Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters











Publication year range
1.
Elife ; 112022 02 08.
Article in English | MEDLINE | ID: mdl-35133274

ABSTRACT

DNA damage response mechanisms have meiotic roles that ensure successful gamete formation. While completion of meiotic double-strand break (DSB) repair requires the canonical RAD9A-RAD1-HUS1 (9A-1-1) complex, mammalian meiocytes also express RAD9A and HUS1 paralogs, RAD9B and HUS1B, predicted to form alternative 9-1-1 complexes. The RAD1 subunit is shared by all predicted 9-1-1 complexes and localizes to meiotic chromosomes even in the absence of HUS1 and RAD9A. Here, we report that testis-specific disruption of RAD1 in mice resulted in impaired DSB repair, germ cell depletion, and infertility. Unlike Hus1 or Rad9a disruption, Rad1 loss in meiocytes also caused severe defects in homolog synapsis, impaired phosphorylation of ATR targets such as H2AX, CHK1, and HORMAD2, and compromised meiotic sex chromosome inactivation. Together, these results establish critical roles for both canonical and alternative 9-1-1 complexes in meiotic ATR activation and successful prophase I completion.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Chromosome Pairing , DNA Repair , Meiosis , Animals , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Male , Mice , Mice, Transgenic , Signal Transduction , Testis/metabolism
2.
Nat Commun ; 12(1): 6926, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34862376

ABSTRACT

Animals are essential genetic tools in scientific research and global resources in agriculture. In both arenas, a single sex is often required in surplus. The ethical and financial burden of producing and culling animals of the undesired sex is considerable. Using the mouse as a model, we develop a synthetic lethal, bicomponent CRISPR-Cas9 strategy that produces male- or female-only litters with one hundred percent efficiency. Strikingly, we observe a degree of litter size compensation relative to control matings, indicating that our system has the potential to increase the yield of the desired sex in comparison to standard breeding designs. The bicomponent system can also be repurposed to generate postnatal sex-specific phenotypes. Our approach, harnessing the technological applications of CRISPR-Cas9, may be applicable to other vertebrate species, and provides strides towards ethical improvements for laboratory research and agriculture.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Sex Determination Processes/genetics , Animal Husbandry , Animals , Female , Litter Size/genetics , Male , Mice , Mice, Transgenic , Models, Animal , Pregnancy , Selective Breeding , Synthetic Lethal Mutations
4.
Nature ; 587(7832): E1, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33067604

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
6.
Annu Rev Cell Dev Biol ; 36: 411-440, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33021826

ABSTRACT

Understanding human embryology has historically relied on comparative approaches using mammalian model organisms. With the advent of low-input methods to investigate genetic and epigenetic mechanisms and efficient techniques to assess gene function, we can now study the human embryo directly. These advances have transformed the investigation of early embryogenesis in nonrodent species, thereby providing a broader understanding of conserved and divergent mechanisms. Here, we present an overview of the major events in human preimplantation development and place them in the context of mammalian evolution by comparing these events in other eutherian and metatherian species. We describe the advances of studies on postimplantation development and discuss stem cell models that mimic postimplantation embryos. A comparative perspective highlights the importance of analyzing different organisms with molecular characterization and functional studies to reveal the principles of early development. This growing field has a fundamental impact in regenerative medicine and raises important ethical considerations.


Subject(s)
Embryonic Development , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Models, Biological , Phylogeny , Zygote/metabolism
7.
Nature ; 586(7830): 612-617, 2020 10.
Article in English | MEDLINE | ID: mdl-32814901

ABSTRACT

Single-cell RNA sequencing of embryos can resolve the transcriptional landscape of development at unprecedented resolution. To date, single-cell RNA-sequencing studies of mammalian embryos have focused exclusively on eutherian species. Analysis of mammalian outgroups has the potential to identify deeply conserved lineage specification and pluripotency factors, and can extend our understanding of X dosage compensation. Metatherian (marsupial) mammals diverged from eutherians around 160 million years ago. They exhibit distinctive developmental features, including late implantation1 and imprinted X chromosome inactivation2, which is associated with expression of the XIST-like noncoding RNA RSX3. Here we perform a single-cell RNA-sequencing analysis of embryogenesis and X chromosome inactivation in a marsupial, the grey short-tailed opossum (Monodelphis domestica). We resolve the developmental trajectory and transcriptional signatures of the epiblast, primitive endoderm and trophectoderm, and identify deeply conserved lineage-specific markers that pre-date the eutherian-marsupial divergence. RSX coating and inactivation of the X chromosome occurs early and rapidly. This observation supports the hypothesis that-in organisms with early X chromosome inactivation-imprinted X chromosome inactivation prevents biallelic X silencing. We identify XSR, an RSX antisense transcript expressed from the active X chromosome, as a candidate for the regulator of imprinted X chromosome inactivation. Our datasets provide insights into the evolution of mammalian embryogenesis and X dosage compensation.


Subject(s)
Embryo, Mammalian/cytology , Embryonic Development/genetics , Monodelphis/embryology , Monodelphis/genetics , Single-Cell Analysis , Transcriptome/genetics , X Chromosome Inactivation/genetics , Animals , Cell Lineage/genetics , Embryo, Mammalian/embryology , Female , Germ Layers/cytology , Germ Layers/embryology , Male , Monodelphis/classification , RNA, Antisense/genetics , RNA, Untranslated/genetics , Up-Regulation , X Chromosome/genetics
8.
Nat Commun ; 11(1): 2598, 2020 05 25.
Article in English | MEDLINE | ID: mdl-32451402

ABSTRACT

DNA double-strand breaks (DSBs) are toxic to mammalian cells. However, during meiosis, more than 200 DSBs are generated deliberately, to ensure reciprocal recombination and orderly segregation of homologous chromosomes. If left unrepaired, meiotic DSBs can cause aneuploidy in gametes and compromise viability in offspring. Oocytes in which DSBs persist are therefore eliminated by the DNA-damage checkpoint. Here we show that the DNA-damage checkpoint eliminates oocytes via the pro-apoptotic BCL-2 pathway members Puma, Noxa and Bax. Deletion of these factors prevents oocyte elimination in recombination-repair mutants, even when the abundance of unresolved DSBs is high. Remarkably, surviving oocytes can extrude a polar body and be fertilised, despite chaotic chromosome segregation at the first meiotic division. Our findings raise the possibility that allelic variants of the BCL-2 pathway could influence the risk of embryonic aneuploidy.


Subject(s)
Mutation , Oocytes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinational DNA Repair/genetics , Aneuploidy , Animals , Apoptosis , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Segregation , DNA Breaks, Double-Stranded , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Female , Fertilization , Genes, bcl-2 , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Phosphate-Binding Proteins/deficiency , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
9.
Reproduction ; 159(4): X1, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32065737

ABSTRACT

The journal and the authors apologise for an error in the above titled article published in this journal (vol 144, pp 433­445). The authors inadvertently presented duplicate sperm images for XY and XESxrbO mouse testes of Fig. 6 (bottom panels). This error does not change the findings of the paper, as this figure does not give a quantitative breakdown of the proportions of different shapes.

10.
Nat Commun ; 11(1): 764, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034154

ABSTRACT

Our understanding of the signalling pathways regulating early human development is limited, despite their fundamental biological importance. Here, we mine transcriptomics datasets to investigate signalling in the human embryo and identify expression for the insulin and insulin growth factor 1 (IGF1) receptors, along with IGF1 ligand. Consequently, we generate a minimal chemically-defined culture medium in which IGF1 together with Activin maintain self-renewal in the absence of fibroblast growth factor (FGF) signalling. Under these conditions, we derive several pluripotent stem cell lines that express pluripotency-associated genes, retain high viability and a normal karyotype, and can be genetically modified or differentiated into multiple cell lineages. We also identify active phosphoinositide 3-kinase (PI3K)/AKT/mTOR signalling in early human embryos, and in both primed and naïve pluripotent culture conditions. This demonstrates that signalling insights from human blastocysts can be used to define culture conditions that more closely recapitulate the embryonic niche.


Subject(s)
Cell Self Renewal/physiology , Human Embryonic Stem Cells/metabolism , Insulin-Like Growth Factor I/metabolism , Activins/metabolism , Animals , Blastocyst/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Culture Media/chemistry , Culture Media/metabolism , Culture Media/pharmacology , Endoderm/cytology , Endoderm/metabolism , Extraembryonic Membranes/cytology , Extraembryonic Membranes/metabolism , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transcriptome , X Chromosome Inactivation/physiology
11.
Genome Biol ; 20(1): 160, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31399122

ABSTRACT

Following publication of the original article [1], the following error was reported: The actin control panel in Fig. 3 of this paper is reproduced from Fig. 7 of Touré et al, 2004 [2] by kind permission of the Genetics Society of America. Touré et al, 2004 used Northern blotting to show that the Y-linked genes Ssty1 and Ssty2 have reduced expression in a range of mouse genotypes with deletions on the Y chromosome long arm. This paper shows that two novel genes, Sly and Asty are also present on mouse Yq and have reduced expression in these deleted genotypes. A further companion paper was published in Human Molecular Genetics (Ellis et al, 2005 [3]) showing that X-linked genes are upregulated in the various deleted genotypes. Since two of the genotypes concerned are sterile and very hard to generate, all the Northern blot experiments in these papers were performed on a single membrane that was stripped and re-probed with a range of different X- and Y-linked genes. The same beta-actin loading control image thus necessarily applies to all the data presented, and was shown in all three papers. We regret that this was not mentioned appropriately in the Methods and figure legends at the time of publication.

12.
PLoS Genet ; 15(7): e1008290, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31329581

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pgen.1002900.].

13.
Dev Cell ; 47(5): 645-659.e6, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30393076

ABSTRACT

Meiotic synapsis and recombination ensure correct homologous segregation and genetic diversity. Asynapsed homologs are transcriptionally inactivated by meiotic silencing, which serves a surveillance function and in males drives meiotic sex chromosome inactivation. Silencing depends on the DNA damage response (DDR) network, but how DDR proteins engage repressive chromatin marks is unknown. We identify the histone H3-lysine-9 methyltransferase SETDB1 as the bridge linking the DDR to silencing in male mice. At the onset of silencing, X chromosome H3K9 trimethylation (H3K9me3) enrichment is downstream of DDR factors. Without Setdb1, the X chromosome accrues DDR proteins but not H3K9me3. Consequently, sex chromosome remodeling and silencing fail, causing germ cell apoptosis. Our data implicate TRIM28 in linking the DDR to SETDB1 and uncover additional factors with putative meiotic XY-silencing functions. Furthermore, we show that SETDB1 imposes timely expression of meiotic and post-meiotic genes. Setdb1 thus unites the DDR network, asynapsis, and meiotic chromosome silencing.


Subject(s)
Chromosome Pairing , DNA Damage , Gene Silencing , Histone Code , Histone-Lysine N-Methyltransferase/metabolism , Animals , Apoptosis , DNA Repair , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Tripartite Motif-Containing Protein 28/genetics , Tripartite Motif-Containing Protein 28/metabolism
14.
Nat Commun ; 9(1): 2621, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29976923

ABSTRACT

Meiotic cells undergo genetic exchange between homologs through programmed DNA double-strand break (DSB) formation, recombination and synapsis. In mice, the DNA damage-regulated phosphatidylinositol-3-kinase-like kinase (PIKK) ATM regulates all of these processes. However, the meiotic functions of the PIKK ATR have remained elusive, because germline-specific depletion of this kinase is challenging. Here we uncover roles for ATR in male mouse prophase I progression. ATR deletion causes chromosome axis fragmentation and germ cell elimination at mid pachynema. This elimination cannot be rescued by deletion of ATM and the third DNA damage-regulated PIKK, PRKDC, consistent with the existence of a PIKK-independent surveillance mechanism in the mammalian germline. ATR is required for synapsis, in a manner genetically dissociable from DSB formation. ATR also regulates loading of recombinases RAD51 and DMC1 to DSBs and recombination focus dynamics on synapsed and asynapsed chromosomes. Our studies reveal ATR as a critical regulator of mouse meiosis.


Subject(s)
Chromosomes, Mammalian/genetics , DNA Breaks, Double-Stranded , Meiosis/genetics , Spermatocytes/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Pairing/genetics , Chromosomes, Mammalian/metabolism , In Situ Hybridization, Fluorescence , Male , Meiotic Prophase I/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphate-Binding Proteins , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
15.
Science ; 357(6354): 932-935, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28818972

ABSTRACT

Having the correct number of chromosomes is vital for normal development and health. Sex chromosome trisomy affects 0.1% of the human population and is associated with infertility. We show that during reprogramming to induced pluripotent stem cells (iPSCs), fibroblasts from sterile trisomic XXY and XYY mice lose the extra sex chromosome through a phenomenon we term trisomy-biased chromosome loss (TCL). Resulting euploid XY iPSCs can be differentiated into the male germ cell lineage and functional sperm that can be used in intracytoplasmic sperm injection to produce chromosomally normal, fertile offspring. Sex chromosome loss is comparatively infrequent during mouse XX and XY iPSC generation. TCL also applies to other chromosomes, generating euploid iPSCs from cells of a Down syndrome mouse model. It can also create euploid iPSCs from human trisomic patient fibroblasts. The findings have relevance to overcoming infertility and other trisomic phenotypes.


Subject(s)
Cellular Reprogramming Techniques , Fertility/genetics , Induced Pluripotent Stem Cells/physiology , Infertility/therapy , Klinefelter Syndrome/therapy , Sex Chromosome Disorders/therapy , Sex Chromosomes/genetics , Trisomy/genetics , Animals , Cellular Reprogramming , Disease Models, Animal , Down Syndrome/genetics , Female , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Infertility/genetics , Klinefelter Syndrome/genetics , Male , Mice , Mice, Inbred C57BL , Sex Chromosome Disorders/genetics , Sperm Injections, Intracytoplasmic , Spermatozoa/physiology , XYY Karyotype/genetics
16.
Dev Cell ; 40(3): 289-301.e3, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28132849

ABSTRACT

Somatic X dosage compensation requires two mechanisms: X inactivation balances X gene output between males (XY) and females (XX), while X upregulation, hypothesized by Ohno and documented in vivo, balances X gene with autosomal gene output. Whether X dosage compensation occurs in germ cells is unclear. We show that mouse and human germ cells exhibit non-canonical X dosage states that differ from the soma and between the sexes. Prior to genome-wide reprogramming, X upregulation is present, consistent with Ohno's hypothesis. Subsequently, however, it is erased. In females, erasure follows loss of X inactivation, causing X dosage excess. Conversely, in males, erasure leads to permanent X dosage decompensation. Sex chromosomally abnormal models exhibit a "sex-reversed" X dosage state: XX males, like XX females, develop X dosage excess, while XO females, like XY males, develop X dosage decompensation. Thus, germline X dosage compensation states are determined by X chromosome number, not phenotypic sex. These unexpected differences in X dosage compensation states between germline and soma offer unique perspectives on sex chromosome infertility.


Subject(s)
Chromosomes, Human, X/genetics , Dosage Compensation, Genetic , Germ Cells/metabolism , Sex Characteristics , X Chromosome/genetics , Animals , Cellular Reprogramming/genetics , Databases, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation , Germ Cells/cytology , Gonads/cytology , Gonads/metabolism , Humans , Male , Mice , Models, Genetic , Sequence Analysis, RNA , Up-Regulation/genetics
17.
Hum Mol Genet ; 25(24): 5300-5310, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27742779

ABSTRACT

During spermatogenesis, germ cells that fail to synapse their chromosomes or fail to undergo meiotic sex chromosome inactivation (MSCI) are eliminated via apoptosis during mid-pachytene. Previous work showed that Y-linked genes Zfy1 and Zfy2 act as 'executioners' for this checkpoint, and that wrongful expression of either gene during pachytene triggers germ cell death. Here, we show that in mice, Zfy genes are also necessary for efficient MSCI and the sex chromosomes are not correctly silenced in Zfy-deficient spermatocytes. This unexpectedly reveals a triple role for Zfy at the mid-pachytene checkpoint in which Zfy genes first promote MSCI, then monitor its progress (since if MSCI is achieved, Zfy genes will be silenced), and finally execute cells with MSCI failure. This potentially constitutes a negative feedback loop governing this critical checkpoint mechanism.


Subject(s)
DNA-Binding Proteins/genetics , Spermatocytes/metabolism , Transcription Factors/genetics , X Chromosome Inactivation/genetics , Animals , Male , Meiosis/genetics , Mice , Spermatocytes/growth & development , Spermatogenesis/genetics , X Chromosome/genetics
18.
PLoS One ; 11(1): e0145398, 2016.
Article in English | MEDLINE | ID: mdl-26765744

ABSTRACT

A previous study indicated that genetic information encoded on the mouse Y chromosome short arm (Yp) is required for efficient completion of the second meiotic division (that generates haploid round spermatids), restructuring of the sperm head, and development of the sperm tail. Using mouse models lacking a Y chromosome but with varying Yp gene complements provided by Yp chromosomal derivatives or transgenes, we recently identified the Y-encoded zinc finger transcription factors Zfy1 and Zfy2 as the Yp genes promoting the second meiotic division. Using the same mouse models we here show that Zfy2 (but not Zfy1) contributes to the restructuring of the sperm head and is required for the development of the sperm tail. The preferential involvement of Zfy2 is consistent with the presence of an additional strong spermatid-specific promotor that has been acquired by this gene. This is further supported by the fact that promotion of sperm morphogenesis is also seen in one of the two markedly Yp gene deficient models in which a Yp deletion has created a Zfy2/1 fusion gene that is driven by the strong Zfy2 spermatid-specific promotor, but encodes a protein almost identical to that encoded by Zfy1. Our results point to there being further genetic information on Yp that also has a role in restructuring the sperm head.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Sperm Head/metabolism , Sperm Tail/metabolism , Spermatogenesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Y Chromosome/genetics , Animals , Male , Mice , Models, Animal , Morphogenesis/genetics , Physical Chromosome Mapping , Seminiferous Tubules/embryology , Seminiferous Tubules/metabolism , Sperm Head/ultrastructure , Sperm Tail/ultrastructure
19.
PLoS Genet ; 11(10): e1005462, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26509888

ABSTRACT

Chromosome abnormalities are common in the human population, causing germ cell loss at meiotic prophase I and infertility. The mechanisms driving this loss are unknown, but persistent meiotic DNA damage and asynapsis may be triggers. Here we investigate the contribution of these lesions to oocyte elimination in mice with chromosome abnormalities, e.g. Turner syndrome (XO) and translocations. We show that asynapsed chromosomes trigger oocyte elimination at diplonema, which is linked to the presence of phosphorylated H2AFX (γH2AFX). We find that DNA double-strand break (DSB) foci disappear on asynapsed chromosomes during pachynema, excluding persistent DNA damage as a likely cause, and demonstrating the existence in mammalian oocytes of a repair pathway for asynapsis-associated DNA DSBs. Importantly, deletion or point mutation of H2afx restores oocyte numbers in XO females to wild type (XX) levels. Unexpectedly, we find that asynapsed supernumerary chromosomes do not elicit prophase I loss, despite being enriched for γH2AFX and other checkpoint proteins. These results suggest that oocyte loss cannot be explained simply by asynapsis checkpoint models, but is related to the gene content of asynapsed chromosomes. A similar mechanistic basis for oocyte loss may operate in humans with chromosome abnormalities.


Subject(s)
Chromosome Pairing/genetics , Histones/genetics , Oocytes/growth & development , Ovary/growth & development , Animals , Chromosome Aberrations , Chromosome Disorders/genetics , DNA Breaks, Double-Stranded , DNA Damage/genetics , Female , Histones/metabolism , Humans , Male , Meiotic Prophase I/genetics , Mice , Oocytes/metabolism , Ovary/metabolism , Prophase/genetics , X Chromosome/genetics
20.
PLoS Genet ; 10(6): e1004444, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24967676

ABSTRACT

Mouse Zfy1 and Zfy2 encode zinc finger transcription factors that map to the short arm of the Y chromosome (Yp). They have previously been shown to promote meiotic quality control during pachytene (Zfy1 and Zfy2) and at the first meiotic metaphase (Zfy2). However, from these previous studies additional roles for genes encoded on Yp during meiotic progression were inferred. In order to identify these genes and investigate their function in later stages of meiosis, we created three models with diminishing Yp and Zfy gene complements (but lacking the Y-long-arm). Since the Y-long-arm mediates pairing and exchange with the X via their pseudoautosomal regions (PARs) we added a minute PAR-bearing X chromosome derivative to enable formation of a sex bivalent, thus avoiding Zfy2-mediated meiotic metaphase I (MI) checkpoint responses to the unpaired (univalent) X chromosome. Using these models we obtained definitive evidence that genetic information on Yp promotes meiosis II, and by transgene addition identified Zfy1 and Zfy2 as the genes responsible. Zfy2 was substantially more effective and proved to have a much more potent transactivation domain than Zfy1. We previously established that only Zfy2 is required for the robust apoptotic elimination of MI spermatocytes in response to a univalent X; the finding that both genes potentiate meiosis II led us to ask whether there was de novo Zfy1 and Zfy2 transcription in the interphase between meiosis I and meiosis II, and this proved to be the case. X-encoded Zfx was also expressed at this stage and Zfx over-expression also potentiated meiosis II. An interphase between the meiotic divisions is male-specific and we previously hypothesised that this allows meiosis II critical X and Y gene reactivation following sex chromosome silencing in meiotic prophase. The interphase transcription and meiosis II function of Zfx, Zfy1 and Zfy2 validate this hypothesis.


Subject(s)
DNA-Binding Proteins/genetics , Interphase/genetics , Meiosis/genetics , Spermatogenesis/genetics , Transcription Factors/genetics , Animals , Apoptosis/physiology , DNA-Binding Proteins/biosynthesis , Female , Genes, Y-Linked , Kruppel-Like Transcription Factors/genetics , Male , Mice , Spermatocytes/physiology , Transcription Factors/biosynthesis , Transcriptional Activation/genetics , Y Chromosome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL