Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Annu Rev Genomics Hum Genet ; 24: 35-61, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37159901

RESUMEN

In meiosis, homologous chromosome synapsis is mediated by a supramolecular protein structure, the synaptonemal complex (SC), that assembles between homologous chromosome axes. The mammalian SC comprises at least eight largely coiled-coil proteins that interact and self-assemble to generate a long, zipper-like structure that holds homologous chromosomes in close proximity and promotes the formation of genetic crossovers and accurate meiotic chromosome segregation. In recent years, numerous mutations in human SC genes have been associated with different types of male and female infertility. Here, we integrate structural information on the human SC with mouse and human genetics to describe the molecular mechanisms by which SC mutations can result in human infertility. We outline certain themes in which different SC proteins are susceptible to different types of disease mutation and how genetic variants with seemingly minor effects on SC proteins may act as dominant-negative mutations in which the heterozygous state is pathogenic.


Asunto(s)
Infertilidad , Complejo Sinaptonémico , Masculino , Femenino , Humanos , Ratones , Animales , Complejo Sinaptonémico/genética , Emparejamiento Cromosómico , Meiosis/genética , Infertilidad/genética , Mutación , Mamíferos/genética
2.
Nat Struct Mol Biol ; 30(2): 188-199, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36635604

RESUMEN

In meiosis, a supramolecular protein structure, the synaptonemal complex (SC), assembles between homologous chromosomes to facilitate their recombination. Mammalian SC formation is thought to involve hierarchical zipper-like assembly of an SYCP1 protein lattice that recruits stabilizing central element (CE) proteins as it extends. Here we combine biochemical approaches with separation-of-function mutagenesis in mice to show that, rather than stabilizing the SYCP1 lattice, the CE protein SYCE3 actively remodels this structure during synapsis. We find that SYCP1 tetramers undergo conformational change into 2:1 heterotrimers on SYCE3 binding, removing their assembly interfaces and disrupting the SYCP1 lattice. SYCE3 then establishes a new lattice by its self-assembly mimicking the role of the disrupted interface in tethering together SYCP1 dimers. SYCE3 also interacts with CE complexes SYCE1-SIX6OS1 and SYCE2-TEX12, providing a mechanism for their recruitment. Thus, SYCE3 remodels the SYCP1 lattice into a CE-binding integrated SYCP1-SYCE3 lattice to achieve long-range synapsis by a mature SC.


Asunto(s)
Emparejamiento Cromosómico , Complejo Sinaptonémico , Animales , Ratones , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/genética , Mamíferos/genética , Meiosis , Proteínas Nucleares/metabolismo , Complejo Sinaptonémico/metabolismo
3.
Genes (Basel) ; 13(10)2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36292683

RESUMEN

Anophthalmia (missing eye) describes a failure of early embryonic ocular development. Mutations in a relatively small set of genes account for 75% of bilateral anophthalmia cases, yet 25% of families currently are left without a molecular diagnosis. Here, we report our experimental work that aimed to uncover the developmental and genetic basis of the anophthalmia characterising the X-linked Ie (eye-ear reduction) X-ray-induced allele in mouse that was first identified in 1947. Histological analysis of the embryonic phenotype showed failure of normal eye development after the optic vesicle stage with particularly severe malformation of the ventral retina. Linkage analysis mapped this mutation to a ~6 Mb region on the X chromosome. Short- and long-read whole-genome sequencing (WGS) of affected and unaffected male littermates confirmed the Ie linkage but identified no plausible causative variants or structural rearrangements. These analyses did reduce the critical candidate interval and revealed evidence of multiple variants within the ancestral DNA, although none were found that altered coding sequences or that were unique to Ie. To investigate early embryonic events at a genetic level, we then generated mouse ES cells derived from male Ie embryos and wild type littermates. RNA-seq and accessible chromatin sequencing (ATAC-seq) data generated from cultured optic vesicle organoids did not reveal any large differences in gene expression or accessibility of putative cis-regulatory elements between Ie and wild type. However, an unbiased TF-footprinting analysis of accessible chromatin regions did provide evidence of a genome-wide reduction in binding of transcription factors associated with ventral eye development in Ie, and evidence of an increase in binding of the Zic-family of transcription factors, including Zic3, which is located within the Ie-refined critical interval. We conclude that the refined Ie critical region at chrX: 56,145,000-58,385,000 contains multiple genetic variants that may be linked to altered cis regulation but does not contain a convincing causative mutation. Changes in the binding of key transcription factors to chromatin causing altered gene expression during development, possibly through a subtle mis-regulation of Zic3, presents a plausible cause for the anophthalmia phenotype observed in Ie, but further work is required to determine the precise causative allele and its genetic mechanism.


Asunto(s)
Anoftalmos , Ratones , Masculino , Animales , Anoftalmos/genética , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cromatina , ADN , Proteínas de Homeodominio/genética
4.
Diagn Microbiol Infect Dis ; 104(1): 115732, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35728458

RESUMEN

Throughout the COVID-19 pandemic nasopharyngeal or nose and/or throat swabs (NTS) have been the primary approach for collecting patient samples for the subsequent detection of viral RNA. However, this procedure, if undertaken correctly, can be unpleasant and therefore deters individuals from providing high quality samples. To overcome these limitations other modes of sample collection have been explored. In a cohort of frontline health care workers we have compared saliva and gargle samples to gold-standard NTS. 93% of individuals preferred providing saliva or gargle samples, with little sex-dependent variation. Viral titers collected in samples were analyzed using standard methods and showed that gargle and saliva were similarly comparable for identifying COVID-19 positive individuals compared to NTS (92% sensitivity; 98% specificity). We suggest that gargle and saliva collection are viable alternatives to NTS swabs and may encourage testing to provide better disease diagnosis and population surveillance.


Asunto(s)
COVID-19 , COVID-19/diagnóstico , Prueba de COVID-19 , Humanos , Antisépticos Bucales , Nasofaringe , Pandemias , ARN Viral/genética , SARS-CoV-2 , Saliva , Manejo de Especímenes/métodos
5.
Nat Commun ; 13(1): 2791, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589755

RESUMEN

Tumour cell plasticity is a major barrier to the efficacy of targeted cancer therapies but the mechanisms that mediate it are poorly understood. Here, we identify dysregulated RNA splicing as a key driver of tumour cell dedifferentiation in colorectal cancer (CRC). We find that Apc-deficient CRC cells have dysregulated RNA splicing machinery and exhibit global rewiring of RNA splicing. We show that the splicing factor SRSF1 controls the plasticity of tumour cells by controlling Kras splicing and is required for CRC invasion in a mouse model of carcinogenesis. SRSF1 expression maintains stemness in human CRC organoids and correlates with cancer stem cell marker expression in human tumours. Crucially, partial genetic downregulation of Srsf1 does not detrimentally affect normal tissue homeostasis, demonstrating that tumour cell plasticity can be differentially targeted. Thus, our findings link dysregulation of the RNA splicing machinery and control of tumour cell plasticity.


Asunto(s)
Plasticidad de la Célula , Neoplasias Colorrectales , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Plasticidad de la Célula/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Ratones , Empalme del ARN/genética , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
6.
Elife ; 102021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34338635

RESUMEN

Shuttling RNA-binding proteins coordinate nuclear and cytoplasmic steps of gene expression. The SR family proteins regulate RNA splicing in the nucleus and a subset of them, including SRSF1, shuttles between the nucleus and cytoplasm affecting post-splicing processes. However, the physiological significance of this remains unclear. Here, we used genome editing to knock-in a nuclear retention signal (NRS) in Srsf1 to create a mouse model harboring an SRSF1 protein that is retained exclusively in the nucleus. Srsf1NRS/NRS mutants displayed small body size, hydrocephalus, and immotile sperm, all traits associated with ciliary defects. We observed reduced translation of a subset of mRNAs and decreased abundance of proteins involved in multiciliogenesis, with disruption of ciliary ultrastructure and motility in cells and tissues derived from this mouse model. These results demonstrate that SRSF1 shuttling is used to reprogram gene expression networks in the context of high cellular demands, as observed here, during motile ciliogenesis.


Asunto(s)
Cilios/metabolismo , Citoplasma/metabolismo , Factores de Empalme Serina-Arginina/genética , Animales , Núcleo Celular/metabolismo , Masculino , Ratones , Factores de Empalme Serina-Arginina/metabolismo
8.
Dev Cell ; 56(1): 22-35.e7, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33278343

RESUMEN

Retrotransposon proliferation poses a threat to germline integrity. While retrotransposons must be activated in developing germ cells in order to survive and propagate, how they are selectively activated in the context of meiosis is unclear. We demonstrate that the transcriptional activation of Ty3/Gypsy retrotransposons and host defense are controlled by master meiotic regulators. We show that budding yeast Ty3/Gypsy co-opts binding sites of the essential meiotic transcription factor Ndt80 upstream of the integration site, thereby tightly linking its transcriptional activation to meiotic progression. We also elucidate how yeast cells thwart Ty3/Gypsy proliferation by blocking translation of the retrotransposon mRNA using amyloid-like assemblies of the RNA-binding protein Rim4. In mammals, several inactive Ty3/Gypsy elements are undergoing domestication. We show that mammals utilize equivalent master meiotic regulators (Stra8, Mybl1, Dazl) to regulate Ty3/Gypsy-derived genes in developing gametes. Our findings inform how genes that are evolving from retrotransposons can build upon existing regulatory networks during domestication.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Germinativas/metabolismo , Meiosis/genética , Proteínas de Unión al ARN/metabolismo , ADN Polimerasa Dirigida por ARN/metabolismo , Retroelementos/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Sitios de Unión , Secuenciación de Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Evolución Molecular , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Meiosis/fisiología , Ratones , Zarigüeyas/genética , Zarigüeyas/metabolismo , Biosíntesis de Proteínas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Unión al ARN/genética , ADN Polimerasa Dirigida por ARN/genética , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética
9.
PLoS Biol ; 18(12): e3001030, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33320856

RESUMEN

With the ongoing COVID-19 (Coronavirus Disease 2019) pandemic, caused by the novel coronavirus SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), there is a need for sensitive, specific, and affordable diagnostic tests to identify infected individuals, not all of whom are symptomatic. The most sensitive test involves the detection of viral RNA using RT-qPCR (quantitative reverse transcription PCR), with many commercial kits now available for this purpose. However, these are expensive, and supply of such kits in sufficient numbers cannot always be guaranteed. We therefore developed a multiplex assay using well-established SARS-CoV-2 targets alongside a human cellular control (RPP30) and a viral spike-in control (Phocine Herpes Virus 1 [PhHV-1]), which monitor sample quality and nucleic acid extraction efficiency, respectively. Here, we establish that this test performs as well as widely used commercial assays, but at substantially reduced cost. Furthermore, we demonstrate >1,000-fold variability in material routinely collected by combined nose and throat swabbing and establish a statistically significant correlation between the detected level of human and SARS-CoV-2 nucleic acids. The inclusion of the human control probe in our assay therefore provides a quantitative measure of sample quality that could help reduce false-negative rates. We demonstrate the feasibility of establishing a robust RT-qPCR assay at approximately 10% of the cost of equivalent commercial assays, which could benefit low-resource environments and make high-volume testing affordable.


Asunto(s)
Prueba de COVID-19/métodos , COVID-19/diagnóstico , ARN Viral/análisis , SARS-CoV-2/aislamiento & purificación , Prueba de COVID-19/economía , Humanos , Reacción en Cadena de la Polimerasa Multiplex/economía , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/economía , SARS-CoV-2/genética
10.
Nucleic Acids Res ; 48(14): 7748-7766, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32585002

RESUMEN

Mouse embryonic stem cells (mESCs) cultured with MEK/ERK and GSK3ß (2i) inhibitors transition to ground state pluripotency. Gene expression changes, redistribution of histone H3K27me3 profiles and global DNA hypomethylation are hallmarks of 2i exposure, but it is unclear whether epigenetic alterations are required to achieve and maintain ground state or occur as an outcome of 2i signal induced changes. Here we show that ESCs with three epitypes, WT, constitutively methylated, or hypomethylated, all undergo comparable morphological, protein expression and transcriptome changes independently of global alterations of DNA methylation levels or changes in H3K27me3 profiles. Dazl and Fkbp6 expression are induced by 2i in all three epitypes, despite exhibiting hypermethylated promoters in constitutively methylated ESCs. We identify a number of activated gene promoters that undergo 2i dependent loss of H3K27me3 in all three epitypes, however genetic and pharmaceutical inhibition experiments show that H3K27me3 is not required for their silencing in non-2i conditions. By separating and defining their contributions, our data suggest that repressive epigenetic systems play minor roles in mESC self-renewal and naïve ground state establishment by core sets of dominant pluripotency associated transcription factor networks, which operate independently from these epigenetic processes.


Asunto(s)
Represión Epigenética , Redes Reguladoras de Genes , Células Madre Embrionarias de Ratones/metabolismo , Animales , Células Cultivadas , Metilación de ADN , Epigénesis Genética , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Histonas/metabolismo , Masculino , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/enzimología , Factores de Transcripción/metabolismo , Transcripción Genética
11.
J Cell Biol ; 219(5)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32232464

RESUMEN

Age-dependent oocyte aneuploidy, a major cause of Down syndrome, is associated with declining sister chromatid cohesion in postnatal oocytes. Here we show that cohesion in postnatal mouse oocytes is regulated by Tex19.1. We show Tex19.1-/- oocytes have defects maintaining chiasmata, missegregate their chromosomes during meiosis, and transmit aneuploidies to the next generation. Furthermore, we show that mouse Tex19.1 inhibits N-end rule protein degradation mediated by its interacting partner UBR2, and that Ubr2 itself has a previously undescribed role in negatively regulating the acetylated SMC3 subpopulation of cohesin in mitotic somatic cells. Lastly, we show that acetylated SMC3 is associated with meiotic chromosome axes in mouse oocytes, and that this population of cohesin is specifically depleted in the absence of Tex19.1. These findings indicate that Tex19.1 regulates UBR protein activity to maintain acetylated SMC3 and sister chromatid cohesion in postnatal oocytes and prevent aneuploidy from arising in the female germline.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ARN/genética , Intercambio de Cromátides Hermanas/genética , Ubiquitina-Proteína Ligasas/genética , Aneuploidia , Animales , Linaje de la Célula/genética , Cromátides/genética , Segregación Cromosómica/genética , Femenino , Células Germinativas/crecimiento & desarrollo , Humanos , Meiosis/genética , Ratones , Ratones Noqueados , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Cohesinas
12.
Cell Rep ; 29(7): 1974-1985.e6, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31722211

RESUMEN

The DNA hypomethylation that occurs when embryonic stem cells (ESCs) are directed to the ground state of naive pluripotency by culturing in two small molecule inhibitors (2i) results in redistribution of polycomb (H3K27me3) away from its target loci. Here, we demonstrate that 3D genome organization is also altered in 2i, with chromatin decompaction at polycomb target loci and a loss of long-range polycomb interactions. By preventing DNA hypomethylation during the transition to the ground state, we are able to restore to ESC in 2i the H3K27me3 distribution, as well as polycomb-mediated 3D genome organization that is characteristic of primed ESCs grown in serum. However, these cells retain the functional characteristics of 2i ground-state ESCs. Our findings demonstrate the central role of DNA methylation in shaping major aspects of 3D genome organization but caution against assuming causal roles for the epigenome and 3D genome in gene regulation and function in ESCs.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Metilación de ADN , Epigenoma , Células Madre Embrionarias de Ratones/metabolismo , Animales , Cromatina/genética , Masculino , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología
13.
FASEB J ; 33(12): 14221-14233, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31659914

RESUMEN

Deleted in azoospermia-like (DAZL) is a germ cell RNA-binding protein that is essential for entry and progression through meiosis. The phenotype of the Dazl knockout mouse has extensive germ cell loss because of incomplete meiosis. We have created a Dazl hypomorph model using short interfering RNA knockdown in mouse fetal ovary cultures, allowing investigation of Dazl function in germ cell maturation. Dazl hypomorph ovaries had a phenotype of impaired germ cell nest breakdown with a 66% reduction in total follicle number and an increase in the proportion of primordial follicles (PMFs), with smaller oocytes within these follicles. There was no significant early germ cell loss or meiotic delay. Immunostaining of intercellular bridge component testis-expressed protein (Tex)14 showed ∼59% reduction in foci number and size, without any change in Tex14 mRNA levels. TEX14 expression was also confirmed in the human fetal ovary across gestation. Using 3'UTR-luciferase reporter assays, translational regulation of TEX14 was demonstrated to be DAZL-dependant. Dazl is therefore essential for normal intercellular bridges within germ cell nests and their timely breakdown, with a major impact on subsequent assembly of PMFs.-Rosario, R., Crichton, J. H., Stewart, H. L., Childs, A. J., Adams, I. R., Anderson, R. A. Dazl determines primordial follicle formation through the translational regulation of Tex14.


Asunto(s)
Ovario/crecimiento & desarrollo , Ovario/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Animales , Clonación Molecular , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Meiosis/fisiología , Ratones , Interferencia de ARN , ARN Mensajero , Proteínas de Unión al ARN/genética , Técnicas de Cultivo de Tejidos , Factores de Transcripción/genética
14.
Biochem J ; 476(15): 2141-2156, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31383821

RESUMEN

The spatial configuration of chromatin is fundamental to ensure any given cell can fulfil its functional duties, from gene expression to specialised cellular division. Significant technological innovations have facilitated further insights into the structure, function and regulation of three-dimensional chromatin organisation. To date, the vast majority of investigations into chromatin organisation have been conducted in interphase and mitotic cells leaving meiotic chromatin relatively unexplored. In combination, cytological and genome-wide contact frequency analyses in mammalian germ cells have recently demonstrated that large-scale chromatin structures in meiotic prophase I are reminiscent of the sequential loop arrays found in mitotic cells, although interphase-like segmentation of transcriptionally active and inactive regions are also evident along the length of chromosomes. Here, we discuss the similarities and differences of such large-scale chromatin architecture, between interphase, mitotic and meiotic cells, as well as their functional relevance and the proposed modulatory mechanisms which underlie them.


Asunto(s)
Cromatina/metabolismo , Células Germinativas/metabolismo , Interfase/fisiología , Meiosis/fisiología , Mitosis/fisiología , Animales , Células Germinativas/citología , Humanos
15.
EMBO J ; 38(9)2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30988016

RESUMEN

The rate of RNA polymerase II (RNAPII) elongation has an important role in the control of alternative splicing (AS); however, the in vivo consequences of an altered elongation rate are unknown. Here, we generated mouse embryonic stem cells (ESCs) knocked in for a slow elongating form of RNAPII We show that a reduced transcriptional elongation rate results in early embryonic lethality in mice. Focusing on neuronal differentiation as a model, we observed that slow elongation impairs development of the neural lineage from ESCs, which is accompanied by changes in AS and in gene expression along this pathway. In particular, we found a crucial role for RNAPII elongation rate in transcription and splicing of long neuronal genes involved in synapse signaling. The impact of the kinetic coupling of RNAPII elongation rate with AS is greater in ESC-differentiated neurons than in pluripotent cells. Our results demonstrate the requirement for an appropriate transcriptional elongation rate to ensure proper gene expression and to regulate AS during development.


Asunto(s)
Empalme Alternativo , Células Madre Embrionarias/patología , Regulación de la Expresión Génica , Células-Madre Neurales/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Transcripción Genética , Animales , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Células-Madre Neurales/patología
16.
Mol Cell ; 74(5): 1069-1085.e11, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31000436

RESUMEN

Orderly segregation of chromosomes during meiosis requires that crossovers form between homologous chromosomes by recombination. Programmed DNA double-strand breaks (DSBs) initiate meiotic recombination. We identify ANKRD31 as a key component of complexes of DSB-promoting proteins that assemble on meiotic chromosome axes. Genome-wide, ANKRD31 deficiency causes delayed recombination initiation. In addition, loss of ANKRD31 alters DSB distribution because of reduced selectivity for sites that normally attract DSBs. Strikingly, ANKRD31 deficiency also abolishes uniquely high rates of recombination that normally characterize pseudoautosomal regions (PARs) of X and Y chromosomes. Consequently, sex chromosomes do not form crossovers, leading to chromosome segregation failure in ANKRD31-deficient spermatocytes. These defects co-occur with a genome-wide delay in assembling DSB-promoting proteins on autosome axes and loss of a specialized PAR-axis domain that is highly enriched for DSB-promoting proteins in wild type. Thus, we propose a model for spatiotemporal patterning of recombination by ANKRD31-dependent control of axis-associated DSB-promoting proteins.


Asunto(s)
Proteínas Portadoras/genética , Roturas del ADN de Doble Cadena , Recombinación Homóloga/genética , Meiosis/genética , Animales , Proteínas Portadoras/química , Segregación Cromosómica/genética , Masculino , Ratones , Regiones Pseudoautosómicas/genética , Espermatocitos/crecimiento & desarrollo , Espermatocitos/metabolismo , Cromosoma X/genética , Cromosoma Y/genética
17.
Elife ; 82019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30674417

RESUMEN

Male germ cells of all placental mammals express an ancient nuclear RNA binding protein of unknown function called RBMXL2. Here we find that deletion of the retrogene encoding RBMXL2 blocks spermatogenesis. Transcriptome analyses of age-matched deletion mice show that RBMXL2 controls splicing patterns during meiosis. In particular, RBMXL2 represses the selection of aberrant splice sites and the insertion of cryptic and premature terminal exons. Our data suggest a Rbmxl2 retrogene has been conserved across mammals as part of a splicing control mechanism that is fundamentally important to germ cell biology. We propose that this mechanism is essential to meiosis because it buffers the high ambient concentrations of splicing activators, thereby preventing poisoning of key transcripts and disruption to gene expression by aberrant splice site selection.


Asunto(s)
Células Germinativas/metabolismo , Sitios de Empalme de ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Exones/genética , Fertilidad , Regulación del Desarrollo de la Expresión Génica , Masculino , Meiosis/genética , Metafase/genética , Ratones Endogámicos C57BL , Modelos Animales , Especificidad de Órganos , Empalme del ARN/genética , Testículo/metabolismo
18.
Chromosoma ; 127(4): 437-459, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29907896

RESUMEN

Recombination, synapsis, chromosome segregation and gene expression are co-ordinately regulated during meiosis to ensure successful execution of this specialised cell division. Studies with multiple mutant mouse lines have shown that mouse spermatocytes possess quality control checkpoints that eliminate cells with persistent defects in chromosome synapsis. In addition, studies on Trip13mod/mod mice suggest that pachytene spermatocytes that successfully complete chromosome synapsis can undergo meiotic arrest in response to defects in recombination. Here, we present additional support for a meiotic recombination-dependent checkpoint using a different mutant mouse line, Tex19.1-/-. The appearance of early recombination foci is delayed in Tex19.1-/- spermatocytes during leptotene/zygotene, but some Tex19.1-/- spermatocytes still successfully synapse their chromosomes and we show that these spermatocytes are enriched for early recombination foci. Furthermore, we show that patterns of axis elongation, chromatin modifications and histone H1t expression are also all co-ordinately skewed towards earlier substages of pachytene in these autosomally synapsed Tex19.1-/- spermatocytes. We also show that this skew towards earlier pachytene substages occurs in the absence of elevated spermatocyte death in the population, that spermatocytes with features of early pachytene are present in late stage Tex19.1-/- testis tubules and that the delay in histone H1t expression in response to loss of Tex19.1 does not occur in a Spo11 mutant background. Taken together, these data suggest that a recombination-dependent checkpoint may be able to modulate pachytene progression in mouse spermatocytes to accommodate some types of recombination defect.


Asunto(s)
Proteínas Nucleares/genética , Fase Paquiteno/genética , Recombinación Genética , Espermatocitos/citología , ATPasas Asociadas con Actividades Celulares Diversas/genética , Animales , Proteínas de Ciclo Celular/genética , Cromatina/genética , Cromatina/metabolismo , Emparejamiento Cromosómico/genética , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino , Meiosis , Ratones Endogámicos C57BL , Ratones Noqueados , Procesamiento Proteico-Postraduccional/genética , Proteínas de Unión al ARN , Túbulos Seminíferos/metabolismo , Túbulos Seminíferos/patología , Espermatocitos/fisiología
19.
Elife ; 62017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28806172

RESUMEN

Mobilization of retrotransposons to new genomic locations is a significant driver of mammalian genome evolution, but these mutagenic events can also cause genetic disorders. In humans, retrotransposon mobilization is mediated primarily by proteins encoded by LINE-1 (L1) retrotransposons, which mobilize in pluripotent cells early in development. Here we show that TEX19.1, which is induced by developmentally programmed DNA hypomethylation, can directly interact with the L1-encoded protein L1-ORF1p, stimulate its polyubiquitylation and degradation, and restrict L1 mobilization. We also show that TEX19.1 likely acts, at least in part, through promoting the activity of the E3 ubiquitin ligase UBR2 towards L1-ORF1p. Moreover, loss of Tex19.1 increases L1-ORF1p levels and L1 mobilization in pluripotent mouse embryonic stem cells, implying that Tex19.1 prevents de novo retrotransposition in the pluripotent phase of the germline cycle. These data show that post-translational regulation of L1 retrotransposons plays a key role in maintaining trans-generational genome stability in mammals.


Asunto(s)
Elementos de Nucleótido Esparcido Largo , Células Madre Embrionarias de Ratones/fisiología , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Recombinación Genética , Animales , Técnicas de Inactivación de Genes , Ratones , Proteínas Nucleares/genética , Unión Proteica , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
20.
PLoS Genet ; 13(7): e1006904, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28708824

RESUMEN

Meiosis relies on the SPO11 endonuclease to generate the recombinogenic DNA double strand breaks (DSBs) required for homologous chromosome synapsis and segregation. The number of meiotic DSBs needs to be sufficient to allow chromosomes to search for and find their homologs, but not excessive to the point of causing genome instability. Here we report that the mammal-specific gene Tex19.1 promotes Spo11-dependent recombination in mouse spermatocytes. We show that the chromosome asynapsis previously reported in Tex19.1-/- spermatocytes is preceded by reduced numbers of recombination foci in leptotene and zygotene. Tex19.1 is required for normal levels of early Spo11-dependent recombination foci during leptotene, but not for upstream events such as MEI4 foci formation or accumulation of H3K4me3 at recombination hotspots. Furthermore, we show that mice carrying mutations in Ubr2, which encodes an E3 ubiquitin ligase that interacts with TEX19.1, phenocopy the Tex19.1-/- recombination defects. These data suggest that Tex19.1 and Ubr2 are required for mouse spermatocytes to accumulate sufficient Spo11-dependent recombination to ensure that the homology search is consistently successful, and reveal a hitherto unknown genetic pathway promoting meiotic recombination in mammals.


Asunto(s)
Endodesoxirribonucleasas/metabolismo , Meiosis/genética , Proteínas Nucleares/metabolismo , Espermatocitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Emparejamiento Cromosómico , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , Endodesoxirribonucleasas/genética , Masculino , Profase Meiótica I/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Proteínas de Unión al ARN , Recombinación Genética , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...