Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
PLoS Genet ; 14(11): e1007832, 2018 11.
Article in English | MEDLINE | ID: mdl-30496175

ABSTRACT

Meiotic recombination plays a critical role in sexual reproduction by creating crossovers between homologous chromosomes. These crossovers, along with sister chromatid cohesion, connect homologs to enable proper segregation at Meiosis I. Recombination is initiated by programmed double strand breaks (DSBs) at particular regions of the genome. The meiotic recombination checkpoint uses meiosis-specific modifications to the DSB-induced DNA damage response to provide time to convert these breaks into interhomolog crossovers by delaying entry into Meiosis I until the DSBs have been repaired. The meiosis-specific kinase, Mek1, is a key regulator of meiotic recombination pathway choice, as well as being required for the meiotic recombination checkpoint. The major target of this checkpoint is the meiosis-specific transcription factor, Ndt80, which is essential to express genes necessary for completion of recombination and meiotic progression. The molecular mechanism by which cells monitor meiotic DSB repair to allow entry into Meiosis I with unbroken chromosomes was unknown. Using genetic and biochemical approaches, this work demonstrates that in the presence of DSBs, activated Mek1 binds to Ndt80 and phosphorylates the transcription factor, thus inhibiting DNA binding and preventing Ndt80's function as a transcriptional activator. Repair of DSBs by recombination reduces Mek1 activity, resulting in removal of the inhibitory Mek1 phosphates. Phosphorylation of Ndt80 by the meiosis-specific kinase, Ime2, then results in fully activated Ndt80. Ndt80 upregulates transcription of its own gene, as well as target genes, resulting in prophase exit and progression through meiosis.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , MAP Kinase Kinase 1/metabolism , Meiosis/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Cell Cycle Checkpoints , Conserved Sequence , DNA Breaks, Double-Stranded , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Genes, Fungal , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase 1/genetics , Meiosis/genetics , Models, Biological , Models, Molecular , Mutation , Pachytene Stage/genetics , Pachytene Stage/physiology , Phosphorylation , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Recombinational DNA Repair , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Transcription Factors/genetics
2.
Chromosoma ; 125(4): 701-8, 2016 09.
Article in English | MEDLINE | ID: mdl-26661581

ABSTRACT

The XX/XY system is the rule among mammals. However, many exceptions from this general pattern have been discovered since the last decades. One of these non-conventional sex chromosome mechanisms is the multiple sex chromosome system, which is evolutionary fixed among many bat species of the family Phyllostomidae, and has arisen by a translocation between one original gonosome (X or Y chromosome), and an autosome, giving rise to a "neo-XY body." The aim of this work is to study the synaptic behavior and the chromatin remodeling of multiple sex chromosomes in different species of phyllostomid bats using electron microscopy and molecular markers. Testicular tissues from adult males of the species Artibeus lituratus, Artibeus planirostris, Uroderma bilobatum, and Vampyrodes caraccioli from the eastern Amazonia were analyzed by optical/electron microscopy and immunofluorescence of meiotic proteins involved in synapsis (SYCP3 and SYCE3), sister-chromatid cohesion (SMC3), and chromatin silencing (BRCA1, γ-H2AX, and RNApol 2). The presence of asynaptic axes-labeled by BRCA1 and γ-H2AX-at meiotic prophase in testes that have a normal development of spermatogenesis, suggests that the basic mechanism that arrests spreading of transcriptional silencing (meiotic sex chromosome inactivation (MSCI)) to the autosomal segments may be per se the formation of a functional synaptonemal complex between homologous or non-homologous regions, and thus, this SC barrier might be probably related to the preservation of fertility in these systems.


Subject(s)
Chiroptera/genetics , Chromatin Assembly and Disassembly/physiology , Chromatin/metabolism , Sex Determination Processes/genetics , X Chromosome/genetics , Y Chromosome/genetics , Animals , Chromosome Pairing/genetics , Male , Pachytene Stage/physiology , Spermatocytes/metabolism , Spermatogenesis/physiology
3.
J Reprod Dev ; 62(6): 623-630, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27665783

ABSTRACT

Cohesins containing a meiosis-specific α-kleisin subunit, RAD21L or REC8, play roles in diverse aspects of meiotic chromosome dynamics including formation of axial elements (AEs), assembly of the synaptonemal complex (SC), recombination of homologous chromosomes (homologs), and cohesion of sister chromatids. However, the exact functions of individual α-kleisins remain to be elucidated. Here, we examined the localization of RAD21L and REC8 within the SC by super-resolution microscopy, 3D-SIM. We found that both RAD21L and REC8 were localized at the connection sites between lateral elements (LEs) and transverse filaments (TFs) of pachynema with RAD21L locating interior to REC8 sites. RAD21L and REC8 were not symmetrical in terms of synaptic homologs, suggesting that the arrangement of different cohesins is not strictly fixed along all chromosome axes. Intriguingly, some RAD21L signals, but not REC8 signals, were observed between unsynapsed regions of AEs of zygonema as if they formed a bridge between homologs. Furthermore, the signals of recombination intermediates overlapped with those of RAD21L to a greater degree than with those of REC8. These results highlight the different properties of two meiotic α-kleisins, and strongly support the previous proposition that RAD21L is an atypical cohesin that establishes the association between homologs rather than sister chromatids.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Meiosis/physiology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Spermatocytes/metabolism , Synaptonemal Complex/metabolism , Animals , Male , Mice , Pachytene Stage/physiology
4.
Biochem J ; 471(2): 211-20, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26268560

ABSTRACT

Spermatogenesis is a highly regulated process that involves both mitotic and meiotic divisions, as well as cellular differentiation to yield mature spermatozoa from undifferentiated germinal stem cells. Although Gpat2 was originally annotated as encoding a glycerol-3-phosphate acyltransferase by sequence homology to Gpat1, GPAT2 is highly expressed in testis but not in lipogenic tissues and is not up-regulated during adipocyte differentiation. New data show that GPAT2 is required for the synthesis of piRNAs (piwi-interacting RNAs), a group of small RNAs that protect the germ cell genome from retrotransposable elements. In order to understand the relationship between GPAT2 and its role in the testis, we focused on Gpat2 expression during the first wave of mouse spermatogenesis. Gpat2 expression was analysed by qPCR (quantitative real-time PCR), in situ hybridization, immunohistochemistry and Western blotting. Gpat2 mRNA content and protein expression were maximal at 15 dpp (days post-partum) and were restricted to pachytene spermatocytes. To achieve this transient expression, both epigenetic mechanisms and trans-acting factors are involved. In vitro assays showed that Gpat2 expression correlates with DNA demethylation and histone acetylation and that it is up-regulated by retinoic acid. Epigenetic regulation by DNA methylation was confirmed in vivo in germ cells by bisulfite sequencing of the Gpat2 promoter. Consistent with the initiation of meiosis at 11 dpp, methylation decreased dramatically. Thus, Gpat2 is expressed at a specific stage of spermatogenesis, consistent with piRNA synthesis and meiosis I prophase, and its on-off expression pattern responds predominantly to epigenetic modifications.


Subject(s)
DNA Methylation/physiology , Epigenesis, Genetic/physiology , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Meiotic Prophase I/physiology , Pachytene Stage/physiology , Promoter Regions, Genetic/physiology , Spermatocytes/metabolism , Spermatogenesis/physiology , Animals , Glycerol-3-Phosphate O-Acyltransferase/genetics , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spermatocytes/cytology
5.
Annu Rev Physiol ; 74: 425-51, 2012.
Article in English | MEDLINE | ID: mdl-22335798

ABSTRACT

We review the critical events in early meiotic prophase in Drosophila melanogaster oocytes. We focus on four aspects of this process: the formation of the synaptonemal complex (SC) and its role in maintaining homologous chromosome pairings, the critical roles of the meiosis-specific process of centromere clustering in the formation of a full-length SC, the mechanisms by which preprogrammed double-strand breaks initiate meiotic recombination, and the checkpoints that govern the progression and coordination of these processes. Central to this discussion are the roles that somatic pairing events play in establishing the necessary conditions for proper SC formation, the roles of centromere pairing in synapsis initiation, and the mechanisms by which oocytes detect failures in SC formation and/or recombination. Finally, we correlate what is known in Drosophila oocytes with our understanding of these processes in other systems.


Subject(s)
Chromosomes/physiology , Drosophila/physiology , Meiosis/physiology , Meiotic Prophase I/physiology , Oocytes/physiology , Animals , Cell Nucleus/physiology , Centromere/physiology , Chromosome Pairing/physiology , DNA Breaks, Double-Stranded , DNA Damage , Female , Humans , Oocytes/growth & development , Pachytene Stage/physiology , Synaptonemal Complex/physiology , Telomere/physiology
6.
Development ; 138(15): 3319-30, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21750041

ABSTRACT

The transcriptional regulation of mammalian meiosis is poorly characterized, owing to few genetic and ex vivo models. From a genetic screen, we identify the transcription factor MYBL1 as a male-specific master regulator of several crucial meiotic processes. Spermatocytes bearing a novel separation-of-function allele (Mybl1(repro9)) had subtle defects in autosome synapsis in pachynema, a high incidence of unsynapsed sex chromosomes, incomplete double-strand break repair on synapsed pachytene chromosomes and a lack of crossing over. MYBL1 protein appears in pachynema, and its mutation caused specific alterations in expression of diverse genes, including some translated postmeiotically. These data, coupled with chromatin immunoprecipitation (ChIP-chip) experiments and bioinformatic analysis of promoters, identified direct targets of MYBL1 regulation. The results reveal that MYBL1 is a master regulator of meiotic genes that are involved in multiple processes in spermatocytes, particularly those required for cell cycle progression through pachynema.


Subject(s)
Gene Expression Regulation, Developmental , Meiosis/physiology , Proto-Oncogene Proteins c-myb/metabolism , Spermatocytes/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , DNA Breaks, Double-Stranded , Female , Gene Expression Profiling , Humans , Infertility, Male/genetics , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Microarray Analysis , Molecular Sequence Data , Mutation , Pachytene Stage/physiology , Proto-Oncogene Proteins c-myb/genetics , Sequence Alignment , Spermatocytes/cytology , Spermatogenesis/physiology , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic
7.
Mol Reprod Dev ; 81(10): 946-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25257909

ABSTRACT

Vasa is a universal marker of the germ line in animals, yet mutations disrupting vasa cause sexually dimorphic infertility, with impaired development of the ovary in some animals and the testis in others. The basis for this sexually dimorphic requirement for Vasa is not clear; in most animals examined, both the male and female gonad express vasa throughout the life of the germ line. Here we characterized a loss-of-function mutation disrupting zebrafish vasa. We show that maternally provided Vasa is stable through the first ten days of development in zebrafish, and thus likely fulfills any early roles for Vasa during germ-line specification, migration, survival, and maintenance. Although zygotic Vasa is not essential for the development of juvenile gonads, vasa mutants develop exclusively as sterile males. Furthermore, phenotypes of vasa;p53 compound mutants are indistinguishable from those of vasa mutants, therefore the failure of vasa mutants to differentiate as females and to support germ-cell development in the testis is not due to p53-mediated apoptosis. Instead, we found that failure to progress beyond the pachytene stage of meiosis causes the loss of germ-line stem cells, leaving empty somatic tubules. Our studies provide insight into the function of zebrafish vasa during female meiosis, differentiation, and maintenance of germ-line stem cells.


Subject(s)
Cell Differentiation/physiology , DEAD-box RNA Helicases/metabolism , Germ Cells/metabolism , Pachytene Stage/physiology , Stem Cells/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Apoptosis/genetics , DEAD-box RNA Helicases/genetics , Female , Germ Cells/cytology , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mutation , Stem Cells/cytology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
8.
Chromosoma ; 121(3): 307-26, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22366883

ABSTRACT

During the first meiotic prophase in male mammals, sex chromosomes undergo a program of transcriptional silencing called meiotic sex chromosome inactivation (MSCI). MSCI is triggered by accumulation of proteins like BRCA1, ATR, and γH2AX on unsynapsed chromosomes, followed by local changes on the sex chromatin, including histone modifications, incorporation of specific histone variants, non-histone proteins, and RNAs. It is generally thought that MSCI represents the transition of unsynapsed chromatin from a transcriptionally active state to a repressed state. However, transcription is generally low in the whole nucleus during the early stages of the first meiotic prophase, when markers of MSCI first appear, and is then reactivated globally during pachytene. Thus, an alternative possibility is that MSCI represents the targeted maintenance and/or reinforcement of a prior repressed state, i.e., a failure to reactivate. Here, we present an analysis of the temporal and spatial appearance of transcriptional and MSCI markers, as well as chromatin modifications related to transcriptional regulation. We show that levels of RNA pol II and histone H3 acetylated at lysine 9 (H3K9ac) are low during leptotene, zygotene, and early pachytene, but increase strongly in mid-pachytene, indicating that reactivation occurs with some delay after synapsis. However, while transcription markers appear abundantly on the autosomes at mid-pachytene, they are not directed to the sex chromosomes. Interestingly, we found that chromatin modifications related to transcriptional silencing and/or MSCI, namely, histone H3 trimethylated at lysine 9 (H3K9me3), histone H3 monomethylated at lysine 4 (H3K4me1), γH2AX, SUMO1, and XMR, appear on the sex chromosomes before autosomes become reactivated. These results suggest that the onset of MSCI during late zygotene and early pachytene may prevent sex chromosome reactivation during mid-pachytene instead of promoting inactivation de novo. Additionally, we found temporal differences between the X and Y chromosomes in the recruitment of DNA repair and MSCI markers, indicating a differential regulation of these processes. We propose that many of the meiotic defects attributed to failure to silence sex chromosomes could be interpreted as a more general process of transcriptional misregulation that occurs under certain pathological circumstances in zygotene and early pachytene.


Subject(s)
Gene Silencing , Meiotic Prophase I/genetics , X Chromosome/metabolism , Y Chromosome/metabolism , Animals , Carrier Proteins , Cell Cycle Proteins , Chromatin/metabolism , Chromosome Pairing/physiology , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins , Histones/metabolism , Male , Mice , Nuclear Proteins/metabolism , Pachytene Stage/physiology , RNA Polymerase II/metabolism , RNA-Binding Proteins , SUMO-1 Protein/metabolism , Transcription, Genetic
9.
J Exp Bot ; 64(8): 2139-54, 2013 May.
Article in English | MEDLINE | ID: mdl-23554258

ABSTRACT

In barley (Hordeum vulgare L.), chiasmata (the physical sites of genetic crossovers) are skewed towards the distal ends of chromosomes, effectively consigning a large proportion of genes to recombination coldspots. This has the effect of limiting potential genetic variability, and of reducing the efficiency of map-based cloning and breeding approaches for this crop. Shifting the sites of recombination to more proximal chromosome regions by forward and reverse genetic means may be profitable in terms of realizing the genetic potential of the species, but is predicated upon a better understanding of the mechanisms governing the sites of these events, and upon the ability to recognize real changes in recombination patterns. The barley MutL Homologue (HvMLH3), a marker for class I interfering crossovers, has been isolated and a specific antibody has been raised. Immunolocalization of HvMLH3 along with the synaptonemal complex transverse filament protein ZYP1, used in conjunction with fluorescence in situ hybridization (FISH) tagging of specific barley chromosomes, has enabled access to the physical recombination landscape of the barley cultivars Morex and Bowman. Consistent distal localization of HvMLH3 foci throughout the genome, and similar patterns of HvMLH3 foci within bivalents 2H and 3H have been observed. A difference in total numbers of HvMLH3 foci between these two cultivars has been quantified, which is interpreted as representing genotypic variation in class I crossover frequency. Discrepancies between the frequencies of HvMLH3 foci and crossover frequencies derived from linkage analysis point to the existence of at least two crossover pathways in barley. It is also shown that interference of HvMLH3 foci is relatively weak compared with other plant species.


Subject(s)
Chromosomes, Plant/genetics , Hordeum/genetics , Pachytene Stage/genetics , Arabidopsis/genetics , Base Sequence , Chromosome Mapping , Chromosomes, Plant/physiology , Crossing Over, Genetic/genetics , Crossing Over, Genetic/physiology , Genetic Linkage/genetics , Genetic Linkage/physiology , Genetic Loci/genetics , Genetic Loci/physiology , Genome, Plant/genetics , Genome, Plant/physiology , Hordeum/physiology , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Pachytene Stage/physiology , Phylogeny , Sequence Alignment , Synaptonemal Complex/genetics , Synaptonemal Complex/physiology
10.
Reproduction ; 145(2): 203-12, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23241345

ABSTRACT

Reactive oxygen species (ROS) and reactive nitrogen species (RNS) like superoxide and nitric oxide are produced by testis and spermatogenic cells in response to heat stress. However, the magnitude and mechanisms of this production in spermatogenic cells have not been described. In this work, we evaluated ROS/RNS production, its pharmacology, mitochondrial oxidative metabolism, membrane potential and antioxidant capacity at different temperatures in isolated rat pachytene spermatocytes and round spermatids. Our results showed an increment in ROS/RNS production by pachytene spermatocytes when increasing the temperature to 40 °C. Instead, ROS/RNS production by round spermatids did not change at temperatures higher than 33 °C. ROS/RNS production was sensitive to NADPH oxidase inhibitor diphenylene iodonium or the mitochondrial complex I inhibitor rotenone. No additive effects were observed for these two compounds. Our results suggest an important mitochondrial ROS/RNS production in spermatogenic cells. Oligomycin-insensitive oxygen consumption (uncoupled oxygen consumption) increased with temperature and was significantly larger in round spermatids than pachytene spermatocytes, indicating a likely round spermatid mitochondrial uncoupling at high temperatures. A similar conclusion can be reached by measuring the mitochondrial membrane potential using rhodamine 123 fluorescence in permeabilized cells or JC-1 fluorescence in intact cells. The antioxidant capacity was higher in round spermatids than pachytene spermatocytes at 40 °C. Our results strongly suggest that at high temperatures (40 °C) pachytene spermatocytes are more susceptible to oxidative stress, but round spermatids are more protected because of a temperature-induced mitochondrial uncoupling together with a larger antioxidant capacity.


Subject(s)
Cold Temperature , Hot Temperature , Pachytene Stage/physiology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Animals , Antioxidants/metabolism , Body Temperature/physiology , Cells, Cultured , Heat-Shock Response/physiology , Male , Rats , Rats, Sprague-Dawley , Spermatids/physiology , Spermatocytes/physiology , Spermatogenesis/physiology
11.
J Biol Chem ; 286(52): 44306-18, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22086916

ABSTRACT

Gonadotropin-regulated testicular RNA helicase (GRTH/DDX25), a testis-specific member of the DEAD-box family, is an essential post-transcriptional regulator of spermatogenesis. Failure of expression of Transition protein 2 (TP2) and Protamine 2 (Prm2) proteins (chromatin remodelers, essential for spermatid elongation and completion of spermatogenesis) with preservation of their mRNA expression was observed in GRTH-null mice (azoospermic due to failure of spermatids to elongate). These were identified as target genes for the testis-specific miR-469, which is increased in the GRTH-null mice. Further analysis demonstrated that miR-469 repressed TP2 and Prm2 protein expression at the translation level with minor effect on mRNA degradation, through binding to the coding regions of TP2 and Prm2 mRNAs. The corresponding primary-microRNAs and the expression levels of Drosha and DGCR8 (both mRNA and protein) were increased significantly in the GRTH-null mice. miR-469 silencing of TP2 and Prm2 mRNA in pachytene spermatocytes and round spermatids is essential for their timely translation at later times of spermiogenesis, which is critical to attain mature sperm. Collectively, these studies indicate that GRTH, a multifunctional RNA helicase, acts as a negative regulator of miRNA-469 biogenesis and consequently their function during spermatogenesis.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Silencing/physiology , MicroRNAs/biosynthesis , Nuclear Proteins/biosynthesis , Open Reading Frames/physiology , Protamines/metabolism , RNA, Messenger/biosynthesis , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/physiology , Animals , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Nuclear Proteins/genetics , Organ Specificity/physiology , Pachytene Stage/physiology , Protamines/genetics , Proteins/genetics , Proteins/metabolism , RNA Stability/physiology , RNA, Messenger/genetics , RNA-Binding Proteins , Ribonuclease III/genetics , Ribonuclease III/metabolism , Spermatids/cytology , Spermatocytes/cytology , Testis/cytology , Testis/metabolism , Up-Regulation/physiology
12.
Exp Cell Res ; 316(2): 158-71, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19686734

ABSTRACT

HORMA domain-containing proteins regulate interactions between homologous chromosomes (homologs) during meiosis in a wide range of eukaryotes. We have identified a mouse HORMA domain-containing protein, HORMAD1, and biochemically and cytologically shown it to be associated with the meiotic chromosome axis. HORMAD1 first accumulates on the chromosomes during the leptotene to zygotene stages of meiotic prophase I. As germ cells progress into the pachytene stage, HORMAD1 disappears from the synapsed chromosomal regions. However, once the chromosomes desynapse during the diplotene stage, HORMAD1 again accumulates on the chromosome axis of the desynapsed homologs. HORMAD1 thus preferentially localizes to unsynapsed or desynapsed chromosomal regions during the prophase I stage of meiosis. Analysis of mutant strains lacking different components of the synaptonemal complex (SC) revealed that establishment of the SC is required for the displacement of HORMAD1 from the chromosome axis. Our results therefore strongly suggest that also mammalian cells use a HORMA domain-containing protein as part of a surveillance system that monitors synapsis or other interactions between homologs.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosome Pairing/physiology , Chromosomes, Mammalian/metabolism , Meiosis/physiology , Animals , BRCA1 Protein/metabolism , COS Cells , Cell Cycle Proteins/chemistry , Cell Nucleus/metabolism , Chlorocebus aethiops , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins , Embryo, Mammalian/metabolism , Female , Histones/metabolism , Male , Meiotic Prophase I/physiology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/metabolism , Pachytene Stage/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Spermatocytes/metabolism , Synaptonemal Complex/metabolism , Testis/metabolism , Transfection , Cohesins
13.
Dev Cell ; 8(6): 949-61, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15935783

ABSTRACT

REC8 is a key component of the meiotic cohesin complex. During meiosis, cohesin is required for the establishment and maintenance of sister-chromatid cohesion, for the formation of the synaptonemal complex, and for recombination between homologous chromosomes. We show that REC8 has an essential role in mammalian meiosis, in that Rec8 null mice of both sexes have germ cell failure and are sterile. In the absence of REC8, early chromosome pairing events appear normal, but synapsis occurs in a novel fashion: between sister chromatids. This implies that a major role for REC8 in mammalian meiosis is to limit synapsis to between homologous chromosomes. In all other eukaryotic species studied to date, REC8 phenotypes have been restricted to meiosis. Unexpectedly, Rec8 null mice are born in sub-Mendelian frequencies and fail to thrive. These findings illuminate hitherto unknown REC8 functions in chromosome dynamics during mammalian meiosis and possibly in somatic development.


Subject(s)
Acetaminophen/analogs & derivatives , Chromatids/metabolism , Chromosome Pairing/physiology , Meiosis/physiology , Nuclear Proteins/deficiency , Nuclear Proteins/physiology , Phosphoproteins/deficiency , Phosphoproteins/physiology , Saccharin/analogs & derivatives , Acetaminophen/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Death/physiology , Chondroitin Sulfate Proteoglycans/metabolism , Chromatids/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Painting/methods , Chromosomes/metabolism , Chromosomes/ultrastructure , Chromosomes, Human, Pair 10/metabolism , Cloning, Molecular/methods , DNA-Binding Proteins/metabolism , Electroporation/methods , Female , Humans , Immunohistochemistry/methods , In Situ Nick-End Labeling/methods , Indoles/metabolism , Male , Meiotic Prophase I/physiology , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Models, Biological , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncorhynchus kisutch/metabolism , Ovary/metabolism , Pachytene Stage/physiology , Phosphate-Binding Proteins , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rad51 Recombinase , Saccharin/metabolism , Spermatogenesis/genetics , Testis/metabolism , Testis/ultrastructure , Trans-Activators/metabolism
14.
J Cell Biol ; 171(2): 241-53, 2005 Oct 24.
Article in English | MEDLINE | ID: mdl-16230461

ABSTRACT

Sister chromatid cohesion and interhomologue recombination are coordinated to promote the segregation of homologous chromosomes instead of sister chromatids at the first meiotic division. During meiotic prophase in Saccharomyces cerevisiae, the meiosis-specific cohesin Rec8p localizes along chromosome axes and mediates most of the cohesion. The mitotic cohesin Mcd1p/Scc1p localizes to discrete spots along chromosome arms, and its function is not clear. In cells lacking Tid1p, which is a member of the SWI2/SNF2 family of helicase-like proteins that are involved in chromatin remodeling, Mcd1p and Rec8p persist abnormally through both meiotic divisions, and chromosome segregation fails in the majority of cells. Genetic results indicate that the primary defect in these cells is a failure to resolve Mcd1p-mediated connections. Tid1p interacts with recombination enzymes Dmc1p and Rad51p and has an established role in recombination repair. We propose that Tid1p remodels Mcd1p-mediated cohesion early in meiotic prophase to facilitate interhomologue recombination and the subsequent segregation of homologous chromosomes.


Subject(s)
Cell Cycle Proteins/metabolism , Fungal Proteins/metabolism , Meiosis/physiology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Anaphase/physiology , Cell Cycle Proteins/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Fungal/genetics , Chromosomes, Fungal/metabolism , DNA Helicases , DNA Repair Enzymes , Fungal Proteins/genetics , Nuclear Proteins/genetics , Pachytene Stage/physiology , Phosphoproteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/genetics , Cohesins
15.
Genetika ; 46(10): 1363-6, 2010 Oct.
Article in Russian | MEDLINE | ID: mdl-21254557

ABSTRACT

Fluorochrome-labeled oligonucleotides (n = 44) corresponding to mouse genome repetitive sequences were hybridized in situ with pachytene nuclei of mouse spermatocytes. Signals of the repetitive sequences MaLR, MER, and (GT)22 were found to be dispersed through chromatin, and signals of BI 1 repeats and minisatellites were mostly attached to synaptonemal complexes immunostained with anti-SYCP3 antibodies. These results suggest that B 1 repeats and minisatellites are candidates for sequences anchoring chromatin to synaptonemal complexes.


Subject(s)
Chromatin/metabolism , Chromosomes, Mammalian/metabolism , Pachytene Stage/physiology , Spermatocytes/metabolism , Synaptonemal Complex/metabolism , Animals , Chromatin/genetics , In Situ Hybridization, Fluorescence , Male , Mice , Minisatellite Repeats/physiology
16.
Curr Biol ; 16(7): R249-51, 2006 Apr 04.
Article in English | MEDLINE | ID: mdl-16581501

ABSTRACT

Faithful recombination and chromosome segregation in meiosis require regulated steps of homolog recognition and association which are monitored by meiotic checkpoints. A recent study in the nematode Caenorhabditis elegans has identified a checkpoint mechanism that monitors chromosome pairing during meiosis.


Subject(s)
Caenorhabditis elegans/genetics , Chromosome Pairing/physiology , Meiosis/genetics , Animals , Apoptosis/physiology , Caenorhabditis elegans/cytology , DNA Damage , Female , Germ Cells/cytology , Germ Cells/physiology , Male , Mutation , Pachytene Stage/physiology , Recombination, Genetic
17.
Reproduction ; 138(3): 463-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19556438

ABSTRACT

Male fertility relies on the highly specialized process of spermatogenesis to continually renew the supply of spermatozoa necessary for reproduction. Central to this unique process is meiosis that is responsible for the production of haploid spermatozoa as well as for generating genetic diversity. During meiosis I, there is a dramatic increase in the number of mitochondria present within the developing spermatocytes, suggesting an increased necessity for ATP production and utilization. Essential for the utilization of ATP is the translocation of ADP and ATP across the inner mitochondrial membrane, which is mediated by the adenine nucleotide translocases (Ant). We recently identified and characterized a novel testis specific Ant, ANT4 (also known as SLC25A31 and Aac4). The generation of Ant4-deficient animals resulted in the severe disruption of the seminiferous epithelium with an apparent spermatocytic arrest of the germ cell population. In the present study utilizing a chromosomal spread technique, we determined that Ant4-deficiency results in an accumulation of leptotene spermatocytes, a decrease in pachytene spermatocytes, and an absence of diplotene spermatocytes, indicating early meiotic arrest. Furthermore, the chromosomes of Ant4-deficient pachytene spermatocyte occasionally demonstrated sustained gammaH2AX association as well as synaptonemal complex protein 1 (SYCP1)/SYCP3 dissociation beyond the sex body. Large ATP supplies from mitochondria may be critical for normal progression of spermatogenesis during early stages of meiotic prophase I, including DNA double-strand break repair and chromosomal synapsis.


Subject(s)
Meiosis/genetics , Membrane Transport Proteins/genetics , Spermatozoa/physiology , Animals , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins , DNA-Binding Proteins , Female , Germ Cells/cytology , Germ Cells/metabolism , Germ Cells/physiology , Histones/metabolism , Male , Meiosis/physiology , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Pachytene Stage/genetics , Pachytene Stage/physiology , Spermatocytes/metabolism , Spermatocytes/physiology , Spermatogenesis/genetics , Spermatogenesis/physiology , Spermatozoa/cytology , Spermatozoa/enzymology , Spermatozoa/metabolism , Testis/cytology , Testis/metabolism , Time Factors
18.
Cell Death Differ ; 26(11): 2194-2207, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30760872

ABSTRACT

Lethal (3) malignant brain tumor like 2 (L3MBTL2) is a member of the MBT-domain proteins, which are involved in transcriptional repression and implicated in chromatin compaction. Our previous study has shown that L3MBTL2 is highly expressed in the testis, but its role in spermatogenesis remains unclear. In the present study, we found that L3MBTL2 was most highly expressed in pachytene spermatocytes within the testis. Germ cell-specific ablation of L3mbtl2 in the testis led to increased abnormal spermatozoa, progressive decrease of sperm counts and premature testicular failure in mice. RNA-sequencing analysis on L3mbtl2 deficient testes confirmed that L3MBTL2 was a transcriptional repressor but failed to reveal any significant changes in spermatogenesis-associated genes. Interestingly, L3mbtl2 deficiency resulted in increased γH2AX deposition in the leptotene spermatocytes, subsequent inappropriate retention of γH2AX on autosomes, and defective crossing-over and synapsis during the pachytene stage of meiosis I, and more germ cell apoptosis and degeneration in aging mice. L3MBTL2 interacted with the histone ubiquitin ligase RNF8. Inhibition of L3MBTL2 reduced nuclear RNF8 and ubH2A levels in GC2 cells. L3mbtl2 deficiency led to decreases in the levels of the RNF8 and ubH2A pathway and in histone acetylation in elongating spermatids, and in protamine 1 deposition and chromatin condensation in sperm. These results suggest that L3MBTL2 plays important roles in chromatin remodeling during meiosis and spermiogenesis.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/metabolism , Nuclear Proteins/genetics , Spermatocytes/metabolism , Spermatogenesis/genetics , Transcription Factors/genetics , Acetylation , Animals , Apoptosis/genetics , Chromatin Assembly and Disassembly/physiology , Histones/metabolism , Male , Meiotic Prophase I/physiology , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Pachytene Stage/physiology , Polycomb-Group Proteins/metabolism , Sperm Count , Testis/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
19.
Genetika ; 44(8): 1101-7, 2008 Aug.
Article in Russian | MEDLINE | ID: mdl-18825959

ABSTRACT

Instability of B-chromosomes was estimated in somatic and germline cells of samples Apodemus peninsulae from different localities of the species range. In 84 out of 188 animals (45%), in cells assessed for B-chromosome mosaicism, bone marrow cells with different B-chromosome number were observed. The numbers of B-chromosomes in spermatocytes at the pachytene stage were estimated in ten males. It was shown that the average number of B-chromosomes and the number of cell clones in germline cells was higher than the corresponding numbers in bone marrow cells. The higher number of B-chromosomes and their higher variability in germline cells than in somatic cells suggest the existence of a mechanism of premeiotic accumulation of B-chromosomes in spermatogenesis of A. peninsulae.


Subject(s)
Chromosomal Instability/physiology , Chromosomes, Mammalian/genetics , Murinae/genetics , Pachytene Stage/physiology , Spermatogenesis/physiology , Animals , Chromosomes, Mammalian/metabolism , Female , Male , Murinae/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism
20.
Curr Biol ; 14(23): 2135-42, 2004 Dec 14.
Article in English | MEDLINE | ID: mdl-15589157

ABSTRACT

In mammalian spermatogenesis, the X and Y chromosomes are transcriptionally silenced during the pachytene stage of meiotic prophase (meiotic sex chromosome inactivation, MSCI), forming a condensed chromatin domain termed the sex or XY body. The nucleosomal core histone H2AX is phosphorylated within the XY chromatin domain just prior to MSCI, and it has been hypothesized that this triggers the chromatin condensation and transcriptional repression. Here, we show that the kinase ATR localizes to XY chromatin at the onset of MSCI and that this localization is disrupted in mice with a mutant form of the tumor suppressor protein BRCA1. In the mutant pachytene cells, ATR is usually present at nonsex chromosomal sites, where it colocalizes with aberrant sites of H2AX phosphorylation; in these cells, there is MSCI failure. In rare pachytene cells, ATR does locate to XY chromatin, H2AX is then phosphorylated, a sex body forms, and MSCI ensues. These observations highlight an important role for BRCA1 in recruiting the kinase ATR to XY chromatin at the onset of MSCI and provide compelling evidence that it is ATR that phosphorylates H2AX and triggers MSCI.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Genes, BRCA1/physiology , Histones/metabolism , Pachytene Stage/physiology , Sex Chromosomes/physiology , Spermatogenesis/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/metabolism , DNA Primers , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization, Fluorescence , Male , Mice , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spermatogenesis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL