Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Genome ; 52(10): 891-6, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19935912

RESUMO

Absence of spermiogenesis in mice with meiotic defects complicates the staging of meiotic arrest using light microscopy. Consequently, new methodologies are required to establish accurate relationships among germ cells. In this study, we utilized a novel approach to analyze germ cell degeneration in juvenile mice. We used terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) in combination with meiosis-specific antibodies. Germ cell degeneration is a normal component of early spermatogenesis in juvenile mice. The incidence of germ cell death was monitored at various postnatal ages of mice using the TUNEL assay to quantify the incidence of apoptosis. Cell death occurred predominantly at 15.5 days after birth. It was found that groups of apoptotic cells were apparent in tubules containing two generations of spermatocytes that form in two progressive cohorts. Electron microscopic observations further illustrated that the majority of cells in the first cohort are in late pachytene, while groups of cells in the second cohort can degenerate in early pachytene. The methodology utilized in this study is significant because it allows one to accurately determine the point at which germ cells arrest. Consequently, we believe that these methods can be applied to study animals with meiotic defects that prevent spermiogenesis.


Assuntos
Meiose , Espermatócitos/ultraestrutura , Espermatogênese , Testículo/ultraestrutura , Animais , Apoptose , DNA Nucleotidilexotransferase/química , Imunofluorescência , Marcação In Situ das Extremidades Cortadas/métodos , Masculino , Camundongos , Microscopia Eletrônica de Transmissão/métodos , Uridina Trifosfato/química
2.
J Cell Biol ; 182(3): 449-58, 2008 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-18678708

RESUMO

Meiosis is critical for sexual reproduction. During meiosis, the dynamics and integrity of homologous chromosomes are tightly regulated. The genetic and molecular mechanisms governing these processes in vivo, however, remain largely unknown. In this study, we demonstrate that Bat3/Scythe is essential for survival and maintenance of male germ cells (GCs). Targeted inactivation of Bat3/Scythe in mice results in widespread apoptosis of meiotic male GCs and complete male infertility. Pachytene spermatocytes exhibit abnormal assembly and disassembly of synaptonemal complexes as demonstrated by abnormal SYCP3 staining and sustained gamma-H2AX and Rad51/replication protein A foci. Further investigation revealed that a testis-specific protein, Hsp70-2/HspA2, is absent in Bat3-deficient male GCs at any stage of spermatogenesis; however, Hsp70-2 transcripts are expressed at normal levels. We found that Bat3 deficiency induces polyubiquitylation and subsequent degradation of Hsp70-2. Inhibition of proteasomal degradation restores Hsp70-2 protein levels. Our findings identify Bat3 as a critical regulator of Hsp70-2 in spermatogenesis, thereby providing a possible molecular target in idiopathic male infertility.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Proteínas Nucleares/deficiência , Processamento de Proteína Pós-Traducional , Espermatogênese , Animais , Apoptose , Proteínas de Transporte/metabolismo , Linhagem Celular , Humanos , Masculino , Camundongos , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Especificidade de Órgãos , Poliubiquitina/metabolismo , Ligação Proteica , Espermatozoides/anormalidades , Espermatozoides/patologia , Complexo Sinaptonêmico/metabolismo , Termodinâmica
3.
J Cell Sci ; 120(Pt 6): 1017-27, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17344431

RESUMO

Programmed double-strand breaks at prophase of meiosis acquire immunologically detectable RAD51-DMC1 foci or early nodules (ENs) that are associated with developing chromosome core segments; each focus is surrounded by a gammaH2AX-modified chromosome domain. The 250-300 ENs per nucleus decline in numbers during the development of full-length cores and the remaining foci are relatively evenly distributed along the mature cores (gamma distribution of nu=2.97). The ENs become transformed nodules (TNs) by the acquisition of RPA, BLM, MSH4 and topoisomerases that function in repair and Holliday junction resolution. At the leptotene-zygotene transition, TNs orient to positions between the aligned cores where they initiate structural interhomolog contacts prior to synaptonemal complex (SC) formation, possibly future crossover sites. Subsequently, TNs are associated with SC extension at the synaptic forks. Dephosphorylation of TN-associated histone gammaH2AX chromatin suggests annealing of single strands or repair of double-strand breaks DSBs at this time. Some 200 TNs per pachytene nucleus are distributed proportional to SC length and are evenly distributed along the SCs (nu= approximately 4). At this stage, gammaH2AX-modified chromatin domains are associated with transcriptionally silenced sex chromosomes and autosomal sites. Immunogold electron microscope evidence shows that one or two TNs of the 10-15 TNs per SC acquire MLH1 protein, the hallmark of reciprocal recombination, whereas the TNs that do not acquire MLH1 protein relocate from their positions along the midline of the SCs to the periphery of the SCs. Relocation of TNs may be associated with the conversion of potential crossovers into non-crossovers.


Assuntos
Troca Genética , Quebras de DNA de Cadeia Dupla , Histonas/fisiologia , Prófase Meiótica I/fisiologia , Recombinação Genética , Complexo Sinaptonêmico/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Cromossomos/fisiologia , Camundongos , Proteína 1 Homóloga a MutL , Proteínas Nucleares/metabolismo , Estágio Paquíteno/fisiologia , Fosforilação , Transporte Proteico , Rad51 Recombinase/metabolismo , Proteína de Replicação A/metabolismo , Cromossomos Sexuais/fisiologia
4.
Genetics ; 175(4): 1561-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17237513

RESUMO

In most eukaryotes, recombination of homologous chromosomes during meiosis is necessary for proper chromosome pairing and subsequent segregation. The molecular mechanisms of meiosis are still relatively unknown, but numerous genes are known to be involved, among which are many mismatch repair genes. One of them, mlh1, colocalizes with presumptive sites of crossing over, but its exact action remains unclear. We studied meiotic processes in a knockout line for mlh1 in zebrafish. Male mlh1 mutants are sterile and display an arrest in spermatogenesis at metaphase I, resulting in increased testis weight due to accumulation of prophase I spermatocytes. In contrast, females are fully fertile, but their progeny shows high rates of dysmorphology and mortality within the first days of development. SNP-based chromosome analysis shows that this is caused by aneuploidy, resulting from meiosis I chromosomal missegregation. Surprisingly, the small percentage of progeny that develops normally has a complete triploid genome, consisting of both sets of maternal and one set of paternal chromosomes. As adults, these triploid fish are infertile males with wild-type appearance. The frequency of triploid progeny of mlh1 mutant females is much higher than could be expected for random chromosome segregation. Together, these results show that multiple solutions exist for meiotic crossover/segregation problems.


Assuntos
Enzimas Reparadoras do DNA/deficiência , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Proteínas de Peixe-Zebra/deficiência , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Aneuploidia , Animais , Sequência de Bases , Troca Genética/genética , Reparo de Erro de Pareamento de DNA , Primers do DNA/genética , Enzimas Reparadoras do DNA/genética , Feminino , Infertilidade Masculina/patologia , Masculino , Meiose/genética , Mutação , Fenótipo , Poliploidia , Túbulos Seminíferos/patologia , Proteínas de Peixe-Zebra/genética
5.
Genome ; 49(3): 205-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16604102

RESUMO

With immunofluorescence microscopy, the positions of centromeres and MLH1 (MutL homolog) foci representing the sites of presumptive chiasmata are shown for zebrafish (Danio rerio Hamilton 1822) synaptonemal complexes (SCs) in spermatocyte nuclei at meiotic prophase. Most SCs have a single focus and a few (7 of 140) have 2 chiasmata. MLH1 foci tend to be in the distal regions of SCs, with progressively fewer occurring towards the middle of the SCs. This non-random distribution suggests chiasma interference. Synaptic initiation, as well as replication protein A (RPA) foci at the chromosome ends, correlates with the distal localization of MLH1 foci. These observations may provide the physical basis for the reported limited genetic recombination in the centromeric region of androgenetic offspring of a male.


Assuntos
Cromossomos/fisiologia , Troca Genética , Peixe-Zebra/genética , Animais , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Centrômero/genética , Centrômero/metabolismo , Pareamento Cromossômico/genética , Pareamento Cromossômico/fisiologia , Cariotipagem , Masculino , Meiose , Recombinação Genética , Espermatócitos/metabolismo , Espermatócitos/ultraestrutura , Espermatogênese , Complexo Sinaptonêmico/genética , Complexo Sinaptonêmico/ultraestrutura
6.
Bioessays ; 27(8): 795-808, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16015600

RESUMO

Several DNA-damage detection and repair mechanisms have evolved to repair double-strand breaks induced by mutagens. Later in evolutionary history, DNA single- and double-strand cuts made possible immune diversity by V(D)J recombination and recombination at meiosis. Such cuts are induced endogenously and are highly regulated and controlled. In meiosis, DNA cuts are essential for the initiation of homologous recombination, and for the formation of joint molecule and crossovers. Many proteins that function during somatic DNA-damage detection and repair are also active during homologous recombination. However, their meiotic functions may be altered from their somatic roles through localization, posttranslational modifications and/or interactions with meiosis-specific proteins. Presumably, somatic repair functions and meiotic recombination diverged during evolution, resulting in adaptations specific to sexual reproduction. (c) 2005 Wiley Periodicals, Inc.


Assuntos
Meiose , Animais , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Pareamento Incorreto de Bases , Cromossomos/ultraestrutura , DNA/metabolismo , Dano ao DNA , Reparo do DNA , Humanos , Camundongos , Modelos Biológicos , Modelos Genéticos , Mutação , Fosforilação , Recombinação Genética , Fatores de Tempo , VDJ Recombinases/metabolismo
7.
Chromosoma ; 114(2): 92-102, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15983832

RESUMO

Genetic modifications causing germ cell death during meiotic prophase in the mouse frequently have sexually dimorphic phenotypes where oocytes reach more advanced stages than spermatocytes. To determine to what extent these dimorphisms are due to differences in male versus female meiotic prophase development, we compared meiotic chromosome events in the two sexes in both wild-type and mutant mice. We report the abundance and time course of appearance of structural and recombination-related proteins of fetal oocyte nuclei. Oocytes at successive days post coitus show rapid, synchronous meiotic prophase development compared with the continuous spermatocyte development in adult testis. Consequently, a genetic defect requiring 2-3 days from the onset of prophase to reach arrest registers pachytene as the developmental endpoint in oocytes. Pachytene spermatocytes, on the other hand, which normally accumulate during days 4-10 after the onset of prophase, will be rare, giving the appearance of an earlier endpoint than in oocytes. We conclude that these different logistics create apparent sexually dimorphic endpoints. For more pronounced sexual dimorphisms, we examined meiotic prophase of mice with genetic modifications of meiotic chromosome core components that cause male but not female sterility. The correlations between male sterility and alterations in the organization of the sex chromosome cores and X-Y chromatin may indicate that impaired signals from the XY domain (XY chromosome cores, chromatin, dense body and sex body) may interfere with the progression of the spermatocyte through prophase. Oocytes, in the absence of the X-Y pair, do not suffer such defects.


Assuntos
Infertilidade Masculina/genética , Espermatócitos/fisiologia , Cromossomo X/genética , Cromossomo Y/genética , Animais , Proteína BRCA1/genética , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Oócitos/fisiologia
8.
Mol Hum Reprod ; 10(12): 917-24, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15489243

RESUMO

During meiotic prophase, chromosomes must undergo highly regulated recombination events, some of which lead to reciprocal exchanges. In yeast, MSH4, a meiosis-specific homologue of the bacterial MutS protein, is required for meiotic recombination. In mice, disruption of the Msh4 gene results in male and female infertility due to meiotic failure. To date, the implication of MSH4 mutations has not been established in human sterility. However, it is noteworthy that mutant mice exhibit a defect in the chromosome synapsis, strikingly similar to the clinical observations found in human infertility. As a step towards understanding the molecular mechanisms underlying the role of MSH4 in human gametogenesis, we decided to determine whether this protein interacts with recombination machinery enzymes. Our results provide biochemical evidence indicating that the human MSH4 protein physically interacts with both RAD51 and DMC1, two RecA homologues known to initiate DNA strand-exchange between homologous chromosomes. Immunolocalization analyses show that some MSH4 foci, located on mouse meiotic chromosomes, colocalize with DMC1/RAD51 complexes. Our data support the view that MSH4 is associated with the early meiotic recombination machinery in mammals. We consider the possibility that MSH4 is involved in the regulation of recombination events by exerting a function closely after DNA strand-exchange has been initiated.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Meiose/fisiologia , Animais , Anticorpos/imunologia , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/genética , Núcleo Celular/química , Núcleo Celular/metabolismo , Cromossomos/química , Cromossomos/metabolismo , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Humanos , Imunoprecipitação , Masculino , Meiose/genética , Camundongos , Proteínas Nucleares , Proteínas de Ligação a Fosfato , Rad51 Recombinase , Recombinação Genética/genética , Recombinação Genética/fisiologia , Espermatócitos/química , Espermatócitos/metabolismo , Técnicas do Sistema de Duplo-Híbrido
9.
Chromosoma ; 112(7): 323-30, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15138768

RESUMO

Topoisomerase IIbeta binding protein 1 (TopBP1), previously shown to localise to sites of DNA damage and to stalled replication forks, has been implicated in DNA replication and in DNA damage response. In this work we showed that TopBP1 was localised in structures other than stalled replication forks. In late mitosis TopBP1 localises to centrosomes in a manner similar to other DNA damage response proteins such as BRCA1 and p53. Spindle checkpoint activation does not affect this centrosomal localisation. Moreover, in the testis, we detected high levels of TopBP1 associated with meiotic prophase chromosome cores and the X-Y pair. Together, these data suggest a direct role of TopBP1 during both mitosis and meiotic prophase I.


Assuntos
Proteínas de Transporte/análise , Centrossomo/química , Cromossomos/química , Prófase Meiótica I , Mitose , Anáfase , Proteína BRCA1/análise , Proteína BRCA1/genética , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Linhagem Celular Tumoral , Cromossomos/ultraestrutura , Proteínas de Ligação a DNA , Ativação Enzimática , Feminino , Células HeLa , Humanos , Masculino , Metáfase , Nocodazol/farmacologia , Proteínas Nucleares , Pirimidinas/farmacologia , Espermatogênese , Testículo/química , Testículo/citologia , Tionas/farmacologia , Fatores de Tempo
10.
Proc Natl Acad Sci U S A ; 101(17): 6496-501, 2004 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-15084742

RESUMO

Telomere shortening can lead to chromosome instability, replicative senescence, and apoptosis in both somatic and male germ cells. To study roles for mammalian telomeres in homologous pairing and recombination, we characterized effects of telomere shortening on spermatogenesis and oogenesis in late-generation telomerase-deficient mice. We show that shortened telomeres of late-generation telomerase-deficient mice impair meiotic synapsis and decrease recombination, in particular, in females. In response to telomere shortening, male germ cells mostly undergo apoptosis, whereas female germ cells preferentially arrest in early meiosis, suggesting sexually dimorphic surveillance mechanisms for telomere dysfunction during meiosis in mice. Further, meiocytes of late-generation telomerase-deficient females with shortened telomeres, bred with early-generation males harboring relatively long telomeres, exhibit severely impaired chromosome pairing and synapsis and reduced meiotic recombination. These findings imply that functional telomeres are important in mammalian meiotic synapsis and recombination.


Assuntos
Pareamento Cromossômico/genética , Recombinação Genética/genética , Telômero , Animais , Feminino , Hibridização in Situ Fluorescente , Marcação In Situ das Extremidades Cortadas , Camundongos , Microscopia de Fluorescência/métodos
11.
Dev Biol ; 268(2): 403-15, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15063176

RESUMO

The acquisition of genomic methylation in the male germ line is initiated prenatally in diploid gonocytes, while DNA methylation in the female germ line is initiated postnatally in growing oocytes. We compared the temporal expression patterns of the DNA methyltransferases, DNMT1, DNMT3a, DNMT3b, and DNMT3l in the male and female germ lines. DNMT1 expression was examined by immunocytochemistry and Northerns with an emphasis on the prenatal period. In the female, there is a gradual down-regulation of DNMT1 protein in prenatal meiotic prophase I oocytes that is not associated with the production of an untranslated transcript, as it is in the male; these results suggest that the mechanism of meiotic down-regulation differs between the sexes. In the male, DNMT1 is unlikely to play a role in the prenatal acquisition of germ line methylation patterns since it is down-regulated in gonocytes between 14.5 and 18.5 days of gestation and is absent at the time of initiation of DNA methylation. To search for candidate DNMTs that could be involved in establishing methylation patterns in both germ lines, real-time RT-PCR was used to simultaneously study the expression profiles of the three DNMT3 enzymes in developing testes and ovaries; DNMT1 expression was included as a control. Expression profiles of DNMT3a and DNMT3l provide support for an interaction of the two enzymes during prenatal germ cell development and de novo methylation in the male. DNMT3l is the predominant DNMT3 enzyme expressed at high levels in the postnatal female germ line at the time of acquisition of DNA methylation patterns. DNMT1 and DNMT3b expression levels peak concomitantly, shortly after birth in the male, consistent with a role in the maintenance of methylation patterns in proliferating spermatogonia. Together, the results provide clues to specific roles for the different DNMT family members in de novo and maintenance methylation in the developing testis and ovary.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA , DNA Metiltransferase 3A , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Imuno-Histoquímica , Masculino , Camundongos , Óvulo/metabolismo , Espermatozoides/metabolismo , DNA Metiltransferase 3B
12.
Mol Biol Cell ; 15(4): 1568-79, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14718568

RESUMO

Mammalian TopBP1 is a BRCT domain-containing protein whose function in mitotic cells is linked to replication and DNA damage checkpoint. Here, we study its possible role during meiosis in mice. TopBP1 foci are abundant during early prophase I and localize mainly to histone gamma-H2AX-positive domains, where DNA double-strand breaks (required to initiate recombination) occur. Strikingly, TopBP1 showed a pattern almost identical to that of ATR, a PI3K-like kinase involved in mitotic DNA damage checkpoint. In the synapsis-defective Fkbp6(-/-) mouse, TopBP1 heavily stains unsynapsed regions of chromosomes. We also tested whether Schizosaccharomyces pombe Cut5 (the TopBP1 homologue) plays a role in the meiotic recombination checkpoint, like spRad3, the ATR homologue. Indeed, we found that a cut5 mutation suppresses the checkpoint-dependent meiotic delay of a meiotic recombination defective mutant, indicating a direct role of the Cut5 protein in the meiotic checkpoint. Our findings suggest that ATR and TopBP1 monitor meiotic recombination and are required for activation of the meiotic recombination checkpoint.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Ciclo Celular/fisiologia , Cromossomos/ultraestrutura , Meiose , Proteínas Serina-Treonina Quinases/fisiologia , Alelos , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte/biossíntese , Proteínas de Ciclo Celular/biossíntese , Núcleo Celular/metabolismo , Sobrevivência Celular , Cruzamentos Genéticos , Dano ao DNA , Proteínas de Ligação a DNA , Diploide , Histonas/metabolismo , Immunoblotting , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Proteínas Nucleares , Fosfatidilinositol 3-Quinases/metabolismo , Prófase , Proteínas Serina-Treonina Quinases/biossíntese , Estrutura Terciária de Proteína , Recombinação Genética , Schizosaccharomyces/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Testículo/metabolismo , Fatores de Tempo
13.
Genome ; 46(6): 936-7, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14663508

RESUMO

The nature of meiotic genetic recombination was resolved at the DNA level by the 1953 Watson and Crick model. What remains to be determined are the roles of the various recombination proteins and the distribution and localization of recombination events in the meiotic prophase nucleus.


Assuntos
DNA/genética , Meiose/genética , Recombinação Genética/genética , DNA/química , Conformação de Ácido Nucleico
14.
Science ; 300(5623): 1291-5, 2003 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-12764197

RESUMO

Meiosis is a critical stage of gametogenesis in which alignment and synapsis of chromosomal pairs occur, allowing for the recombination of maternal and paternal genomes. Here we show that FK506 binding protein (Fkbp6) localizes to meiotic chromosome cores and regions of homologous chromosome synapsis. Targeted inactivation of Fkbp6 in mice results in aspermic males and the absence of normal pachytene spermatocytes. Moreover, we identified the deletion of Fkbp6 exon 8 as the causative mutation in spontaneously male sterile as/as mutant rats. Loss of Fkbp6 results in abnormal pairing and misalignments between homologous chromosomes, nonhomologous partner switches, and autosynapsis of X chromosome cores in meiotic spermatocytes. Fertility and meiosis are normal in Fkbp6 mutant females. Thus, Fkbp6 is a component of the synaptonemal complex essential for sex-specific fertility and for the fidelity of homologous chromosome pairing in meiosis.


Assuntos
Pareamento Cromossômico/fisiologia , Fertilidade/fisiologia , Infertilidade Masculina/fisiopatologia , Meiose , Complexo Sinaptonêmico/fisiologia , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/fisiologia , Sequência de Aminoácidos , Animais , Apoptose , Proteínas de Ciclo Celular , Clonagem Molecular , Proteínas de Ligação a DNA , Éxons , Feminino , Marcação de Genes , Humanos , Infertilidade Masculina/genética , Masculino , Camundongos , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oogênese , Ovário/fisiologia , Prófase , Ratos , Deleção de Sequência , Espermátides/fisiologia , Espermatócitos/fisiologia , Espermatócitos/ultraestrutura , Espermatogênese , Proteínas de Ligação a Tacrolimo/química , Testículo/fisiologia , Cromossomo X/fisiologia
15.
Proc Natl Acad Sci U S A ; 100(5): 2526-31, 2003 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-12591952

RESUMO

We report that disruption of the mouse TOP3 beta gene encoding DNA topoisomerase III beta, one of the two mammalian type IA DNA topoisomerases, leads to a progressive reduction in fecundity. The litter size in crosses of top3 beta(-/-) mice decreases over time and through successive generations, and this decrease seems to reflect embryonic death rather than impaired fertilization. These observations are suggestive of a gradual accumulation of chromosomal defects in germ cells lacking DNA topoisomerase III beta, and this interpretation is supported by the observation of a high incidence of aneuploidy in the spermatocytes of infertile top3 beta(-/-) males. Cytogenetic examination of spermatocytes of wild-type mice also indicates that DNA topoisomerase III beta becomes prominently associated with the asynaptic regions of the XY bivalents during pachytene, and that there is a time lag between the appearance of chromosome-bound DNA topoisomerase III beta and Rad51, a protein known to be involved in an early step of homologous recombination. We interpret these findings, together with the known mechanistic characteristics of different subfamilies of DNA topoisomerases, in terms of a specific role of a type IA DNA topoisomerase in the resolution of meiotic double-Holliday junctions without crossing over. This interpretation is most likely applicable to mitotic cells as well and can explain the universal presence of at least one type IA DNA topoisomerase in all organisms.


Assuntos
Aneuploidia , DNA Topoisomerases Tipo I/genética , DNA Topoisomerases Tipo I/fisiologia , Infertilidade , Isoenzimas/genética , Isoenzimas/fisiologia , Animais , Aberrações Cromossômicas , Cruzamentos Genéticos , Proteínas de Ligação a DNA/metabolismo , Feminino , Genótipo , Hibridização in Situ Fluorescente , Masculino , Meiose , Metáfase , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Rad51 Recombinase , Recombinação Genética , Espermatócitos/citologia , Fatores de Tempo
16.
Nat Genet ; 31(4): 385-90, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12091911

RESUMO

MutL homolog 3 (Mlh3) is a member of a family of proteins conserved during evolution and having dual roles in DNA mismatch repair and meiosis. The pathway in eukaryotes consists of the DNA-binding components, which are the homologs of the bacterial MutS protein (MSH 2 6), and the MutL homologs, which bind to the MutS homologs and are essential for the repair process. Three of the six homologs of MutS that function in these processes, Msh2, Msh3 and Msh6, are involved in the mismatch repair of mutations, frameshifts and replication errors, and two others, Msh4 and Msh5, have specific roles in meiosis. Of the four MutL homologs, Mlh1, Mlh3, Pms1 and Pms2, three are involved in mismatch repair and at least two, Pms2 and Mlh1, are essential for meiotic progression in both yeast and mice. To assess the role of Mlh3 in mammalian meiosis, we have generated and characterized Mlh3(-/-) mice. Here we show that Mlh3(-/-) mice are viable but sterile. Mlh3 is required for Mlh1 binding to meiotic chromosomes and localizes to meiotic chromosomes from the mid pachynema stage of prophase I. Mlh3(-/-) spermatocytes reach metaphase before succumbing to apoptosis, but oocytes fail to complete meiosis I after fertilization. Our results show that Mlh3 has an essential and distinct role in mammalian meiosis.


Assuntos
Aneuploidia , Proteínas de Transporte/genética , Enzimas Reparadoras do DNA , Infertilidade Feminina/genética , Meiose , Proteínas Adaptadoras de Transdução de Sinal , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Apoptose/genética , Proteínas de Transporte/metabolismo , Cromossomos/genética , Cromossomos/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Éxons , Feminino , Fertilização in vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Repetições de Microssatélites , Endonuclease PMS2 de Reparo de Erro de Pareamento , Dados de Sequência Molecular , Proteína 1 Homóloga a MutL , Proteínas MutL , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Oócitos/patologia , Análise de Sequência , Espermatócitos/metabolismo , Testículo/anormalidades
17.
J Cell Sci ; 115(Pt 8): 1611-22, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11950880

RESUMO

During mouse meiosis, the early prophase RAD51/DMC1 recombination protein sites, which are associated with the chromosome cores and which serve as markers for ongoing DNA-DNA interactions, are in ten-fold excess of the eventual reciprocal recombinant events. Most, if not all, of these early interactions are eliminated as prophase progresses. The manner in which these sites are eliminated is the focus of this investigation. We report that these sites acquire replication protein A, RPA and the Escherichia coli MUTS homologue, MSH4p, and somewhat later the Bloom helicase, BLM, while simultaneously losing the RAD51/DMC1 component. Eventually the RPA component is also lost and BLM sites remain. At that time, the MUTL homologue, MLH1p, which is essential for reciprocal recombination in the mouse, appears in numbers and locations that correspond to the distribution of reciprocal recombination events. However, the MLH1 foci do not appear to coincide with the remaining BLM sites. The MLH1p is specifically localized to electron-microscope-defined recombination nodules. We consider the possibility that the homology-search RAD51/DMC1 complexes are involved in homologous chromosome synapsis but that most of these early DNA-DNA interactions are later resolved by the anti-recombination RPA/MSH4/BLM-topoisomerase complex, thereby preventing the formation of superfluous reciprocal recombinant events.


Assuntos
Núcleo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos/metabolismo , DNA/metabolismo , Meiose/fisiologia , Recombinação Genética , Proteínas de Saccharomyces cerevisiae , Proteínas Adaptadoras de Transdução de Sinal , Adenosina Trifosfatases/metabolismo , Animais , Cromossomos/ultraestrutura , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Masculino , Camundongos , Microscopia Imunoeletrônica , Modelos Genéticos , Proteína 1 Homóloga a MutL , Proteínas Nucleares/metabolismo , Rad51 Recombinase , Ratos , RecQ Helicases , Espermatócitos/citologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA