Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
DNA Res ; 29(3)2022 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-35652718

RESUMEN

Recent advances in single-cell analysis technology have made it possible to analyse tens of thousands of cells at a time. In addition, sample multiplexing techniques, which allow the analysis of several types of samples in a single run, are very useful for reducing experimental costs and improving experimental accuracy. However, a problem with this technique is that antigens and antibodies for universal labelling of various cell types may not be fully available. To overcome this issue, we developed a universal labelling technique, Universal Surface Biotinylation (USB), which does not depend on specific cell surface proteins. By introducing biotin into the amine group of any cell surface protein, we have obtained good labelling results in all the cell types we have tested. Combining with DNA-tagged streptavidin, it is possible to label each cell sample with specific DNA 'hashtag'. Compared with the conventional cell hashing method, the USB procedure seemed to have no discernible adverse effect on the acquisition of the transcriptome in each cell, according to the model experiments using differentiating mouse embryonic stem cells. This method can be theoretically used for any type of cells, including cells to which the conventional cell hashing method has not been applied successfully.


Asunto(s)
Biotina , Animales , Biotinilación , Análisis Costo-Beneficio , Ratones , Análisis de Secuencia de ARN , Estreptavidina
2.
Biomaterials ; 284: 121491, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35395453

RESUMEN

The loss of nucleus pulposus (NP) precedes the intervertebral disk (IVD) degeneration that causes back pain. Here, we demonstrate that the implantation of human iPS cell-derived cartilaginous tissue (hiPS-Cart) restores this loss by replacing lost NP spatially and functionally. NP cells consist of notochordal NP cells and chondrocyte-like NP cells. Single cell RNA sequencing (scRNA-seq) analysis revealed that cells in hiPS-Cart corresponded to chondrocyte-like NP cells but not to notochordal NP cells. The implantation of hiPS-Cart into a nuclectomized space of IVD in nude rats prevented the degeneration of the IVD and preserved its mechanical properties. hiPS-Cart survived and occupied the nuclectomized space for at least six months after implantation, indicating spatial and functional replacement of lost NP by hiPS-Cart. Further scRNA-seq analysis revealed that hiPS-Cart cells changed their profile after implantation, differentiating into two lineages that are metabolically distinct from each other. However, post-implanted hiPS-Cart cells corresponded to chondrocyte-like NP cells only and did not develop into notochordal NP cells, suggesting that chondrocyte-like NP cells are nearly sufficient for NP function. The data collectively indicate that hiPS-Cart is a candidate implant for regenerating NP spatially and functionally and preventing IVD degeneration.


Asunto(s)
Células Madre Pluripotentes Inducidas , Degeneración del Disco Intervertebral , Disco Intervertebral , Núcleo Pulposo , Animales , Cartílago , Humanos , Degeneración del Disco Intervertebral/terapia , Ratas , Regeneración
3.
Genes Cells ; 27(1): 14-24, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34808017

RESUMEN

LincRNA-p21 is a long intergenic non-coding RNA (LincRNA) gene reported to activate the transcription of the adjacent Cdkn1a (p21) gene in cis. The importance of the enhancer elements in the LincRNA-p21 gene region has also been reported; however, the involvement of the LincRNA-p21 transcripts in regulating Cdkn1a in vivo is still unclear. In this study, we used a LincRNA-p21-trapped mouse line (LincRNA-p21Gt ) in which ßgeo was inserted into intron 1, and all enhancer elements were retained. In LincRNA-p21Gt/Gt mice, the transcription of LincRNA-p21 was repressed due to the ßgeo sequence, and the expression of exon 1 of LincRNA-p21 was restored through its deletion or replacement by another sequence, and Cdkn1a expression was also upregulated. Furthermore, regardless of the full-length transcripts, the expression of Cdkn1a correlated with the transcription of the exon 1 of LincRNA-p21. This result indicates that the LincRNA-p21 transcripts are not functional, but the transcriptional activity around exon 1 is important for Cdkn1a expression.


Asunto(s)
ARN Largo no Codificante , Animales , Proliferación Celular , Exones , Ratones , ARN Largo no Codificante/genética
4.
Cell Rep ; 31(8): 107686, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32460033

RESUMEN

Meiotic recombination is critical for genetic exchange and generation of chiasmata that ensures faithful chromosome segregation during meiosis I. Meiotic recombination is initiated by DNA double-strand break (DSB) followed by multiple processes of DNA repair. The exact mechanisms for how recombinases localize to DSB remain elusive. Here, we show that C19orf57/4930432K21Rik/BRME1 is a player for meiotic recombination in mice. C19orf57/4930432K21Rik/BRME1 associates with single-stranded DNA (ssDNA) binding proteins, BRCA2 and MEILB2/HSF2BP, which are critical recruiters of recombinases onto DSB sites. Disruption of C19orf57/4930432K21Rik/BRME1 shows severe impact on DSB repair and male fertility. Remarkably, removal of ssDNA binding proteins from DSB sites is delayed, and reciprocally, the loading of RAD51 and DMC1 onto resected ssDNA is impaired in Brme1 knockout (KO) spermatocytes. We propose that C19orf57/4930432K21Rik/BRME1 modulates localization of recombinases to meiotic DSB sites through the interaction with the BRCA2-MEILB2/HSF2BP complex during meiotic recombination.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Recombinación Homóloga/genética , Meiosis/genética , Proteínas de Unión a Fosfato/metabolismo , Recombinasa Rad51/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/genética , Animales , Roturas del ADN de Doble Cadena , Masculino , Ratones
5.
Dev Cell ; 52(4): 429-445.e10, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32032549

RESUMEN

The mechanisms regulating meiotic initiation in mammals are enigmatic. It is known that retinoic acid (RA) signaling plays a pivotal role during meiotic initiation. STRA8, which is expressed in response to RA, is thought to be a key factor promoting meiotic initiation. However, the specific role of STRA8 in meiotic initiation has remained elusive. Here, we identified MEIOSIN as a germ-cell-specific factor that associates with STRA8. MEIOSIN, like STRA8, is expressed in response to RA and plays an essential role in meiotic initiation in both males and females. Functional analyses revealed that MEIOSIN acts as a transcription factor together with STRA8, and that both factors are critical for driving meiotic gene activation. Furthermore, temporally restricted expression of MEIOSIN leads to meiotic entry decision during spermatogenesis. The present study demonstrates that MEIOSIN, in collaboration with STRA8, plays a central role in regulating the mitosis to meiosis germ cell fate decision in mammals.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Ciclo Celular , Regulación de la Expresión Génica , Células Germinativas/fisiología , Meiosis , Mitosis , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Femenino , Células Germinativas/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Espermatogénesis
6.
Endocrinology ; 160(11): 2673-2691, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31436800

RESUMEN

Isolated growth hormone deficiency type II (IGHD2) is mainly caused by heterozygous splice-site mutations in intron 3 of the GH1 gene. A dominant-negative effect of the mutant GH lacking exon 3 on wild-type GH secretion has been proposed; however, the molecular mechanisms involved are elusive. To uncover the molecular systems underlying GH deficiency in IGHD2, we established IGHD2 model mice, which carry both wild-type and mutant copies of the human GH1 gene, replacing each of the endogenous mouse Gh loci. Our IGHD2 model mice exhibited growth retardation along with intact cellular architecture and mildly activated endoplasmic reticulum stress in the pituitary gland, caused by decreased GH-releasing hormone receptor (Ghrhr) and Gh gene promoter activities. Decreased Ghrhr and Gh promoter activities were likely caused by reduced levels of nuclear CREB3L2, which was demonstrated to stimulate Ghrhr and Gh promoter activity. To our knowledge, this is the first in vivo study to reveal a novel molecular mechanism of GH deficiency in IGHD2, representing a new paradigm that differs from widely accepted models.


Asunto(s)
Enanismo Hipofisario/etiología , Hormona del Crecimiento/metabolismo , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Enanismo Hipofisario/patología , Femenino , Hormona del Crecimiento/genética , Humanos , Masculino , Ratones , Hipófisis/metabolismo , Hipófisis/ultraestructura , Regiones Promotoras Genéticas , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética
7.
Stem Cell Reports ; 12(5): 1113-1128, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31056481

RESUMEN

CpG islands (CGIs) including those at imprinting control regions (ICRs) are protected from de novo methylation in somatic cells. However, many cancers often exhibit CGI hypermethylation, implying that the machinery is impaired in cancer cells. Here, we conducted a comprehensive analysis of CGI methylation during somatic cell reprogramming. Although most CGIs remain hypomethylated, a small subset of CGIs, particularly at several ICRs, was often de novo methylated in reprogrammed pluripotent stem cells (PSCs). Such de novo ICR methylation was linked with the silencing of reprogramming factors, which occurs at a late stage of reprogramming. The ICR-preferred CGI hypermethylation was similarly observed in human PSCs. Mechanistically, ablation of Dnmt3a prevented PSCs from de novo ICR methylation. Notably, the ICR-preferred CGI hypermethylation was observed in pediatric cancers, while adult cancers exhibit genome-wide CGI hypermethylation. These results may have important implications in the pathogenesis of pediatric cancers and the application of PSCs.


Asunto(s)
Reprogramación Celular/genética , Metilación de ADN/genética , Impresión Genómica/genética , Células Madre Pluripotentes/metabolismo , Adulto , Animales , Células Cultivadas , Islas de CpG/genética , Epigénesis Genética/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos ICR , Células Madre Pluripotentes/citología
8.
J Hum Genet ; 63(11): 1119-1128, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30115950

RESUMEN

Intracranial vertebral-basilar artery dissection (IVAD) is an arterial disorder leading to life-threatening consequences. Genetic factors are known to be causative to certain syndromic forms of IVAD. However, systematic study of the molecular basis of sporadic and isolated IVAD is lacking. To identify genetic variants contributing to the etiology of IVAD, we enrolled a cohort of 44 unrelated cases with a clinical diagnosis of isolated IVAD and performed whole-exome sequencing (WES) for all the participants; a trio exome sequencing approach was used when samples from both parents were available. Four previously reported disease-causing heterozygous variants (three in COL3A1 and one in FBN1) and seven novel heterozygous variants in IVAD-related genes were identified. In addition, six variants in novel IVAD genes including two de novo heterozygous nonsynonymous variants (each in VPS52 and CDK18), two stop-gain variants (each in MYH9 and LYL1), and two heterozygous biallelic variants in TNXB were considered to be possibly contributing to the phenotype, with unknown significance according to the existing knowledge. A significantly higher mutational rate of IVAD candidate genes was observed in patients versus our in-house controls (P = 0.002) (DISCO study, http://www.discostudy.org/ , n = 2248). Our study provided a mutational landscape for patients with isolated IVAD.


Asunto(s)
Disección Aórtica/genética , Exoma , Secuenciación de Nucleótidos de Alto Rendimiento , Aneurisma Intracraneal/genética , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Estudios de Cohortes , Colágeno Tipo III/genética , Femenino , Fibrilina-1/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas Motoras Moleculares/genética , Cadenas Pesadas de Miosina/genética , Proteínas de Neoplasias/genética
9.
Curr Protoc Stem Cell Biol ; 46(1): e60, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30005143

RESUMEN

Epiblast stem cells (EpiSCs) are primed pluripotent stem cells (PSCs) derived from mouse postimplantation embryos. Interestingly, EpiSCs share many characteristics with human PSCs such as human embryonic stem cells (hESCs) and human induced PSCs (hiPSC). Thus, EpiSCs can serve as a model for studying primed states of pluripotency. This article describes a simple yet highly efficient protocol for EpiSC derivation and maintenance of homogenous EpiSCs using an inhibitor of WNT secretion. Using this method, EpiSCs can be readily derived from mouse strains with different genetic background including C57BL/6N. The EpiSCs derived by this protocol maintain a homogenous, undifferentiated status, yet retain high differentiation potential. Unlike EpiSCs established by the original protocol, the new EpiSC lines require the continued presence of WNT inhibitor, suggesting intrinsic differences from EpiSCs made by the original method. This new version of EpiSCs will provide clues to understand the nature of primed states of mammalian pluripotent cells and may facilitate establishment of a better protocol for directed differentiation from the primed state. © 2018 by John Wiley & Sons, Inc.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/citología , Proteínas Wnt/metabolismo , Animales , Separación Celular , Células Cultivadas , Criopreservación , Disección , Embrión de Mamíferos/citología , Fibroblastos/citología , Estratos Germinativos/citología , Ratones Endogámicos C57BL , Células Madre Pluripotentes/metabolismo
10.
Sci Rep ; 7: 40056, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28059165

RESUMEN

Ovo, which encodes a transcription factor with Zn-finger domains, is evolutionarily conserved among animals. In Drosophila, in addition to its zygotic function for egg production, maternal ovo activity is required in primordial germ cells (PGCs) for expression of germline genes such as vasa and nanos. In this study, we found that maternal Ovo accumulates in PGC nuclei during embryogenesis. In these cells, ovo serves a dual function: activation of genes expressed predominantly in PGCs, and conversely suppression of somatic genes. Reduction of ovo activity in PGCs makes them unable to develop normally into germ cells of both sexes. In mice, knockout of the ovo ortholog, Ovol2, which is expressed in PGCs, decreases the number of PGCs during early embryogenesis. These data strongly suggest that ovo acts as part of an evolutionarily conserved mechanism that regulates germline development in animals.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Desarrollo Embrionario , Células Germinativas/fisiología , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Drosophila , Proteínas de Drosophila/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética
11.
Biol Reprod ; 94(6): 122, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27122635

RESUMEN

Mouse trophoblast stem cells (TSCs) proliferate indefinitely in vitro, despite their highly heterogeneous nature. In this study, we sought to characterize TSC colony types by using methods based on cell biology and biochemistry for a better understanding of how TSCs are maintained over multiple passages. Colonies of TSCs could be classified into four major types: type 1 is compact and dome-shaped, type 4 is flattened but with a large multilayered cell cluster, and types 2 and 3 are their intermediates. A time-lapse analysis indicated that type 1 colonies predominantly appeared after passaging, and a single type 1 colony gave rise to all other types. These colony transitions were irreversible, but at least some type 1 colonies persisted throughout culture. The typical cells comprising type 1 colonies were small and highly motile, and they aggregated together to form primary colonies. A hierarchical clustering based on global gene expression profiles suggested that a TSC line containing more type 1 colony cells was similar to in vivo extraembryonic tissues. Among the known TSC genes examined, Elf5 showed a differential expression pattern according to colony type, indicating that this gene might be a reliable marker of undifferentiated TSCs. When aggregated with fertilized embryos, cells from types 1 and 2, but not from type 4, distributed to the polar trophectoderm in blastocysts. These findings indicate that cells typically found in type 1 colonies can persist indefinitely as stem cells and are responsible for the maintenance of TSC lines. They may provide key information for future improvements in the quality of TSC lines.


Asunto(s)
Células Madre/citología , Trofoblastos/citología , Animales , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Trofoblastos/metabolismo
12.
Stem Cell Reports ; 4(4): 744-57, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25818811

RESUMEN

Epiblast stem cells (EpiSCs) are pluripotent stem cells derived from epiblasts of postimplantation mouse embryos, and thus provide a useful model for studying "primed" pluripotent states. Here, we devised a simple and robust technique to derive high-quality EpiSCs using an inhibitor of WNT secretion. Using this method, we readily established EpiSC lines with high efficiency and were able to use whole embryonic portions without having to separate the epiblast from the visceral endoderm (VE). Expression analyses revealed that these EpiSCs maintained a homogeneous, undifferentiated status, yet showed high potential for differentiation both in vitro and in teratomas. Unlike EpiSCs derived by the original protocol, new EpiSC lines required continuous treatment with the Wnt inhibitor, suggesting some intrinsic differences from the existing EpiSCs. The homogeneous properties of this new version of EpiSCs should facilitate studies on the establishment and maintenance of a "primed" pluripotent state, and directed differentiation from the primed state.


Asunto(s)
Estratos Germinativos/citología , Células Madre/citología , Células Madre/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Línea Celular , Autorrenovación de las Células/efectos de los fármacos , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Células Madre/efectos de los fármacos
14.
Genes Genet Syst ; 89(3): 109-20, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25475934

RESUMEN

The proximal third of mouse chromosome 17 is known as the t-complex. The t-haplotype is a variant form of this region containing four tandem inversions compared with the wild-type t-complex, and thus recombination in heterozygotes of the t-haplotype is strongly suppressed along the entire t-complex region. Within this genetically locked t-haplotype, many mutations related to various interesting phenotypes (e.g., taillessness, transmission ratio distortion, recessive lethality) have accumulated, and many mouse geneticists have been attracted to t-haplotype research. Many recessive lethal mutations known as t-complex lethal mutations have been found, and detailed phenotypic analyses have revealed that the functions of t-lethal genes are related to important developmental events. Therefore, identification of the genes responsible for these lethal mutations may contribute to our understanding of the mechanisms of mammalian development. In this review, I introduce the phenotypes of t-lethal mutations and describe recent findings, including our results regarding the molecular identification of a t-lethal gene.


Asunto(s)
Genes Letales , Ratones/genética , Región del Complejo T del Genoma , Animales , Cromosomas de los Mamíferos , Desarrollo Embrionario , Ratones/embriología , Mutación , Fenotipo
15.
DNA Res ; 20(6): 549-65, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23861320

RESUMEN

To understand the epigenetic regulation required for germ cell-specific gene expression in the mouse, we analysed DNA methylation profiles of developing germ cells using a microarray-based assay adapted for a small number of cells. The analysis revealed differentially methylated sites between cell types tested. Here, we focused on a group of genomic sequences hypomethylated specifically in germline cells as candidate regions involved in the epigenetic regulation of germline gene expression. These hypomethylated sequences tend to be clustered, forming large (10 kb to ~9 Mb) genomic domains, particularly on the X chromosome of male germ cells. Most of these regions, designated here as large hypomethylated domains (LoDs), correspond to segmentally duplicated regions that contain gene families showing germ cell- or testis-specific expression, including cancer testis antigen genes. We found an inverse correlation between DNA methylation level and expression of genes in these domains. Most LoDs appear to be enriched with H3 lysine 9 dimethylation, usually regarded as a repressive histone modification, although some LoD genes can be expressed in male germ cells. It thus appears that such a unique epigenomic state associated with the LoDs may constitute a basis for the specific expression of genes contained in these genomic domains.


Asunto(s)
Metilación de ADN , Células Germinativas/metabolismo , Cromosoma X/metabolismo , Animales , Carcinogénesis/genética , Línea Celular , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Femenino , Duplicación de Gen , Perfilación de la Expresión Génica , Histonas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Duplicaciones Segmentarias en el Genoma , Testículo/metabolismo , Cromosoma X/genética
16.
J Reprod Dev ; 59(3): 231-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23363561

RESUMEN

In mice, one of the major epigenetic errors associated with somatic cell nuclear transfer (SCNT) is ectopic expression of Xist during the preimplantation period in both sexes. We found that this aberrant Xist expression could be impeded by deletion of Xist from the putative active X chromosome in donor cells. In male clones, it was also found that prior injection of Xist-specific siRNA could significantly improve the postimplantation development of cloned embryos as a result of a significant repression of Xist at the morula stage. In this study, we examined whether the same knockdown strategy could work as well in female SCNT-derived embryos. Embryos were reconstructed with cumulus cell nuclei and injected with Xist-specific siRNA at 6-7 h after oocyte activation. RNA FISH analysis revealed that siRNA treatment successfully repressed Xist RNA at the morula stage, as shown by the significant decrease in the number of cloud-type Xist signals in the blastomere nuclei. However, blastomeres with different sizes (from "pinpoint" to "cloud") and numbers of Xist RNA signals remained within single embryos. After implantation, the dysregulated Xist expression was normalized autonomously, as in male clones, to a state of monoallelic expression in both embryonic and extraembryonic tissues. However, at term there was no significant improvement in the survival of the siRNA-injected cloned embryos. Thus, siRNA injection was largely effective in repressing the Xist overexpression in female cloned embryos but failed to rescue them, probably because of an inability to mimic consistent monoallelic Xist expression in these embryos. This could only be achieved in female embryos by applying a gene knockout strategy rather than an siRNA approach.


Asunto(s)
Clonación de Organismos/métodos , Interferencia de ARN , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Alelos , Animales , Núcleo Celular/metabolismo , Células del Cúmulo/metabolismo , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ácidos Hidroxámicos/química , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Mórula/metabolismo , Técnicas de Transferencia Nuclear , Oocitos/citología , ARN Interferente Pequeño/metabolismo
17.
Cell Rep ; 2(5): 1363-74, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23142660

RESUMEN

After implantation, pluripotent epiblasts are converted to embryonic ectoderm through cell-cell interactions that significantly change the transcriptional and epigenetic networks. An entrée to understanding this vital developmental transition is the t(w5) mutation of the mouse t complex. This mutation produces highly specific defects in the embryonic ectoderm before gastrulation, leading to death of the embryonic ectoderm. Using a positional cloning approach, we have now identified the mutated gene, completing a decades-long search. The gene, vacuolar protein sorting 52 (Vps52), is a mouse homolog of yeast VPS52 that is involved in the retrograde trafficking of endosomes. Our data suggest that Vps52 acts in extraembryonic tissues to support the growth and differentiation of embryonic ectoderm via cell-cell interactions. It is also required in the formation of embryonic structures at a later stage of development, revealing hitherto unknown functions of Vps52 in the development of a multicellular organism.


Asunto(s)
Células Madre Embrionarias/citología , Proteínas/metabolismo , Región del Complejo T del Genoma , Animales , Secuencia de Bases , Comunicación Celular , Diferenciación Celular , Células Cultivadas , Ectodermo/metabolismo , Ectodermo/patología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Desarrollo Embrionario , Células Madre Embrionarias/metabolismo , Exones , Mutación del Sistema de Lectura , Haplotipos , Complejo Mayor de Histocompatibilidad/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitosis , Datos de Secuencia Molecular , Proteínas/genética , Proteínas de Transporte Vesicular
18.
Proc Natl Acad Sci U S A ; 108(51): 20621-6, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22065773

RESUMEN

Cloning mammals by somatic cell nuclear transfer (SCNT) is highly inefficient. Most SCNT-generated embryos die after implantation because of unidentified, complex epigenetic errors in the process of postimplantation embryonic development. Here we identify the most upstream level of dysfunction leading to impaired development of clones by using RNAi against Xist, a gene responsible for X chromosome inactivation (XCI). A prior injection of Xist-specific siRNA into reconstructed oocytes efficiently corrected SCNT-specific aberrant Xist expression at the morula stage, but failed to do so thereafter at the blastocyst stage. However, we found that shortly after implantation, this aberrant XCI status in cloned embryos had been corrected autonomously in both embryonic and extraembryonic tissues, probably through a newly established XCI control for postimplantation embryos. Embryo transfer experiments revealed that siRNA-treated embryos showed 10 times higher survival than controls as early as embryonic day 5.5 and this high survival persisted until term, resulting in a remarkable improvement in cloning efficiency (12% vs. 1% in controls). Importantly, unlike control clones, these Xist-siRNA clones at birth showed only a limited dysregulation of their gene expression, indicating that correction of Xist expression in preimplantation embryos had a long-term effect on their postnatal normality. Thus, contrary to the general assumption, our results suggest that the fate of cloned embryos is determined almost exclusively before implantation by their XCI status. Furthermore, our strategy provides a promising breakthrough for mammalian SCNT cloning, because RNAi treatment of oocytes is readily applicable to most mammal species.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Interferencia de ARN , ARN no Traducido/metabolismo , Animales , Blastocisto/citología , Clonación Molecular , Desarrollo Embrionario , Ácidos Hidroxámicos/farmacología , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Mórula/metabolismo , Oocitos/citología , ARN/metabolismo , ARN Largo no Codificante , ARN Interferente Pequeño/metabolismo , Factores de Tiempo
19.
Science ; 330(6003): 496-9, 2010 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-20847234

RESUMEN

Cloning mammals by means of somatic cell nuclear transfer (SCNT) is highly inefficient because of erroneous reprogramming of the donor genome. Reprogramming errors appear to arise randomly, but the nature of nonrandom, SCNT-specific errors remains elusive. We found that Xist, a noncoding RNA that inactivates one of the two X chromosomes in females, was ectopically expressed from the active X (Xa) chromosome in cloned mouse embryos of both sexes. Deletion of Xist on Xa showed normal global gene expression and resulted in about an eight- to ninefold increase in cloning efficiency. We also identified an Xist-independent mechanism that specifically down-regulated a subset of X-linked genes through somatic-type repressive histone blocks. Thus, we have identified nonrandom reprogramming errors in mouse cloning that can be altered to improve the efficiency of SCNT methods.


Asunto(s)
Clonación de Organismos/métodos , Técnicas de Transferencia Nuclear , ARN no Traducido/fisiología , Cromosoma X , Animales , Regulación hacia Abajo , Embrión de Mamíferos/metabolismo , Femenino , Eliminación de Gen , Perfilación de la Expresión Génica , Masculino , Ratones , ARN Largo no Codificante , ARN no Traducido/biosíntesis , ARN no Traducido/genética
20.
PLoS Genet ; 5(12): e1000756, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19997484

RESUMEN

In mammals, observations of rapid shifts in mitochondrial DNA (mtDNA) variants between generations have led to the creation of the bottleneck theory for the transmission of mtDNA. The bottleneck could be attributed to a marked decline of mtDNA content in germ cells giving rise to the next generation, to a small effective number of mtDNA segregation units resulting from homoplasmic nucleoids rather than the single mtDNA molecule serving as the units of segregation, or to the selective transmission of a subgroup of the mtDNA population to the progeny. We have previously determined mtDNA copy number in single germ cells and shown that the bottleneck occurs without the reduction in germline mtDNA content. Recently one study suggested that the bottleneck is driven by a remarkable decline of mtDNA copies in early primordial germ cells (PGCs), while another study reported that the mtDNA genetic bottleneck results from replication of a subpopulation of the mtDNA genome during postnatal oocyte maturation and not during embryonic oogenesis, despite a detected a reduction in mtDNA content in early PGCs. To clarify these contradictory results, we examined the mtDNA copy number in PGCs isolated from transgenic mice expressing fluorescent proteins specifically in PGCs as in the aforementioned two other studies. We provide clear evidence to confirm that no remarkable reduction in mtDNA content occurs in PGCs and reinforce that the bottleneck is generated without reduction of mtDNA content in germ cells.


Asunto(s)
ADN Mitocondrial/metabolismo , Células Germinativas/metabolismo , Mitocondrias/genética , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/metabolismo , Separación Celular , Proteínas Cromosómicas no Histona , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/citología , Células Germinativas/enzimología , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados , Coloración y Etiquetado , Factores de Transcripción/metabolismo , Proteína Fluorescente Roja
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...