manner, suggesting that SRPK1-catalyzed phosphorylation initiates a highly synchronized reorganization program in both parental genomes.


Subject(s)
Chromatin/metabolism , Protamines/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Chromatin/physiology , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Fertilization/genetics , Histones/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oocytes/metabolism , Oocytes/physiology , Phosphorylation , Protamine Kinase/genetics , Protamine Kinase/metabolism , Protamines/genetics , Protein Serine-Threonine Kinases/physiology , RNA Splicing/genetics , RNA Splicing/physiology , Spermatozoa/metabolism , Transcription Factors/metabolism , Zygote/metabolism
14.
Cell ; 180(3): 585-600.e19, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32004457

ABSTRACT

Molecular mechanisms of ovarian aging and female age-related fertility decline remain unclear. We surveyed the single-cell transcriptomic landscape of ovaries from young and aged non-human primates (NHPs) and identified seven ovarian cell types with distinct gene-expression signatures, including oocyte and six types of ovarian somatic cells. In-depth dissection of gene-expression dynamics of oocytes revealed four subtypes at sequential and stepwise developmental stages. Further analysis of cell-type-specific aging-associated transcriptional changes uncovered the disturbance of antioxidant signaling specific to early-stage oocytes and granulosa cells, indicative of oxidative damage as a crucial factor in ovarian functional decline with age. Additionally, inactivated antioxidative pathways, increased reactive oxygen species, and apoptosis were observed in granulosa cells from aged women. This study provides a comprehensive understanding of the cell-type-specific mechanisms underlying primate ovarian aging at single-cell resolution, revealing new diagnostic biomarkers and potential therapeutic targets for age-related human ovarian disorders.


Subject(s)
Aging/genetics , Ovary/physiology , Single-Cell Analysis/methods , Transcriptome , Aged , Animals , Antioxidants/metabolism , Apoptosis/physiology , Atlases as Topic , Biomarkers , Cell Line, Tumor , Female , Granulosa Cells/metabolism , Humans , Macaca fascicularis , Oocytes/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
15.
Cell ; 182(1): 127-144.e23, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32502394

ABSTRACT

Before zygotic genome activation (ZGA), the quiescent genome undergoes reprogramming to transition into the transcriptionally active state. However, the mechanisms underlying euchromatin establishment during early embryogenesis remain poorly understood. Here, we show that histone H4 lysine 16 acetylation (H4K16ac) is maintained from oocytes to fertilized embryos in Drosophila and mammals. H4K16ac forms large domains that control nucleosome accessibility of promoters prior to ZGA in flies. Maternal depletion of MOF acetyltransferase leading to H4K16ac loss causes aberrant RNA Pol II recruitment, compromises the 3D organization of the active genomic compartments during ZGA, and causes downregulation of post-zygotically expressed genes. Germline depletion of histone deacetylases revealed that other acetyl marks cannot compensate for H4K16ac loss in the oocyte. Moreover, zygotic re-expression of MOF was neither able to restore embryonic viability nor onset of X chromosome dosage compensation. Thus, maternal H4K16ac provides an instructive function to the offspring, priming future gene activation.


Subject(s)
Histones/metabolism , Lysine/metabolism , Transcriptional Activation/genetics , Acetylation , Animals , Base Sequence , Chromosome Segregation/genetics , Conserved Sequence , Dosage Compensation, Genetic , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , Evolution, Molecular , Female , Genome , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Male , Mammals/genetics , Mice , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Oocytes/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , X Chromosome/metabolism , Zygote/metabolism
16.
Cell ; 178(5): 1132-1144.e10, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31402175

ABSTRACT

Asymmetric division in female meiosis creates selective pressure favoring selfish centromeres that bias their transmission to the egg. This centromere drive can explain the paradoxical rapid evolution of both centromere DNA and centromere-binding proteins despite conserved centromere function. Here, we define a molecular pathway linking expanded centromeres to histone phosphorylation and recruitment of microtubule destabilizing factors, leading to detachment of selfish centromeres from spindle microtubules that would direct them to the polar body. Exploiting centromere divergence between species, we show that selfish centromeres in two hybrid mouse models use the same molecular pathway but modulate it differently to enrich destabilizing factors. Our results indicate that increasing microtubule destabilizing activity is a general strategy for drive in both models, but centromeres have evolved distinct mechanisms to increase that activity. Furthermore, we show that drive depends on slowing meiotic progression, suggesting that selfish centromeres can be suppressed by regulating meiotic timing.


Subject(s)
Centromere/genetics , Meiosis , Animals , Chromosome Segregation , Female , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Oocytes/metabolism , Protein Serine-Threonine Kinases/metabolism
17.
Cell ; 176(6): 1379-1392.e14, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30773315

ABSTRACT

Cell fate specification by lateral inhibition typically involves contact signaling through the Delta-Notch signaling pathway. However, whether this is the only signaling mode mediating lateral inhibition remains unclear. Here we show that in zebrafish oogenesis, a group of cells within the granulosa cell layer at the oocyte animal pole acquire elevated levels of the transcriptional coactivator TAZ in their nuclei. One of these cells, the future micropyle precursor cell (MPC), accumulates increasingly high levels of nuclear TAZ and grows faster than its surrounding cells, mechanically compressing those cells, which ultimately lose TAZ from their nuclei. Strikingly, relieving neighbor-cell compression by MPC ablation or aspiration restores nuclear TAZ accumulation in neighboring cells, eventually leading to MPC re-specification from these cells. Conversely, MPC specification is defective in taz-/- follicles. These findings uncover a novel mode of lateral inhibition in cell fate specification based on mechanical signals controlling TAZ activity.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Oogenesis/physiology , Zebrafish Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Communication/physiology , Cell Differentiation/physiology , Cell Lineage , Cell Nucleus/metabolism , Female , Granulosa Cells/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Oocytes/metabolism , Oocytes/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcriptional Activation/physiology , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors
18.
Cell ; 177(6): 1463-1479.e18, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31080065

ABSTRACT

Segregation of maternal determinants within the oocyte constitutes the first step in embryo patterning. In zebrafish oocytes, extensive ooplasmic streaming leads to the segregation of ooplasm from yolk granules along the animal-vegetal axis of the oocyte. Here, we show that this process does not rely on cortical actin reorganization, as previously thought, but instead on a cell-cycle-dependent bulk actin polymerization wave traveling from the animal to the vegetal pole of the oocyte. This wave functions in segregation by both pulling ooplasm animally and pushing yolk granules vegetally. Using biophysical experimentation and theory, we show that ooplasm pulling is mediated by bulk actin network flows exerting friction forces on the ooplasm, while yolk granule pushing is achieved by a mechanism closely resembling actin comet formation on yolk granules. Our study defines a novel role of cell-cycle-controlled bulk actin polymerization waves in oocyte polarization via ooplasmic segregation.


Subject(s)
Actins/metabolism , Cell Cycle/physiology , Oocytes/metabolism , Actins/physiology , Animals , Cell Polarity/physiology , Cytoplasm/metabolism , Egg Yolk/physiology , Polymerization , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zygote
19.
Cell ; 172(4): 647-649, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29425487

ABSTRACT

In this issue of Cell, Liu et al. (2018) report the birth of two healthy cloned macaque monkeys using fetal fibroblasts. By artificially enhancing the arsenal of epigenetic modifiers in the oocyte, the authors overcome the earliest roadblocks that take place during somatic cell nuclear transfer (SCNT).


Subject(s)
Haplorhini , Macaca , Animals , Cloning, Organism , Fibroblasts , Nuclear Transfer Techniques , Oocytes , Primates
20.
Cell ; 174(5): 1082-1094.e12, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30057117

ABSTRACT

Although animals have evolved multiple mechanisms to suppress transposons, "leaky" mobilizations that cause mutations and diseases still occur. This suggests that transposons employ specific tactics to accomplish robust propagation. By directly tracking mobilization, we show that, during a short and specific time window of oogenesis, retrotransposons achieve massive amplification via a cell-type-specific targeting strategy. Retrotransposons rarely mobilize in undifferentiated germline stem cells. However, as oogenesis proceeds, they utilize supporting nurse cells-which are highly polyploid and eventually undergo apoptosis-as factories to massively manufacture invading products. Moreover, retrotransposons rarely integrate into nurse cells themselves but, instead, via microtubule-mediated transport, they preferentially target the DNA of the interconnected oocytes. Blocking microtubule-dependent intercellular transport from nurse cells significantly alleviates damage to the oocyte genome. Our data reveal that parasitic genomic elements can efficiently hijack a host developmental process to propagate robustly, thereby driving evolutionary change and causing disease.


Subject(s)
Drosophila melanogaster/genetics , Long Interspersed Nucleotide Elements , Oogenesis , RNA, Small Interfering , Retroelements , Retroviridae/genetics , Animals , Drosophila Proteins , Female , Gene Library , Gene Silencing , Germ Cells , Green Fluorescent Proteins/metabolism , In Situ Hybridization, Fluorescence , Male , Oocytes/metabolism , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL