Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
PLoS Genet ; 15(6): e1008177, 2019 06.
Article in English | MEDLINE | ID: mdl-31170160

ABSTRACT

During meiotic prophase I, double-strand breaks (DSBs) initiate homologous recombination leading to non-crossovers (NCOs) and crossovers (COs). In mouse, 10% of DSBs are designated to become COs, primarily through a pathway dependent on the MLH1-MLH3 heterodimer (MutLγ). Mlh3 contains an endonuclease domain that is critical for resolving COs in yeast. We generated a mouse (Mlh3DN/DN) harboring a mutation within this conserved domain that is predicted to generate a protein that is catalytically inert. Mlh3DN/DN males, like fully null Mlh3-/- males, have no spermatozoa and are infertile, yet spermatocytes have grossly normal DSBs and synapsis events in early prophase I. Unlike Mlh3-/- males, mutation of the endonuclease domain within MLH3 permits normal loading and frequency of MutLγ in pachynema. However, key DSB repair factors (RAD51) and mediators of CO pathway choice (BLM helicase) persist into pachynema in Mlh3DN/DN males, indicating a temporal delay in repair events and revealing a mechanism by which alternative DSB repair pathways may be selected. While Mlh3DN/DN spermatocytes retain only 22% of wildtype chiasmata counts, this frequency is greater than observed in Mlh3-/- males (10%), suggesting that the allele may permit partial endonuclease activity, or that other pathways can generate COs from these MutLγ-defined repair intermediates in Mlh3DN/DN males. Double mutant mice homozygous for the Mlh3DN/DN and Mus81-/- mutations show losses in chiasmata close to those observed in Mlh3-/- males, indicating that the MUS81-EME1-regulated crossover pathway can only partially account for the increased residual chiasmata in Mlh3DN/DN spermatocytes. Our data demonstrate that mouse spermatocytes bearing the MLH1-MLH3DN/DN complex display the proper loading of factors essential for CO resolution (MutSγ, CDK2, HEI10, MutLγ). Despite these functions, mice bearing the Mlh3DN/DN allele show defects in the repair of meiotic recombination intermediates and a loss of most chiasmata.


Subject(s)
DNA-Binding Proteins/genetics , Endonucleases/genetics , Meiotic Prophase I/genetics , MutL Proteins/genetics , Animals , Chromosome Pairing/genetics , Crossing Over, Genetic , DNA Breaks, Double-Stranded , DNA Repair/genetics , Homologous Recombination/genetics , Male , Meiosis/genetics , Mice , MutL Protein Homolog 1/genetics , MutS Proteins/genetics , Rad51 Recombinase/genetics , Spermatocytes/growth & development , Spermatocytes/metabolism
2.
Chromosoma ; 125(2): 237-52, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26490168

ABSTRACT

Fancj, the gene associated with Fanconi anemia (FA) Complementation Group J, encodes a DNA helicase involved in homologous recombination repair and the cellular response to replication stress. FANCJ functions in part through its interaction with key DNA repair proteins, including MutL homolog-1 (MLH1), Breast Cancer Associated gene-1 (BRCA1), and Bloom syndrome helicase (BLM). All three of these proteins are involved in a variety of events that ensure genome stability, including the events of DNA double strand break (DSB) repair during prophase I of meiosis. Meiotic DSBs are repaired through homologous recombination resulting in non-crossovers (NCO) or crossovers (CO). The frequency and placement of COs are stringently regulated to ensure that each chromosome receives at least one CO event, and that longer chromosomes receive at least one additional CO, thus facilitating the accurate segregation of homologous chromosomes at the first meiotic division. In the present study, we investigated the role of Fancj during prophase I using a gene trap mutant allele. Fancj (GT/GT) mutants are fertile, but their testes are very much smaller than wild-type littermates, predominantly as a result of impeded spermatogonial proliferation and mildly increased apoptosis during testis development in the fetus. This defect in spermatogonial proliferation is consistent with mutations in other FA genes. During prophase I, early events of synapsis and DSB induction/repair appear mostly normal in Fancj (GT/GT) males, and the FANCJ-interacting protein BRCA1 assembles normally on meiotic chromosome cores. However, MLH1 focus frequency is increased in Fancj (GT/GT) males, indicative of increased DSB repair via CO, and is concomitant with increased chiasmata at diakinesis. This increase in COs in the absence of FANCJ is associated with increased localization of BLM helicase protein, indicating that BLM may facilitate the increased rate of crossing over in Fancj (GT/GT) males. Taken together, these results demonstrate a critical role for FANCJ in spermatogenesis at two stages: firstly in the proliferative activity that gives rise to the full complement of testicular spermatogonia and secondly in the establishment of appropriate CO numbers during prophase I.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Crossing Over, Genetic , Fanconi Anemia Complementation Group Proteins/metabolism , Meiotic Prophase I , Mice/embryology , Mice/metabolism , Spermatogonia/metabolism , Alleles , Animals , Basic-Leucine Zipper Transcription Factors/genetics , DNA Breaks, Double-Stranded , DNA Repair , Fanconi Anemia Complementation Group Proteins/genetics , Male , Mice/genetics , RNA Helicases , Recombination, Genetic , Spermatogenesis , Spermatogonia/cytology , Spermatogonia/growth & development
3.
J Cell Biol ; 205(5): 633-41, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24891606

ABSTRACT

Meiotic crossovers (COs) are crucial for ensuring accurate homologous chromosome segregation during meiosis I. Because the double-strand breaks (DSBs) that initiate meiotic recombination greatly outnumber eventual COs, this process requires exquisite regulation to narrow down the pool of DSB intermediates that may form COs. In this paper, we identify a cyclin-related protein, CNTD1, as a critical mediator of this process. Disruption of Cntd1 results in failure to localize CO-specific factors MutLγ and HEI10 at designated CO sites and also leads to prolonged high levels of pre-CO intermediates marked by MutSγ and RNF212. These data show that maturation of COs is intimately coupled to deselection of excess pre-CO sites to yield a limited number of COs and that CNTD1 coordinates these processes by regulating the association between the RING finger proteins HEI10 and RNF212 and components of the CO machinery.


Subject(s)
Crossing Over, Genetic , Cyclins/genetics , Cyclins/physiology , Meiosis , Animals , Cell Cycle Proteins , Cell Differentiation , Chromosome Segregation , Chromosomes/ultrastructure , DNA Breaks, Double-Stranded , Ligases/metabolism , Male , Mice , Mice, Transgenic , Mutation , Phenotype , Recombination, Genetic , Sperm Count , Spermatocytes/cytology , Spermatocytes/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
Methods Mol Biol ; 957: 1-18, 2013.
Article in English | MEDLINE | ID: mdl-23138941

ABSTRACT

Meiosis is the specialized cell division in sexually reproducing organisms in which haploid gametes are produced. Meiotic prophase I is the defining stage of meiosis, when pairing and synapsis occur between homologous chromosomes, concurrent with reciprocal recombination (or crossing over) events that arise between them. Any disruption of these events during prophase I can lead to improper segregation of homologous chromosomes which can cause severe birth defects in the resulting progeny, and this occurs with alarming frequency in human oocytes. Thus, while the pathways that regulate these events in prophase I are highly conserved in both males and females, the stringency with which these events are monitored and/or controlled appears to be dramatically lower in females. These observations underscore the need to examine and compare meiotic mechanisms across the sexes. However, the study of female meiosis is impeded by the early start of meiosis during fetal development and the very limited amount of ovarian tissue available for meiotic analyses. Here we describe three different techniques which are useful for meiotic prophase I analysis in mouse/human oocytes, ranging from early prophase I events through until the resolution of crossing over at the first and second meiotic divisions.


Subject(s)
Cytogenetic Analysis/methods , Oocytes/cytology , Oocytes/metabolism , Recombination, Genetic , Animals , Chromosomes, Mammalian/genetics , Female , Fetus/cytology , Fetus/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Male , Meiotic Prophase I/genetics , Metaphase/genetics , Mice , Ovary/cytology , Ovary/metabolism
5.
PLoS Genet ; 7(6): e1002094, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21655083

ABSTRACT

The mammalian ortholog of yeast Slx4, BTBD12, is an ATM substrate that functions as a scaffold for various DNA repair activities. Mutations of human BTBD12 have been reported in a new sub-type of Fanconi anemia patients. Recent studies have implicated the fly and worm orthologs, MUS312 and HIM-18, in the regulation of meiotic crossovers arising from double-strand break (DSB) initiating events and also in genome stability prior to meiosis. Using a Btbd12 mutant mouse, we analyzed the role of BTBD12 in mammalian gametogenesis. BTBD12 localizes to pre-meiotic spermatogonia and to meiotic spermatocytes in wildtype males. Btbd12 mutant mice have less than 15% normal spermatozoa and are subfertile. Loss of BTBD12 during embryogenesis results in impaired primordial germ cell proliferation and increased apoptosis, which reduces the spermatogonial pool in the early postnatal testis. During prophase I, DSBs initiate normally in Btbd12 mutant animals. However, DSB repair is delayed or impeded, resulting in persistent γH2AX and RAD51, and the choice of repair pathway may be altered, resulting in elevated MLH1/MLH3 focus numbers at pachynema. The result is an increase in apoptosis through prophase I and beyond. Unlike yeast Slx4, therefore, BTBD12 appears to function in meiotic prophase I, possibly during the recombination events that lead to the production of crossovers. In line with its expected regulation by ATM kinase, BTBD12 protein is reduced in the testis of Atm(-/-) males, and Btbd12 mutant mice exhibit increased genomic instability in the form of elevated blood cell micronucleus formation similar to that seen in Atm(-/-) males. Taken together, these data indicate that BTBD12 functions throughout gametogenesis to maintain genome stability, possibly by co-ordinating repair processes and/or by linking DNA repair events to the cell cycle via ATM.


Subject(s)
Genomic Instability , Recombinases/genetics , Spermatogenesis/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Male , Mammals/genetics , Mammals/metabolism , Meiotic Prophase I , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinases/metabolism , Recombination, Genetic , Spermatocytes/metabolism , Testis/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Biol Reprod ; 78(3): 462-71, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18057311

ABSTRACT

The DNA mismatch repair (MMR) family functions in a variety of contexts to preserve genome integrity in most eukaryotes. In particular, members of the MMR family are involved in the process of meiotic recombination in germ cells. MMR gene mutations in mice result in meiotic disruption during prophase I, but the extent of this disruption often differs between male and female meiocytes. To address the role of MMR proteins specifically in female meiosis, we explored the progression of oocytes through prophase I and the meiotic divisions in mice harboring deletions in members of the MMR pathway (Mlh1, Mlh3, Exo1, and an ATPase-deficient variant of Mlh1, Mlh1(G67R)). The colocalization of MLH1 and MLH3, key proteins involved in stabilization of nascent crossovers, was dependent on intact heterodimer formation and was highly correlated with the ability of oocytes to progress through to metaphase II. The exception was Exo1(-/-) oocytes, in which normal MLH1/MLH3 localization was observed followed by failure to proceed to metaphase II. All mutant oocytes were able to resume meiosis after dictyate arrest, but they showed a dramatic decline in chiasmata (to less than 25% of normal), accompanied by varied progression through metaphase I. Taken together, these results demonstrate that MMR function is required for the formation and stabilization of crossovers in mammalian oocytes and that, in the absence of a functional MMR system, the failure to maintain chiasmata results in a reduced ability to proceed normally through the first and second meiotic divisions, despite near-normal levels of meiotic resumption after dictyate arrest.


Subject(s)
DNA Mismatch Repair , Germ-Line Mutation , Meiosis/genetics , Meiosis/physiology , Pregnancy, Animal , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Exodeoxyribonucleases/genetics , Female , Gene Frequency , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MutL Protein Homolog 1 , MutL Proteins , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/metabolism , Pregnancy , Signal Transduction/genetics
7.
Mol Biol Rep ; 31(3): 197-202, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15560375

ABSTRACT

We have isolated a novel cDNA from the human fetal brain cDNA library with homology to the Mg2+ -dependent serine/threonine protein phosphatase 2C (PP2C) family. The cDNA is 3055 bp in length, and the predicted coding region encodes a 360-amino-acid protein, which shows 99% identity to the PP2C epsilon from rat and mouse. Then we term it human PP2C epsilon gene. The gene is mapped to chromosome 3q26.1 and contains 4 exons. RT-PCR analysis shows that the PP2C epsilon is widely expressed in human tissues and the expression levels in heart, placenta, lung, liver, kidney, and pancreas are relatively high.


Subject(s)
Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/embryology , Chromosomes, Human, Pair 3 , Cloning, Molecular , Computational Biology , DNA, Complementary/metabolism , Exons , Gene Library , Humans , Introns , MAP Kinase Signaling System , Magnesium/chemistry , Mice , Models, Genetic , Molecular Sequence Data , Phosphoprotein Phosphatases/chemistry , Protein Phosphatase 2C , Protein Structure, Tertiary , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL