Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Biol Res ; 57(1): 36, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822414

RESUMEN

BACKGROUND: Helicase for meiosis 1 (HFM1), a putative DNA helicase expressed in germ-line cells, has been reported to be closely associated with premature ovarian insufficiency (POI). However, the underlying molecular mechanism has not been clearly elucidated. The aim of this study was to investigate the function of HFM1 in the first meiotic prophase of mouse oocytes. RESULTS: The results suggested that the deficiency of HFM1 resulting in increased apoptosis and depletion of oocytes in mice, while the oocytes were arrested in the pachytene stage of the first meiotic prophase. In addition, impaired DNA double-strand break repair and disrupted synapsis were observed in the absence of HFM1. Further investigation revealed that knockout of HFM1 promoted ubiquitination and degradation of FUS protein mediated by FBXW11. Additionally, the depletion of HFM1 altered the intranuclear localization of FUS and regulated meiotic- and oocyte development-related genes in oocytes by modulating the expression of BRCA1. CONCLUSIONS: These findings elaborated that the critical role of HFM1 in orchestrating the regulation of DNA double-strand break repair and synapsis to ensure meiosis procession and primordial follicle formation. This study provided insights into the pathogenesis of POI and highlighted the importance of HFM1 in maintaining proper meiotic function in mouse oocytes.


Asunto(s)
Profase Meiótica I , Oocitos , Ubiquitinación , Animales , Femenino , Ratones , Apoptosis/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Meiosis/fisiología , Profase Meiótica I/fisiología , Ratones Noqueados , Oocitos/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteína FUS de Unión a ARN/genética
2.
PLoS Genet ; 18(2): e1010040, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35130272

RESUMEN

During meiotic prophase I, homologous chromosomes pair, synapse and recombine in a tightly regulated process that ensures the generation of genetically variable haploid gametes. Although the mechanisms underlying meiotic cell division have been well studied in model species, our understanding of the dynamics of meiotic prophase I in non-traditional model mammals remains in its infancy. Here, we reveal key meiotic features in previously uncharacterised marsupial species (the tammar wallaby and the fat-tailed dunnart), plus the fat-tailed mouse opossum, with a focus on sex chromosome pairing strategies, recombination and meiotic telomere homeostasis. We uncovered differences between phylogroups with important functional and evolutionary implications. First, sex chromosomes, which lack a pseudo-autosomal region in marsupials, had species specific pairing and silencing strategies, with implications for sex chromosome evolution. Second, we detected two waves of γH2AX accumulation during prophase I. The first wave was accompanied by low γH2AX levels on autosomes, which correlated with the low recombination rates that distinguish marsupials from eutherian mammals. In the second wave, γH2AX was restricted to sex chromosomes in all three species, which correlated with transcription from the X in tammar wallaby. This suggests non-canonical functions of γH2AX on meiotic sex chromosomes. Finally, we uncover evidence for telomere elongation in primary spermatocytes of the fat-tailed dunnart, a unique strategy within mammals. Our results provide new insights into meiotic progression and telomere homeostasis in marsupials, highlighting the importance of capturing the diversity of meiotic strategies within mammals.


Asunto(s)
Emparejamiento Cromosómico/fisiología , Cromosomas Sexuales/fisiología , Telómero/fisiología , Animales , Macropodidae/genética , Marsupiales/genética , Meiosis/genética , Meiosis/fisiología , Profase Meiótica I/fisiología , Zarigüeyas/genética , Cromosomas Sexuales/genética , Telómero/genética , Cromosoma X/genética , Cromosoma Y/genética
3.
Cell Rep ; 38(4): 110289, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35081355

RESUMEN

The meiosis-specific telomere-binding protein TERB1 anchors telomeres to the nuclear envelope and drives chromosome movements for the pairing of homologous chromosomes. TERB1 has an MYB-like DNA-binding (MYB) domain, which is a hallmark of telomeric DNA-binding proteins. Here, we demonstrate that the TERB1 MYB domain has lost its canonical DNA-binding activity. The analysis of Terb1 point mutant mice expressing TERB1 lacking its MYB domain showed that the MYB domain is dispensable for telomere localization of TERB1 and the downstream TERB2-MAJIN complex, the promotion of homologous pairing, and even fertility. Instead, the TERB1 MYB domain regulates the enrichment of cohesin and promotes the remodeling of axial elements in the early-to-late pachytene transition, which suppresses telomere erosion. Considering its conservation across metazoan phyla, the TERB1 MYB domain is likely to be important for the maintenance of telomeric DNA and thus for genomic integrity by suppressing meiotic telomere erosion over long evolutionary timescales.


Asunto(s)
Profase Meiótica I/fisiología , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Dominios Proteicos
4.
PLoS Genet ; 17(8): e1009753, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34388164

RESUMEN

Meiosis is essential for the generation of gametes and sexual reproduction, yet the factors and underlying mechanisms regulating meiotic progression remain largely unknown. Here, we showed that MTL5 translocates into nuclei of spermatocytes during zygotene-pachytene transition and ensures meiosis advances beyond pachytene stage. MTL5 shows strong interactions with MuvB core complex components, a well-known transcriptional complex regulating mitotic progression, and the zygotene-pachytene transition of MTL5 is mediated by its direct interaction with the component LIN9, through MTL5 C-terminal 443-475 residues. Male Mtl5c-mu/c-mu mice expressing the truncated MTL5 (p.Ser445Arg fs*3) that lacks the interaction with LIN9 and is detained in cytoplasm showed male infertility and spermatogenic arrest at pachytene stage, same as that of Mtl5 knockout mice, indicating that the interaction with LIN9 is essential for the nuclear translocation and function of MTL5 during meiosis. Our data demonstrated MTL5 translocates into nuclei during the zygotene-pachytene transition to initiate its function along with the MuvB core complex in pachytene spermatocytes, highlighting a new mechanism regulating the progression of male meiosis.


Asunto(s)
Meiosis/fisiología , Metalotioneína/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Proteínas de Ciclo Celular/metabolismo , Emparejamiento Cromosómico/genética , Citoplasma , Proteínas de Unión al ADN , Fertilidad/genética , Fertilidad/fisiología , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Masculino , Profase Meiótica I/fisiología , Metalotioneína/genética , Ratones , Ratones Endogámicos C57BL , Fase Paquiteno/genética , Espermatocitos/fisiología , Espermatogénesis/fisiología , Testículo , Proteínas Supresoras de Tumor/fisiología
5.
PLoS Genet ; 16(11): e1009067, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33206637

RESUMEN

Mammalian oogonia proliferate without completing cytokinesis, forming cysts. Within these, oocytes differentiate and initiate meiosis, promoting double-strand break (DSBs) formation, which are repaired by homologous recombination (HR) causing the pairing and synapsis of the homologs. Errors in these processes activate checkpoint mechanisms, leading to apoptosis. At the end of prophase I, in contrast with what is observed in spermatocytes, oocytes accumulate unrepaired DSBs. Simultaneously to the cyst breakdown, there is a massive oocyte death, which has been proposed to be necessary to enable the individualization of the oocytes to form follicles. Based upon all the above-mentioned information, we hypothesize that the apparently inefficient HR occurring in the oocytes may be a requirement to first eliminate most of the oocytes and enable cyst breakdown and follicle formation. To test this idea, we compared perinatal ovaries from control and mutant mice for the effector kinase of the DNA Damage Response (DDR), CHK2. We found that CHK2 is required to eliminate ~50% of the fetal oocyte population. Nevertheless, the number of oocytes and follicles found in Chk2-mutant ovaries three days after birth was equivalent to that of the controls. These data revealed the existence of another mechanism capable of eliminating oocytes. In vitro inhibition of CHK1 rescued the oocyte number in Chk2-/- mice, implying that CHK1 regulates postnatal oocyte death. Moreover, we found that CHK1 and CHK2 functions are required for the timely breakdown of the cyst and to form follicles. Thus, we uncovered a novel CHK1 function in regulating the oocyte population in mice. Based upon these data, we propose that the CHK1- and CHK2-dependent DDR controls the number of oocytes and is required to properly break down oocyte cysts and form follicles in mammals.


Asunto(s)
Daño del ADN/genética , Oogonios/metabolismo , Folículo Ovárico/metabolismo , Animales , Apoptosis/fisiología , Proteínas de Ciclo Celular/genética , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , Quistes/metabolismo , Daño del ADN/fisiología , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Femenino , Meiosis/fisiología , Profase Meiótica I/fisiología , Ratones , Ratones Endogámicos C57BL , Oocitos/metabolismo , Oocitos/fisiología , Oogonios/fisiología , Folículo Ovárico/fisiología , Ovario/metabolismo , Progesterona/metabolismo
6.
Reproduction ; 160(3): 331-341, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32520724

RESUMEN

In female mammals, reproductive potential is determined during fetal life by the formation of a non-renewable pool of primordial follicles. Initiation of meiosis is one of the defining features of germ cell differentiation and is well established to commence in response to retinoic acid. WIN 18,446 inhibits the conversion of retinol to retinoic acid, and therefore it was used to explore the impact of reduced retinoic acid synthesis on meiotic progression and thus germ cell development and subsequent primordial follicle formation. e13.5 mouse fetal ovaries were cultured in vitro and treated with WIN 18,446 for the first 3 days of a total of up to 12 days. Doses as low as 0.01 µM reduced transcript levels of the retinoic acid response genes Stra8 and Rarß without affecting germ cell number. Higher doses resulted in germ cell loss, rescued with the addition of retinoic acid. WIN 18,446 significantly accelerated the progression of prophase I; this was seen as early as 48 h post treatment using meiotic chromosome spreads and was still evident after 12 days of culture using Tra98/Msy2 immunostaining. Furthermore, ovaries treated with WIN 18,446 at e13.5 but not at P0 had a higher proportion of growing follicles compared to vehicle controls, thus showing evidence of increased follicle activation. These data therefore indicate that retinoic acid is not necessary for meiotic progression but may have a role in the regulation of its progression and germ cell survival at that time and provide evidence for a link between meiotic arrest and follicle growth initiation.


Asunto(s)
Feto/fisiología , Profase Meiótica I/fisiología , Folículo Ovárico/fisiología , Ovario/fisiología , Tretinoina/metabolismo , Animales , Femenino , Feto/citología , Ratones , Folículo Ovárico/citología , Ovario/citología , Tretinoina/antagonistas & inhibidores
7.
Cell ; 181(6): 1442-1442.e1, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32531249

RESUMEN

Meiosis is the specialized cell division that generates haploid gametes and is therefore essential for sexual reproduction. This SnapShot encompasses key events taking place during prophase I of meiosis that are required for achieving proper chromosome segregation and highlights how these are both conserved and diverged throughout five different species. To view this SnapShot, open or download the PDF.


Asunto(s)
Meiosis/fisiología , Profase Meiótica I/fisiología , Animales , Arabidopsis/fisiología , Caenorhabditis elegans/fisiología , Segregación Cromosómica/fisiología , Drosophila melanogaster/fisiología , Ratones , Saccharomyces cerevisiae/fisiología
9.
Mol Reprod Dev ; 87(1): 91-101, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31749232

RESUMEN

Formation and subsequent break down of ovarian germ cell (GC) cysts is a key and an evolutionary-conserved developmental event, described in phylogenetically diverse species of invertebrates and vertebrates. In mammals, cyst break down (CBD) ends at the time of, or soon after, birth with the formation of primordial follicles enclosing single oocytes, which constitute the sole reservoir of gametes available through the whole female's reproductive life. In this study, we challenge this paradigm demonstrating the constitutive presence of a large number of cysts, enclosing two-thirty GCs, in the ovary of the adult armadillo Chaetophractus villosus, belonging to the superorder Xenarthra, one of the earliest offshoots among placentals. We also describe that (a) GCs enclosed within cysts are connected by intercellular bridges-intercellular bridges-markers of their clonal origin; (b) CBD occurs through four main phases, ending with primordial follicles containing single oocytes; (c) GCs encompass meiotic prophase I stages, from leptotene to diplotene; (d) seasonal variations in the number of GCs enclosed within cysts, suggesting the presence of a GC multiplying activity. The armadillo C. villosus''s ovary emerges as an extraordinary resource to investigate folliculogenesis and to explore the evolutionary past of the mammalian ovary.


Asunto(s)
Armadillos/crecimiento & desarrollo , Profase Meiótica I/fisiología , Oocitos/citología , Oogénesis/fisiología , Folículo Ovárico/crecimiento & desarrollo , Animales , Femenino , Folículo Ovárico/citología , Estaciones del Año
10.
Cell Cycle ; 18(21): 2986-2997, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31530151

RESUMEN

Calcium (Ca2+)/calmodulin-dependent protein kinase II (CaMKII) had been reported to play a role in the process of fertilization. However, the role of CaMKII in the release of diplotene-arrested oocytes is poorly understood. In this study, we explored the potential effect of CaMKII on Akt1 and the relationship among CaMKII, Akt1 and phosphatidylinositol (3,4,5)-trisphosphate (PIP3) during the meiotic resumption of mouse oocytes. We found that inhibition of CaMKII aggravated diplotene arrest. We detected the expression and distribution of pCaMKII (Thr286), pAkt1 (Ser473), Cdc25B and pCdc2 (Tyr15) when oocytes were treated with KN-93, SH-6, LY294002 or PIP3, respectively. Our data showed that down-regulated CaMKII by KN-93 decreased the levels of pAkt1 (Ser473) and rearranged the distribution of pAkt1 (Ser473). Meanwhile, down-regulated pAkt1 (Ser473) by SH-6 also decreased the levels of pCaMKII (Thr286), Cdc25B and pCdc2 (Tyr15) significantly and rearranged the distributions of pCaMKII (Thr286). Furthermore, our data showed that exogenous PIP3 up-regulated GVBD rates significantly and increased the levels of pCaMKII (Thr286) and pAkt1 (Ser473). On the contrary, down-regulation of PIP3 by LY294002 decreased GVBD rates and the levels of pCaMKII (Thr286) and pAkt1 (Ser473), respectively. Our results showed that Akt1 and CaMKII regulated each other, and PIP3 may be involved in these regulations during the release of mouse oocytes from diplotene arrest.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Profase Meiótica I/fisiología , Oocitos/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Proteína Quinasa CDC2/biosíntesis , Femenino , Ratones , Oocitos/crecimiento & desarrollo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Fosfatasas cdc25/biosíntesis
11.
J Cell Biol ; 218(9): 2896-2918, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31350280

RESUMEN

Meiosis generates four genetically distinct haploid gametes over the course of two reductional cell divisions. Meiotic divisions are characterized by the coordinated deposition and removal of various epigenetic marks. Here we propose that nuclear respiratory factor 1 (NRF1) regulates transcription of euchromatic histone methyltransferase 1 (EHMT1) to ensure normal patterns of H3K9 methylation during meiotic prophase I. We demonstrate that cyclin-dependent kinase (CDK2) can bind to the promoters of a number of genes in male germ cells including that of Ehmt1 through interaction with the NRF1 transcription factor. Our data indicate that CDK2-mediated phosphorylation of NRF1 can occur at two distinct serine residues and negatively regulates NRF1 DNA binding activity in vitro. Furthermore, induced deletion of Cdk2 in spermatocytes results in increased expression of many NRF1 target genes including Ehmt1 We hypothesize that the regulation of NRF1 transcriptional activity by CDK2 may allow the modulation of Ehmt1 expression, therefore controlling the dynamic methylation of H3K9 during meiotic prophase.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Regulación Enzimológica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/biosíntesis , Profase Meiótica I/fisiología , Factor Nuclear 1 de Respiración/metabolismo , Espermatocitos/metabolismo , Animales , Quinasa 2 Dependiente de la Ciclina/genética , Eliminación de Gen , N-Metiltransferasa de Histona-Lisina/genética , Masculino , Ratones , Ratones Noqueados , Factor Nuclear 1 de Respiración/genética , Espermatocitos/citología
12.
J Cell Biochem ; 120(9): 15057-15068, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31081178

RESUMEN

Late diplotene oocytes are characterized by an essential decrease in transcriptional activity. At this time, chromosomes condense and form a compact structure named a karyosphere. The karyosphere of grass frogs Rana temporaria is surrounded by a fibrillar karyosphere capsule (KC). One of the main protein constituents of R. temporaria KC is actin. In this study, we used antibodies against different actin epitopes to trace different forms of actin in the KC. We also investigated the effect of F-actin depolymerization on the oocyte nuclear structures and transcription of chromatin DNA and rDNA in the amplified nucleoli. It was determined that disruption of actin filaments leads to chromosome shrinkage, nucleoli fusion, and distortion of the KC structure, but does not inhibit residual transcription in both the karyosphere and the nucleoli.


Asunto(s)
Citoesqueleto de Actina/fisiología , Actinas/metabolismo , Nucléolo Celular/metabolismo , Oocitos/metabolismo , Transcripción Genética/fisiología , Actinas/inmunología , Animales , Cromatina/metabolismo , Cromosomas/metabolismo , Epítopos/inmunología , Femenino , Profase Meiótica I/fisiología , Rana temporaria
13.
BMC Biol ; 17(1): 23, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30866939

RESUMEN

BACKGROUND: Female mammals have a limited reproductive lifespan determined by the size of the primordial follicle pool established perinatally. Over two thirds of fetal oocytes are abolished via programmed cell death during early folliculogenesis. However, the underlying mechanisms governing fetal oocyte attrition remain largely elusive. RESULTS: Here, we demonstrate that glycogen synthase kinase-3 beta (GSK-3ß) is indispensable for fetal oocyte maintenance during meiotic prophase I in mice. In vitro inhibition of GSK-3ß activity or in vivo conditional deletion of Gsk-3ß in the germline led to a dramatic loss of fetal oocytes via apoptosis, which subsequently resulted in a reduced capacity of the primordial follicle pool. Inhibition of GSK-3ß also impeded meiotic progression in fetal oocytes and led to a deficiency in DNA double-strand break (DSB) repair associated with premature upregulation of Tap63, the major genome guardian of the female germline, following GSK-3ß inhibition in fetal ovaries. Mechanistically, we demonstrated that aberrant nuclear translocation of ß-catenin was responsible for the abnormal expression of TAp63 and global fetal oocyte attrition following GSK-3ß inhibition. CONCLUSIONS: In summary, GSK-3ß was essential for sustaining fetal oocyte survival and folliculogenesis via fine-tuning the cytoplasmic-nuclear translocation of ß-catenin, which in turn modulates timely TAp63 expression during meiotic prophase I in mice. Our study provides a perspective on the physiological regulatory role of DNA damage checkpoint signaling in fetal oocyte guardianship and female fertility.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Oocitos/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Transactivadores/metabolismo , Animales , Apoptosis/fisiología , Daño del ADN/fisiología , Femenino , Glucógeno Sintasa Quinasa 3 beta/genética , Profase Meiótica I/fisiología , Ratones , Fosfoproteínas/genética , Transactivadores/genética , Regulación hacia Arriba , beta Catenina/metabolismo
14.
Chromosoma ; 128(3): 199-214, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30826870

RESUMEN

Meiosis is a conserved cell division process that is used by sexually reproducing organisms to generate haploid gametes. Males and females produce different end products of meiosis: eggs (females) and sperm (males). In addition, these unique end products demonstrate sex-specific differences that occur throughout meiosis to produce the final genetic material that is packaged into distinct gametes with unique extracellular morphologies and nuclear sizes. These sexually dimorphic features of meiosis include the meiotic chromosome architecture, in which both the lengths of the chromosomes and the requirement for specific meiotic axis proteins being different between the sexes. Moreover, these changes likely cause sex-specific changes in the recombination landscape with the sex that has the longer chromosomes usually obtaining more crossovers. Additionally, epigenetic regulation of meiosis may contribute to sexually dimorphic recombination landscapes. Here we explore the sexually dimorphic features of both the chromosome axis and crossing over for each stage of meiotic prophase I in Mus musculus, Caenorhabditis elegans, and Arabidopsis thaliana. Furthermore, we consider how sex-specific changes in the meiotic chromosome axes and the epigenetic landscape may function together to regulate crossing over in each sex, indicating that the mechanisms controlling crossing over may be different in oogenesis and spermatogenesis.


Asunto(s)
Profase Meiótica I/fisiología , Caracteres Sexuales , Desarrollo Sexual , Animales , Proteínas de Ciclo Celular/metabolismo , Intercambio Genético , Roturas del ADN de Doble Cadena , Femenino , Recombinación Homóloga , Humanos , Masculino , Proteínas Nucleares/metabolismo , Oogénesis , Unión Proteica , Desarrollo Sexual/genética , Espermatogénesis
15.
Nat Commun ; 10(1): 1251, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30890697

RESUMEN

Male gametes are generated through a specialised differentiation pathway involving a series of developmental transitions that are poorly characterised at the molecular level. Here, we use droplet-based single-cell RNA-Sequencing to profile spermatogenesis in adult animals and at multiple stages during juvenile development. By exploiting the first wave of spermatogenesis, we both precisely stage germ cell development and enrich for rare somatic cell-types and spermatogonia. To capture the full complexity of spermatogenesis including cells that have low transcriptional activity, we apply a statistical tool that identifies previously uncharacterised populations of leptotene and zygotene spermatocytes. Focusing on post-meiotic events, we characterise the temporal dynamics of X chromosome re-activation and profile the associated chromatin state using CUT&RUN. This identifies a set of genes strongly repressed by H3K9me3 in spermatocytes, which then undergo extensive chromatin remodelling post-meiosis, thus acquiring an active chromatin state and spermatid-specific expression.


Asunto(s)
Histonas/metabolismo , Espermatocitos/crecimiento & desarrollo , Espermatogénesis/fisiología , Transcripción Genética/fisiología , Cromosoma X/metabolismo , Animales , Separación Celular/métodos , Cromatina/metabolismo , Mapeo Cromosómico/métodos , Cromosomas Humanos Par 21/genética , Epigénesis Genética/fisiología , Femenino , Citometría de Flujo/métodos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/fisiología , Histonas/genética , Humanos , Masculino , Profase Meiótica I/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Espermatocitos/metabolismo , Testículo/citología
16.
PLoS Genet ; 15(3): e1008004, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30921322

RESUMEN

Germ cell immortality, or transgenerational maintenance of the germ line, could be promoted by mechanisms that could occur in either mitotic or meiotic germ cells. Here we report for the first time that the GSP-2 PP1/Glc7 phosphatase promotes germ cell immortality. Small RNA-induced genome silencing is known to promote germ cell immortality, and we identified a separation-of-function allele of C. elegans gsp-2 that is compromised for germ cell immortality and is also defective for small RNA-induced genome silencing and meiotic but not mitotic chromosome segregation. Previous work has shown that GSP-2 is recruited to meiotic chromosomes by LAB-1, which also promoted germ cell immortality. At the generation of sterility, gsp-2 and lab-1 mutant adults displayed germline degeneration, univalents, histone methylation and histone phosphorylation defects in oocytes, phenotypes that mirror those observed in sterile small RNA-mediated genome silencing mutants. Our data suggest that a meiosis-specific function of GSP-2 ties small RNA-mediated silencing of the epigenome to germ cell immortality. We also show that transgenerational epigenomic silencing at hemizygous genetic elements requires the GSP-2 phosphatase, suggesting a functional link to small RNAs. Given that LAB-1 localizes to the interface between homologous chromosomes during pachytene, we hypothesize that small localized discontinuities at this interface could promote genomic silencing in a manner that depends on small RNAs and the GSP-2 phosphatase.


Asunto(s)
Células Germinativas/metabolismo , Proteína Fosfatasa 1/fisiología , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica , Genoma , Células Germinativas/fisiología , Meiosis/fisiología , Profase Meiótica I/fisiología , Metilación , Monoéster Fosfórico Hidrolasas , Proteína Fosfatasa 1/metabolismo , Interferencia de ARN/fisiología , ARN Interferente Pequeño
17.
Sci Adv ; 5(1): eaau9780, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30746471

RESUMEN

Segregation of homologous chromosomes in meiosis I is tightly regulated by their physical links, or crossovers (COs), generated from DNA double-strand breaks (DSBs) through meiotic homologous recombination. In budding yeast, three ZMM (Zip1/2/3/4, Mer3, Msh4/5) proteins, Zip2, Zip4, and Spo16, form a "ZZS" complex, functioning to promote meiotic recombination via a DSB repair pathway. Here, we identified the mammalian ortholog of Spo16, termed SPO16, which interacts with the mammalian ortholog of Zip2 (SHOC1/MZIP2), and whose functions are evolutionarily conserved to promote the formation of COs. SPO16 localizes to the recombination nodules, as SHOC1 and TEX11 do. SPO16 is required for stabilization of SHOC1 and proper localization of other ZMM proteins. The DSBs formed in SPO16-deleted meiocytes were repaired without COs formation, although synapsis is less affected. Therefore, formation of SPO16-SHOC1 complex-associated recombination intermediates is a key step facilitating meiotic recombination that produces COs from yeast to mammals.


Asunto(s)
Intercambio Genético , Proteínas de Unión al ADN/metabolismo , Profase Meiótica I/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Proteínas de Transporte de Catión/metabolismo , Segregación Cromosómica/fisiología , Cromosomas de los Mamíferos/genética , Roturas del ADN de Doble Cadena , Femenino , Células HeLa , Humanos , Masculino , Ratones
18.
Cell Death Differ ; 26(11): 2194-2207, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30760872

RESUMEN

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.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/metabolismo , Proteínas Nucleares/genética , Espermatocitos/metabolismo , Espermatogénesis/genética , Factores de Transcripción/genética , Acetilación , Animales , Apoptosis/genética , Ensamble y Desensamble de Cromatina/fisiología , Histonas/metabolismo , Masculino , Profase Meiótica I/fisiología , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo , Fase Paquiteno/fisiología , Proteínas del Grupo Polycomb/metabolismo , Recuento de Espermatozoides , Testículo/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
19.
Nat Struct Mol Biol ; 26(3): 175-184, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30778237

RESUMEN

Germ cells manifest a unique gene expression program and regain totipotency in the zygote. Here, we perform Hi-C analysis to examine 3D chromatin organization in male germ cells during spermatogenesis. We show that the highly compartmentalized 3D chromatin organization characteristic of interphase nuclei is attenuated in meiotic prophase. Meiotic prophase is predominated by short-range intrachromosomal interactions that represent a condensed form akin to that of mitotic chromosomes. However, unlike mitotic chromosomes, meiotic chromosomes display weak genomic compartmentalization, weak topologically associating domains, and localized point interactions in prophase. In postmeiotic round spermatids, genomic compartmentalization increases and gives rise to the strong compartmentalization seen in mature sperm. The X chromosome lacks domain organization during meiotic sex-chromosome inactivation. We propose that male meiosis occurs amid global reprogramming of 3D chromatin organization and that strengthening of chromatin compartmentalization takes place in spermiogenesis to prepare the next generation of life.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Meiosis/fisiología , Espermátides/crecimiento & desarrollo , Espermatocitos/crecimiento & desarrollo , Espermatogénesis/fisiología , Animales , Cromatina/metabolismo , Cromosomas/metabolismo , Interfase/fisiología , Masculino , Profase Meiótica I/fisiología , Ratones , Ratones Endogámicos C57BL , Dominios Proteicos/fisiología
20.
Reprod Sci ; 26(11): 1519-1537, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29587616

RESUMEN

Mechanisms of meiotic prophase I arrest maintenance (germinal vesicle [GV] stage) and meiotic resumption (germinal vesicle breakdown [GVBD] stage) in mammalian oocytes seem to be very complicated. These processes are regulated via multiple molecular cascades at transcriptional, translational, and posttranslational levels, and many of them are interrelated. There are many molecular cascades of meiosis maintaining and meiotic resumption in oocyte which are orchestrated by multiple molecules produced by pituitary gland and follicular cells. Furthermore, many of these molecular cascades are duplicated, thus ensuring the stability of the entire system. Understanding mechanisms of oocyte maturation is essential to assess the oocyte status, develop effective protocols of oocyte in vitro maturation, and design novel contraceptive drugs. Mechanisms of meiotic arrest maintenance at prophase I and meiotic resumption in mammalian oocytes are covered in the present article.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Meiosis/fisiología , Profase Meiótica I/fisiología , Oocitos/metabolismo , Oogénesis/fisiología , Animales , Femenino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA