Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Struct Mol Biol ; 30(2): 188-199, 2023 02.
Article in English | MEDLINE | ID: mdl-36635604

ABSTRACT

In meiosis, a supramolecular protein structure, the synaptonemal complex (SC), assembles between homologous chromosomes to facilitate their recombination. Mammalian SC formation is thought to involve hierarchical zipper-like assembly of an SYCP1 protein lattice that recruits stabilizing central element (CE) proteins as it extends. Here we combine biochemical approaches with separation-of-function mutagenesis in mice to show that, rather than stabilizing the SYCP1 lattice, the CE protein SYCE3 actively remodels this structure during synapsis. We find that SYCP1 tetramers undergo conformational change into 2:1 heterotrimers on SYCE3 binding, removing their assembly interfaces and disrupting the SYCP1 lattice. SYCE3 then establishes a new lattice by its self-assembly mimicking the role of the disrupted interface in tethering together SYCP1 dimers. SYCE3 also interacts with CE complexes SYCE1-SIX6OS1 and SYCE2-TEX12, providing a mechanism for their recruitment. Thus, SYCE3 remodels the SYCP1 lattice into a CE-binding integrated SYCP1-SYCE3 lattice to achieve long-range synapsis by a mature SC.


Subject(s)
Chromosome Pairing , Synaptonemal Complex , Animals , Mice , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , Mammals/genetics , Meiosis , Nuclear Proteins/metabolism , Synaptonemal Complex/metabolism
2.
J Cell Biol ; 219(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32232464

ABSTRACT

Age-dependent oocyte aneuploidy, a major cause of Down syndrome, is associated with declining sister chromatid cohesion in postnatal oocytes. Here we show that cohesion in postnatal mouse oocytes is regulated by Tex19.1. We show Tex19.1-/- oocytes have defects maintaining chiasmata, missegregate their chromosomes during meiosis, and transmit aneuploidies to the next generation. Furthermore, we show that mouse Tex19.1 inhibits N-end rule protein degradation mediated by its interacting partner UBR2, and that Ubr2 itself has a previously undescribed role in negatively regulating the acetylated SMC3 subpopulation of cohesin in mitotic somatic cells. Lastly, we show that acetylated SMC3 is associated with meiotic chromosome axes in mouse oocytes, and that this population of cohesin is specifically depleted in the absence of Tex19.1. These findings indicate that Tex19.1 regulates UBR protein activity to maintain acetylated SMC3 and sister chromatid cohesion in postnatal oocytes and prevent aneuploidy from arising in the female germline.


Subject(s)
Cell Cycle Proteins/genetics , Chondroitin Sulfate Proteoglycans/genetics , Chromosomal Proteins, Non-Histone/genetics , RNA-Binding Proteins/genetics , Sister Chromatid Exchange/genetics , Ubiquitin-Protein Ligases/genetics , Aneuploidy , Animals , Cell Lineage/genetics , Chromatids/genetics , Chromosome Segregation/genetics , Female , Germ Cells/growth & development , Humans , Meiosis/genetics , Mice , Mice, Knockout , Oocytes/growth & development , Oocytes/metabolism , Cohesins
3.
FASEB J ; 33(12): 14221-14233, 2019 12.
Article in English | MEDLINE | ID: mdl-31659914

ABSTRACT

Deleted in azoospermia-like (DAZL) is a germ cell RNA-binding protein that is essential for entry and progression through meiosis. The phenotype of the Dazl knockout mouse has extensive germ cell loss because of incomplete meiosis. We have created a Dazl hypomorph model using short interfering RNA knockdown in mouse fetal ovary cultures, allowing investigation of Dazl function in germ cell maturation. Dazl hypomorph ovaries had a phenotype of impaired germ cell nest breakdown with a 66% reduction in total follicle number and an increase in the proportion of primordial follicles (PMFs), with smaller oocytes within these follicles. There was no significant early germ cell loss or meiotic delay. Immunostaining of intercellular bridge component testis-expressed protein (Tex)14 showed ∼59% reduction in foci number and size, without any change in Tex14 mRNA levels. TEX14 expression was also confirmed in the human fetal ovary across gestation. Using 3'UTR-luciferase reporter assays, translational regulation of TEX14 was demonstrated to be DAZL-dependant. Dazl is therefore essential for normal intercellular bridges within germ cell nests and their timely breakdown, with a major impact on subsequent assembly of PMFs.-Rosario, R., Crichton, J. H., Stewart, H. L., Childs, A. J., Adams, I. R., Anderson, R. A. Dazl determines primordial follicle formation through the translational regulation of Tex14.


Subject(s)
Ovary/growth & development , Ovary/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Cloning, Molecular , Female , Gene Expression Regulation, Developmental , Humans , Meiosis/physiology , Mice , RNA Interference , RNA, Messenger , RNA-Binding Proteins/genetics , Tissue Culture Techniques , Transcription Factors/genetics
4.
Elife ; 82019 01 24.
Article in English | MEDLINE | ID: mdl-30674417

ABSTRACT

Male germ cells of all placental mammals express an ancient nuclear RNA binding protein of unknown function called RBMXL2. Here we find that deletion of the retrogene encoding RBMXL2 blocks spermatogenesis. Transcriptome analyses of age-matched deletion mice show that RBMXL2 controls splicing patterns during meiosis. In particular, RBMXL2 represses the selection of aberrant splice sites and the insertion of cryptic and premature terminal exons. Our data suggest a Rbmxl2 retrogene has been conserved across mammals as part of a splicing control mechanism that is fundamentally important to germ cell biology. We propose that this mechanism is essential to meiosis because it buffers the high ambient concentrations of splicing activators, thereby preventing poisoning of key transcripts and disruption to gene expression by aberrant splice site selection.


Subject(s)
Germ Cells/metabolism , RNA Splice Sites/genetics , RNA-Binding Proteins/metabolism , Animals , Exons/genetics , Fertility , Gene Expression Regulation, Developmental , Male , Meiosis/genetics , Metaphase/genetics , Mice, Inbred C57BL , Models, Animal , Organ Specificity , RNA Splicing/genetics , Testis/metabolism
5.
Chromosoma ; 127(4): 437-459, 2018 12.
Article in English | MEDLINE | ID: mdl-29907896

ABSTRACT

Recombination, synapsis, chromosome segregation and gene expression are co-ordinately regulated during meiosis to ensure successful execution of this specialised cell division. Studies with multiple mutant mouse lines have shown that mouse spermatocytes possess quality control checkpoints that eliminate cells with persistent defects in chromosome synapsis. In addition, studies on Trip13mod/mod mice suggest that pachytene spermatocytes that successfully complete chromosome synapsis can undergo meiotic arrest in response to defects in recombination. Here, we present additional support for a meiotic recombination-dependent checkpoint using a different mutant mouse line, Tex19.1-/-. The appearance of early recombination foci is delayed in Tex19.1-/- spermatocytes during leptotene/zygotene, but some Tex19.1-/- spermatocytes still successfully synapse their chromosomes and we show that these spermatocytes are enriched for early recombination foci. Furthermore, we show that patterns of axis elongation, chromatin modifications and histone H1t expression are also all co-ordinately skewed towards earlier substages of pachytene in these autosomally synapsed Tex19.1-/- spermatocytes. We also show that this skew towards earlier pachytene substages occurs in the absence of elevated spermatocyte death in the population, that spermatocytes with features of early pachytene are present in late stage Tex19.1-/- testis tubules and that the delay in histone H1t expression in response to loss of Tex19.1 does not occur in a Spo11 mutant background. Taken together, these data suggest that a recombination-dependent checkpoint may be able to modulate pachytene progression in mouse spermatocytes to accommodate some types of recombination defect.


Subject(s)
Nuclear Proteins/genetics , Pachytene Stage/genetics , Recombination, Genetic , Spermatocytes/cytology , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Cell Cycle Proteins/genetics , Chromatin/genetics , Chromatin/metabolism , Chromosome Pairing/genetics , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Histones/genetics , Histones/metabolism , Male , Meiosis , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational/genetics , RNA-Binding Proteins , Seminiferous Tubules/metabolism , Seminiferous Tubules/pathology , Spermatocytes/physiology
6.
PLoS Genet ; 13(7): e1006904, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28708824

ABSTRACT

Meiosis relies on the SPO11 endonuclease to generate the recombinogenic DNA double strand breaks (DSBs) required for homologous chromosome synapsis and segregation. The number of meiotic DSBs needs to be sufficient to allow chromosomes to search for and find their homologs, but not excessive to the point of causing genome instability. Here we report that the mammal-specific gene Tex19.1 promotes Spo11-dependent recombination in mouse spermatocytes. We show that the chromosome asynapsis previously reported in Tex19.1-/- spermatocytes is preceded by reduced numbers of recombination foci in leptotene and zygotene. Tex19.1 is required for normal levels of early Spo11-dependent recombination foci during leptotene, but not for upstream events such as MEI4 foci formation or accumulation of H3K4me3 at recombination hotspots. Furthermore, we show that mice carrying mutations in Ubr2, which encodes an E3 ubiquitin ligase that interacts with TEX19.1, phenocopy the Tex19.1-/- recombination defects. These data suggest that Tex19.1 and Ubr2 are required for mouse spermatocytes to accumulate sufficient Spo11-dependent recombination to ensure that the homology search is consistently successful, and reveal a hitherto unknown genetic pathway promoting meiotic recombination in mammals.


Subject(s)
Endodeoxyribonucleases/metabolism , Meiosis/genetics , Nuclear Proteins/metabolism , Spermatocytes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Chromosome Pairing , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , Endodeoxyribonucleases/genetics , Male , Meiotic Prophase I/genetics , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA-Binding Proteins , Recombination, Genetic , Ubiquitin-Protein Ligases/genetics
7.
Semin Cell Dev Biol ; 45: 68-76, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26454098

ABSTRACT

Meiosis is one of the defining events in gametogenesis. Male and female germ cells both undergo one round of meiotic cell division during their development in order to reduce the ploidy of the gametes, and thereby maintain the ploidy of the species after fertilisation. However, there are some aspects of meiosis in the female germline, such as the prolonged arrest in dictyate, that appear to predispose oocytes to missegregate their chromosomes and transmit aneuploidies to the next generation. These maternally-derived aneuploidies are particularly problematic in humans where they are major contributors to miscarriage, age-related infertility, and the high incidence of Down's syndrome in human conceptions. This review will discuss how events that occur in foetal oocyte development and during the oocytes' prolonged dictyate arrest can influence meiotic chromosome segregation and the incidence of aneuploidy in adult oocytes.


Subject(s)
Meiosis , Oocytes/physiology , Trisomy , Animals , Chromosome Segregation , Crossing Over, Genetic , Female , Humans , Oogenesis , Recombination, Genetic
8.
J Genet Genomics ; 41(3): 97-106, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24656230

ABSTRACT

Meiosis is a key event in gametogenesis that generates new combinations of genetic information and is required to reduce the chromosome content of the gametes. Meiotic chromosomes undergo a number of specialised events during prophase to allow meiotic recombination, homologous chromosome synapsis and reductional chromosome segregation to occur. In mammalian cells, DNA physically associates with histones to form chromatin, which can be modified by methylation, phosphorylation, ubiquitination and acetylation to help regulate higher order chromatin structure, gene expression, and chromosome organisation. Recent studies have identified some of the enzymes responsible for generating chromatin modifications in meiotic mammalian cells, and shown that these chromatin modifying enzymes are required for key meiosis-specific events that occur during meiotic prophase. This review will discuss the role of chromatin modifications in meiotic recombination, homologous chromosome synapsis and regulation of meiotic gene expression in mammals.


Subject(s)
Chromatin/metabolism , Prophase/physiology , Acetylation , Animals , Centromere/physiology , DNA/metabolism , Gene Expression , Histones/metabolism , Methylation , Methyltransferases/metabolism , Mice , Phosphorylation , Polycomb-Group Proteins/metabolism , Ubiquitination
9.
Cell Mol Life Sci ; 71(9): 1581-605, 2014 May.
Article in English | MEDLINE | ID: mdl-24045705

ABSTRACT

The viability of any species requires that the genome is kept stable as it is transmitted from generation to generation by the germ cells. One of the challenges to transgenerational genome stability is the potential mutagenic activity of transposable genetic elements, particularly retrotransposons. There are many different types of retrotransposon in mammalian genomes, and these target different points in germline development to amplify and integrate into new genomic locations. Germ cells, and their pluripotent developmental precursors, have evolved a variety of genome defence mechanisms that suppress retrotransposon activity and maintain genome stability across the generations. Here, we review recent advances in understanding how retrotransposon activity is suppressed in the mammalian germline, how genes involved in germline genome defence mechanisms are regulated, and the consequences of mutating these genome defence genes for the developing germline.


Subject(s)
Genome , Germ Cells/metabolism , Retroelements/genetics , Animals , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Germ Cells/cytology , Humans , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA, Small Interfering/metabolism
10.
PLoS Comput Biol ; 8(4): e1002486, 2012.
Article in English | MEDLINE | ID: mdl-22570599

ABSTRACT

Retrotransposons are highly prevalent in mammalian genomes due to their ability to amplify in pluripotent cells or developing germ cells. Host mechanisms that silence retrotransposons in germ cells and pluripotent cells are important for limiting the accumulation of the repetitive elements in the genome during evolution. However, although silencing of selected individual retrotransposons can be relatively well-studied, many mammalian retrotransposons are seldom analysed and their silencing in germ cells, pluripotent cells or somatic cells remains poorly understood. Here we show, and experimentally verify, that cryptic repetitive element probes present in Illumina and Affymetrix gene expression microarray platforms can accurately and sensitively monitor repetitive element expression data. This computational approach to genome-wide retrotransposon expression has allowed us to identify the histone deacetylase Hdac1 as a component of the retrotransposon silencing machinery in mouse embryonic stem cells, and to determine the retrotransposon targets of Hdac1 in these cells. We also identify retrotransposons that are targets of other retrotransposon silencing mechanisms such as DNA methylation, Eset-mediated histone modification, and Ring1B/Eed-containing polycomb repressive complexes in mouse embryonic stem cells. Furthermore, our computational analysis of retrotransposon silencing suggests that multiple silencing mechanisms are independently targeted to retrotransposons in embryonic stem cells, that different genomic copies of the same retrotransposon can be differentially sensitive to these silencing mechanisms, and helps define retrotransposon sequence elements that are targeted by silencing machineries. Thus repeat annotation of gene expression microarray data suggests that a complex interplay between silencing mechanisms represses retrotransposon loci in germ cells and embryonic stem cells.


Subject(s)
Embryonic Stem Cells/physiology , Gene Expression Profiling/methods , Histone Deacetylase 1/genetics , Interspersed Repetitive Sequences/genetics , Oligonucleotide Array Sequence Analysis/methods , Retroelements/genetics , Animals , Cells, Cultured , Gene Expression Regulation, Developmental/genetics , Gene Silencing/physiology , Mice , Regulatory Sequences, Nucleic Acid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...