Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell Rep Methods ; 3(9): 100590, 2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37714158

RESUMEN

Non-human primates (NHPs) are the closest animal model to humans; thus, gene engineering technology in these species holds great promise for the elucidation of higher brain functions and human disease models. Knockin (KI) gene targeting is a versatile approach to modify gene(s) of interest; however, it generally suffers from the low efficiency of homology-directed repair (HDR) in mammalian cells, especially in non-expressed gene loci. In the current study, we generated a tyrosine hydroxylase (TH)-2A-Cre KI model of the common marmoset monkey (marmoset; Callithrix jacchus) using an HDR-biased CRISPR-Cas9 genome editing approach using Cas9-DN1S and RAD51. This model should enable labeling and modification of a specific neuronal lineage using the Cre-loxP system. Collectively, the current study paves the way for versatile gene engineering in NHPs, which may be a significant step toward further biomedical and preclinical applications.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Animales , Sistemas CRISPR-Cas/genética , Tirosina 3-Monooxigenasa/genética , Primates/genética , Mamíferos/genética
2.
Biology (Basel) ; 11(7)2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-36101367

RESUMEN

Recent success in derivation of functional gametes (oocytes and spermatozoa) from pluripotent stem cells (PSCs) of rodents has made it feasible for future application to large animals including endangered species and to ultimately humans. Here, we summarize backgrounds and recent studies on in vitro gametogenesis from rodent PSCs, and similar approaches using PSCs from large animals, including livestock, nonhuman primates (NHPs), and humans. We also describe additional developing approaches for in vitro reconstitution of reproductive organs, such as the ovary (ovarioid), testis (testisoid), and future challenges in the uterus (uteroid) and oviduct (oviductoid), all of which may be derived from PSCs. Once established, these in vitro systems may serve as a robust platform for elucidating the pathology of infertility-related disorders and ectopic pregnancy, principle of reproduction, and artificial biogenesis. Therefore, these possibilities, especially when using human cells, require consideration of ethical issues, and international agreements and guidelines need to be raised before opening "Pandora's Box".

3.
Dev Growth Differ ; 64(6): 325-341, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35841539

RESUMEN

Potentials of immortal proliferation and unlimited differentiation into all the three germ layers and germ cells in induced pluripotent stem cells (iPSCs) render them important bioresources for in vitro reconstitution and modeling of intravital tissues and organs in various animal models, thus contributing to the elucidation of pathomechanisms, drug discovery and stem cell-based regenerative medicine. We previously reported promising approaches for deriving transgene-free iPSCs from somatic fibroblasts of multiple mammalian species by episomal vector or RNA transfection, although the respective step-by-step protocols and the combinatorial usage of these methods, which achieved high induction efficiency, have not been described in the literature so far. Here, we provide a detailed step-by-step description of these methods with critical tips and slight modifications (improvements) to previously reported methods. We also report a novel method for the establishment of iPSCs from the Syrian hamster (also known as golden hamster; Mesocricetus auratus), a unique animal model of hibernation. We anticipate this methodology will contribute to stem cell biology and regenerative medicine research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular , Reprogramación Celular , Fibroblastos , Mamíferos , Transgenes
4.
Dev Growth Differ ; 63(6): 277-284, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34133763

RESUMEN

Urodele amphibian newts have unique biological properties in male gametogenesis, in addition to their extreme regenerative capacity. Male newts are able to regenerate new testes even after reaching sexual maturity and can possess multiple testes. Notably, these animals maintain primordial germ cell-like cells in a tissue adjacent to the testis. Spermatogenesis proceeds while synchronizing in a region-specific manner in the testis. However, the newt species that have been used most commonly require 2-3 years to achieve sexual maturity, and spermatogenesis in these species shows seasonality. These traits have restricted the use of newts for studies on testicular development and spermatogenesis, and testis development in newts remains poorly characterized. Recently, the Iberian ribbed newt Pleurodeles waltl has been established as an emerging model organism. P. waltl reaches sexual maturity more quick after birth than do other newts and is capable of breeding year-round. Thus, P. waltl is expected to serve as an appealing experimental model for studying the mechanisms of male gametogenesis in the urodeles. In the present study, we use P. waltl to describe the entire developmental process of the newt testis from primordial gonad to maturity. Notably, the mature testes show synchronized progression of spermatogenesis along the anteroposterior axis. Additionally, we demonstrate that the process of spermatogenesis in P. waltl proceeds irrespective of day length. Our results show that P. waltl newts are a suitable model for investigating the process of testicular development. We also expect that these results will be useful for the maintenance of P. waltl bioresources.


Asunto(s)
Fotoperiodo , Pleurodeles , Animales , Células Germinativas , Masculino , Salamandridae , Testículo
5.
Stem Cell Reports ; 16(4): 754-770, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33798453

RESUMEN

Induced pluripotent stem cells (iPSCs) are capable of providing an unlimited source of cells from all three germ layers and germ cells. The derivation and usage of iPSCs from various animal models may facilitate stem cell-based therapy, gene-modified animal production, and evolutionary studies assessing interspecies differences. However, there is a lack of species-wide methods for deriving iPSCs, in particular by means of non-viral and non-transgene-integrating (NTI) approaches. Here, we demonstrate the iPSC derivation from somatic fibroblasts of multiple mammalian species from three different taxonomic orders, including the common marmoset (Callithrix jacchus) in Primates, the dog (Canis lupus familiaris) in Carnivora, and the pig (Sus scrofa) in Cetartiodactyla, by combinatorial usage of chemical compounds and NTI episomal vectors. Interestingly, the fibroblasts temporarily acquired a neural stem cell-like state during the reprogramming. Collectively, our method, robustly applicable to various species, holds a great potential for facilitating stem cell-based research using various animals in Mammalia.


Asunto(s)
Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Mamíferos/metabolismo , Transgenes , Animales , Callithrix , Perros , Perfilación de la Expresión Génica , Vectores Genéticos/metabolismo , Estratos Germinativos/metabolismo , Células-Madre Neurales/metabolismo , Plásmidos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , RNA-Seq , Especificidad de la Especie , Porcinos , Virus
6.
Stem Cell Res ; 53: 102308, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33799281

RESUMEN

POU class 5 homeobox 1 (POU5F1, also known as OCT4) is critical for maintenance of pluripotency, germ cell fate, reprogramming into a pluripotent state, and early embryogenesis. We generated an embryonic stem cell (ESC) line of the common marmoset (Callithrix jacchus) harboring a heterozygous knock-in allele of OCT4-P2A-mCerulean-T2A-pac. The ESC line (CMES40-OC) will be valuable for investigation of primed/naïve pluripotency and germ cell fate. Homozygous OCT4 knock-in clones were generated but could not be sustained in an undifferentiated state in long-term culture. The OCT4 knock-in system facilitated simultaneous knock-in of a reporter construct at another locus, DDX4 (VASA).


Asunto(s)
Callithrix , Genes Homeobox , Alelos , Animales , Diferenciación Celular , Línea Celular , Células Madre Embrionarias , Factor 3 de Transcripción de Unión a Octámeros/genética
7.
Stem Cell Res ; 44: 101740, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32151954

RESUMEN

BLIMP1 (PRDM1) and VASA (DDX4) play pivotal roles in the development of the germ cell linage. Importantly, these genes are specifically expressed in germ cells; BLIMP1 in primordial germ cells (PGCs) to early-stage gonocytes, and VASA in migration-stage PGCs to mature gametes. The high reproductive efficiency of common marmosets (marmosets; Callithrix jacchus) makes them advantageous for use in germ cell research. We herein report the generation of a male marmoset embryonic stem cell (ESC) line harboring BLIMP1 and DDX4 double reporters. This ESC line will be a useful tool for investigating male gametogenesis in non-human primates.


Asunto(s)
Callithrix , Línea Celular , Células Madre Embrionarias , Transposasas , Animales , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Células Germinativas , Masculino
8.
PLoS One ; 12(6): e0179585, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28658256

RESUMEN

A search for early response genes that are activated following germ cell induction from mouse embryonic stem cells in vitro led us to the isolation of a long noncoding RNA that contains a SINE (short interspersed element)-B1F motif that was named R53. In situ hybridization and northern blot analyses revealed that the R53 subfragment RNA bears a B1F motif, is processed from the primary transcript, is expressed in adult testis and is predominantly localized in meiotic metaphase chromatin during spermatogenesis. Recent studies of chromosome-associated RNAs have explored novel functions of noncoding RNAs. Specifically, chromosome-bound noncoding RNAs function not only as structural components of chromosome but also as scaffolds that recruit epigenetic modulators for transcriptional regulation, and they are dynamically rearranged during the cell cycle. However, few studies have explored meiotic chromatin; thus, R53 RNA appears to be the first long noncoding RNA to be tightly associated with the metaphase chromatin during spermatogenesis. Furthermore, R53 knockdown using a lentivirus-mediated RNAi injected into mouse testis and organ culture of the fragments revealed a remarkable reduction in postmeiotic cells and irregular up-regulation of several postmeiotic genes, which suggests the possibility that the SINE-B1-derived noncoding RNA R53 plays an indispensable role in the transcriptional regulation of key spermatogenesis genes.


Asunto(s)
Cromatina/metabolismo , Metafase/fisiología , ARN no Traducido/metabolismo , Elementos de Nucleótido Esparcido Corto/genética , Espermatogénesis/fisiología , Animales , Regulación de la Expresión Génica , Masculino , Ratones , Interferencia de ARN , ARN no Traducido/genética , Testículo/metabolismo
9.
Sci Rep ; 6: 32932, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27608931

RESUMEN

Epigenetic modifications play crucial roles on establishment of tissue-specific transcription profiles and cellular characteristics. Direct conversions of fibroblasts into differentiated tissue cells by over-expression of critical transcription factors have been reported, but the epigenetic mechanisms underlying these conversions are still not fully understood. In addition, conversion of somatic cells into germ cells has not yet been achieved. To understand epigenetic mechanisms that underlie germ cell characteristics, we attempted to use defined epigenetic factors to directly convert mouse embryonic fibroblasts (MEFs) into germ cells. Here, we successfully induced germ cell-specific genes by inhibiting repressive epigenetic modifications via RNAi or small-molecule compounds. Under these conditions, some tissue-specific genes and stimulus-inducible genes were also induced. Meanwhile, the treatments did not result in genome-wide transcriptional activation. These results suggested that a permissive epigenetic environment resulted in selective de-repression of stimulus- and differentiation-inducible genes including germ cell-specific genes in MEFs.


Asunto(s)
Transdiferenciación Celular , Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/fisiología , Animales , Células Cultivadas , Ratones , Factores de Transcripción/metabolismo , Activación Transcripcional
10.
PLoS One ; 11(9): e0163466, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27649575

RESUMEN

Jmjd1C is one of the Jmjd1 family genes that encode putative demethylases against histone H3K9 and non-histone proteins and has been proven to play an indispensable role in mouse spermatogenesis. Here, we analyzed a newly-bred transgenic mouse strain carrying a Jmjd1C loss-of-function allele in which a ß-geo cassette was integrated into the intron of the Jmjd1C locus. Jmjd1C gene-trap homozygous testes exhibited malformations in postmeiotic processes and a deficiency in the long-term maintenance of undifferentiated spermatogonia. Some groups of spermatids in the homozygous testis showed abnormal organization and incomplete elongation from the first wave of spermatogenesis onwards. Moreover, histone H4K16 acetylation, which is required for the onset of chromatin remodeling, appeared to be remarkably decreased. These effects may not have been a result of the drastic decrease in gene expression related to the events but instead may have been due to the lack of interaction between JMJD1C and its partner proteins, such as MDC1 and HSP90. Additionally, significant decreases in Oct4 expression and NANOG- and OCT4-expressing spermatogonia were found in the Jmjd1C homozygous mature testis, suggesting that JMJD1C may participate in the maintenance of spermatogonial stem cell self-renewal by up-regulating Oct4 expression. These results indicate that JMJD1C has multiple functions during spermatogenesis through interactions with different partners during the spermatogenic stages.


Asunto(s)
Epigénesis Genética , Histona Demetilasas con Dominio de Jumonji/genética , Espermatogénesis/genética , Animales , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Espermatogonias/metabolismo , Testículo/metabolismo
11.
Mol Reprod Dev ; 82(10): 774-86, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26096940

RESUMEN

Development of female gonads in the chicken is asymmetric. This asymmetry affects gene expression, morphology, and germ cell development; consequently only the left ovary develops into a functional organ, whereas the right ovary remains vestigial. In males, on the other hand, both gonads develop into functional testes. Here, we revisited the development of asymmetric traits in female (and male) chicken gonads between Hamburger Hamilton stage 16 (HH16) and hatching. At HH16, primordial germ cells migrated preferentially to the left gonad, accumulating in the left coelomic hinge between the gut mesentery and developing gonad in both males and females. Using the meiotic markers SYCP3 and phosphorylated H2AFX, we identified a previously undescribed, pronounced asymmetryc meiotic progression in the germ cells located in the central, lateral, and extreme cortical regions of the left female gonad from HH38 until hatching. Moreover, we observed that--in contrast to the current view--medullary germ cells are not apoptotic, but remain arrested in pre-leptotene until hatching. In addition to the systematic analysis of the asymmetric distribution of germ cells in female chicken gonads, we propose an updated model suggesting that the localization of germ cells--in the left or right gonad; in the cortex or medulla of the left gonad; and in the central part or the extremities of the left cortex--has direct consequences for their development and participation in adult reproduction.


Asunto(s)
Tipificación del Cuerpo , Células Germinales Embrionarias/citología , Ovario/embriología , Animales , Movimiento Celular , Pollos , Desarrollo Embrionario , Femenino , Masculino , Meiosis , Profase Meiótica I , Caracteres Sexuales , Testículo/embriología
12.
Proc Natl Acad Sci U S A ; 111(3): 1120-5, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24398516

RESUMEN

For normal fertilization in mammals, it is important that functionally mature sperm are motile and have a fully formed acrosome. The glycosyltransferase-like gene, human polypeptide N-acetylgalactosaminyltransferase-like protein 5 (GALNTL5), belongs to the polypeptide N-acetylgalactosamine-transferase (pp-GalNAc-T) gene family because of its conserved glycosyltransferase domains, but it uniquely truncates the C-terminal domain and is expressed exclusively in human testis. However, glycosyltransferase activity of the human GALNTL5 protein has not been identified by in vitro assay thus far. Using mouse Galntl5 ortholog, we have examined whether GALNTL5 is a functional molecule in spermatogenesis. It was observed that mouse GALNTL5 localizes in the cytoplasm of round spermatids in the region around the acrosome of elongating spermatids, and finally in the neck region of spermatozoa. We attempted to establish Galntl5-deficient mutant mice to investigate the role of Galntl5 in spermiogenesis and found that the heterozygous mutation affected male fertility due to immotile sperm, which is diagnosed as asthenozoospermia, an infertility syndrome in humans. Furthermore, the heterozygous mutation of Galntl5 attenuated glycolytic enzymes required for motility, disrupted protein loading into acrosomes, and caused aberrant localization of the ubiquitin-proteasome system. By comparing the protein compositions of sperm from infertile males, we found a deletion mutation of the exon of human GALNTL5 gene in a patient with asthenozoospermia. This strongly suggests that the genetic mutation of human GALNTL5 results in male infertility with the reduction of sperm motility and that GALNTL5 is a functional molecule essential for mammalian sperm formation.


Asunto(s)
Infertilidad Masculina/genética , Mutación , N-Acetilgalactosaminiltransferasas/genética , Motilidad Espermática , Espermatozoides/metabolismo , Acrosoma/metabolismo , Animales , Astenozoospermia/metabolismo , Citoplasma/metabolismo , Heterocigoto , Humanos , Lectinas/metabolismo , Masculino , Ratones , N-Acetilgalactosaminiltransferasas/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Espermátides/metabolismo , Espermatogénesis , Testículo/metabolismo , Ubiquitina/química , Polipéptido N-Acetilgalactosaminiltransferasa
13.
Nat Commun ; 4: 1754, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23612295

RESUMEN

Embryonic stem cells and primordial germ cells (PGCs) express many pluripotency-associated genes, but embryonic stem cells do not normally undergo conversion into primordial germ cells. Thus, we predicted that there is a mechanism that represses primordial germ cell-related gene expression in embryonic stem cells. Here we identify genes involved in this putative mechanism, by using an embryonic stem cell line with a Vasa reporter in an RNA interference screen of transcription factor genes expressed in embryonic stem cells. We identify five genes that result in the expression of Vasa when silenced. Of these, Max is the most striking. Transcriptome analysis reveals that Max knockdown in embryonic stem cells results in selective, global derepression of germ cell-specific genes. Max interacts with histone H3K9 methyltransferases and associates with the germ cell-specific genes in embryonic stem cells. In addition, Max knockdown results in a decrease in histone H3K9 dimethylation at their promoter regions. We propose that Max is part of protein complex that acts as a repressor of germ cell-related genes in embryonic stem cells.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Biomarcadores/metabolismo , Línea Celular , ARN Helicasas DEAD-box/genética , Células Madre Embrionarias/citología , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Genes Reporteros , Células Germinativas/citología , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Meiosis/genética , Metilación , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Proteínas Represoras , Espermatogénesis/genética , Factores de Tiempo , Regulación hacia Arriba/genética
14.
Biol Open ; 1(11): 1146-52, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23213395

RESUMEN

During gastrulation, chicken primordial germ cells (PGCs) are present in an extraembryonic region of the embryo from where they migrate towards the genital ridges. This is also observed in mammals, but in chicken the vehicle used by the migratory PGCs is the vascular system. We have analysed the migratory pathway of chicken PGCs, focusing on the period of transition from the extraembryonic region to the intraembryonic vascular system.Our findings show that at Hamburger and Hamilton developmental stage HH12-HH14 the majority of PGCs concentrate axially in the sinus terminalis and favour transport axially via the anterior vitelline veins into the embryonic circulation. Moreover, directly blocking the blood flow through the anterior vitelline veins resulted in an accumulation of PGCs in the anterior region and a decreased number of PGCs in the genital ridges. We further confirmed the key role for the anterior vitelline veins in the correct migration of PGCs using an ex ovo culture method that resulted in defective morphogenetic development of the anterior vitelline veins.We propose a novel model for the migratory pathway of chicken PGCs whereby the anterior vitelline veins play a central role at the extraembryonic and embryonic interface. The chicken model of PGC migration through the vasculature may be a powerful tool to study the process of homing (inflammation and metastasis) due to the striking similarities in regulatory signaling pathways (SDF1-CXCR4) and the transient role of the vasculature.

15.
Genes Dev ; 26(22): 2477-82, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23154982

RESUMEN

Transcription elongation is stimulated by positive transcription elongation factor b (P-TEFb), for which activity is repressed in the 7SK small nuclear ribonucleoprotein (7SK snRNP) complex. We show here a critical role of 7SK snRNP in growth control of primordial germ cells (PGCs). The expression of p15(INK4b), a cyclin-dependent kinase inhibitor (CDKI) gene, in PGCs is selectively activated by P-TEFb and its recruiting molecule, Brd4, when the amount of active P-TEFb is increased due to reduction of the 7SK snRNP, and PGCs consequently undergo growth arrest. These results indicate that CDKI gene-specific control of transcription by 7SK snRNP plays a pivotal role in the maintenance of PGC proliferation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genes cdc/genética , Células Germinativas/citología , Células Germinativas/metabolismo , Animales , Ciclo Celular , Proliferación Celular , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Proteínas Nucleares/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Factores de Transcripción/metabolismo
16.
Primates ; 53(2): 205-13, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22075965

RESUMEN

Induced pluripotent stem (iPS) cells have the potential to become a universal resource for cell-based therapies in regenerative medicine; however, prior to the use of such iPS cell-based therapies, preclinical assessment of their safety and efficacy is essential. Non-human primates serve as valuable animal models for human diseases or biomedical research; therefore, in this study, we generated cynomolgus monkey iPS cells from adult skin and fetal fibroblast cells by the retrovirally mediated introduction of four human transcription factors: c-Myc, Klf4, Oct3/4, and Sox2 (the so-called "Yamanaka factors"). Twenty to 30 days after the introduction of these factors, several cynomolgus monkey embryonic stem (ES) cell-like colonies appeared on SNL and mouse embryonic fibroblast (MEF) feeder layers. These colonies were picked and cultivated in primate ES medium. Seven iPS cell lines were established, and we detected the expression of pluripotent markers that are also expressed in ES cells. Reverse transcription polymerase chain reaction (PCR) showed that these iPS cells expressed endogenous c-Myc, Klf4, Oct3/4, and Sox2 genes, whereas several transgenes were silenced. Embryoid body and teratoma formation showed that the cynomolgus iPS cells had the developmental potential to differentiate into cells of all three primary germ layers. In summary, we generated cynomolgus monkey iPS cells by retrovirus-mediated transduction of the human transcription factors, c-Myc, Klf4, Oct3/4, and Sox2 into adult cynomolgus monkey skin cells and fetal fibroblasts. The cynomolgus monkey is the most relevant primate model for human disease, and the highly efficient generation of monkey iPS cells would allow investigation of the treatments of various diseases in this model via therapeutic cloning.


Asunto(s)
Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Macaca fascicularis/fisiología , Células Madre Pluripotentes/citología , Factores de Transcripción/farmacología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Fibroblastos/fisiología , Humanos , Factor 4 Similar a Kruppel , Ratones , Ratones SCID , Células Madre Pluripotentes/fisiología , Piel/citología , Teratoma
17.
Dev Cell ; 20(3): 364-75, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21397847

RESUMEN

MITOPLD is a member of the phospholipase D superfamily proteins conserved among diverse species. Zucchini (Zuc), the Drosophila homolog of MITOPLD, has been implicated in primary biogenesis of Piwi-interacting RNAs (piRNAs). By contrast, MITOPLD has been shown to hydrolyze cardiolipin in the outer membrane of mitochondria to generate phosphatidic acid, which is a signaling molecule. To assess whether the mammalian MITOPLD is involved in piRNA biogenesis, we generated Mitopld mutant mice. The mice display meiotic arrest during spermatogenesis, demethylation and derepression of retrotransposons, and defects in primary piRNA biogenesis. Furthermore, in mutant germ cells, mitochondria and the components of the nuage, a perinuclear structure involved in piRNA biogenesis/function, are mislocalized to regions around the centrosome, suggesting that MITOPLD may be involved in microtubule-dependent localization of mitochondria and these proteins. Our results indicate a conserved role for MITOPLD/Zuc in the piRNA pathway and link mitochondrial membrane metabolism/signaling to small RNA biogenesis.


Asunto(s)
Células Germinativas , Isoenzimas/metabolismo , Proteínas Mitocondriales/metabolismo , Fosfolipasa D/metabolismo , ARN Interferente Pequeño/metabolismo , Espermatogénesis/fisiología , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Células Germinativas/citología , Células Germinativas/fisiología , Humanos , Isoenzimas/genética , Masculino , Meiosis/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Fosfolipasa D/genética , ARN Interferente Pequeño/genética , Retroelementos , Transducción de Señal/fisiología , Testículo/citología
18.
Biochem Biophys Res Commun ; 402(4): 680-6, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20977886

RESUMEN

A novel member of the human ppGalNAc-T family, ppGalNAc-T20, was identified and characterized. Amino acid alignment revealed a high sequence identity between ppGalNAc-T20 and -T10. In the GalNAc transfer assay towards mucin-derived peptide substrates, the recombinant ppGalNAc-T20 demonstrated to be a typical glycopeptide GalNAc-transferase that exhibits activity towards mono-GalNAc-glycosylated peptide EA2 derived from rat submandibular gland mucin but no activity towards non-modified EA2. The in vitro catalytic property of ppGalNAc-T20 was compared with that of ppGalNAc-T10 to show different acceptor substrate specificities and kinetic constants. The ppGalNAc-T20 transcript was found exclusively in testis and brain. In situ hybridization further reveals that ppGalNAc-T20 was specifically localized in primary and secondary spermatocytes of the two meiotic periods, suggesting that it may involve in O-glycosylation during mouse spermatogenesis.


Asunto(s)
Encéfalo/enzimología , N-Acetilgalactosaminiltransferasas/metabolismo , Testículo/enzimología , Secuencia de Aminoácidos , Animales , Catálisis , Glicosilación , Humanos , Hibridación in Situ , Masculino , Ratones , Datos de Secuencia Molecular , N-Acetilgalactosaminiltransferasas/clasificación , N-Acetilgalactosaminiltransferasas/genética , Filogenia , Ratas , Transcripción Genética , Polipéptido N-Acetilgalactosaminiltransferasa
19.
PLoS One ; 5(9)2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20824066

RESUMEN

BACKGROUND: During fertilization, pronuclear envelope breakdown (PNEB) is followed by the mingling of male and female genomes. Dynamic chromatin and protein rearrangements require posttranslational modification (PTM) for the postfertilization development. METHODOLOGY/PRINCIPAL FINDINGS: Inhibition of poly(ADP-ribose) polymerase activity (PARylation) by either PJ-34 or 5-AIQ resulted in developmental arrest of fertilized embryos at the PNEB. PARylation inhibition affects spindle bundle formation and phosphorylation of Erk molecules of metaphase II (MII) unfertilized oocytes. We found a frequent appearance of multiple pronuclei (PN) in the PARylation-inhibited embryos, suggesting defective polymerization of tubulins. Attenuated phosphorylation of lamin A/C by PARylation was detected in the PARylation-inhibited embryos at PNEB. This was associated with sustained localization of heterodomain protein 1 (HP1) at the PN of the one-cell embryos arrested by PARylation inhibition. CONCLUSIONS/SIGNIFICANCE: Our findings indicate that PARylation is required for pronuclear fusion during postfertilization processes. These data further suggest that PARylation regulates protein dynamics essential for the beginning of mouse zygotic development. PARylation and its involving signal-pathways may represent potential targets as contraceptives.


Asunto(s)
División del Núcleo Celular , Núcleo Celular/metabolismo , Fertilización , Poli Adenosina Difosfato Ribosa/metabolismo , Animales , Núcleo Celular/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Poli(ADP-Ribosa) Polimerasas/metabolismo , Cigoto/citología , Cigoto/metabolismo
20.
Mol Reprod Dev ; 77(9): 802-11, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20722049

RESUMEN

Pluripotent stem cells can be established by various methods, but they share several cytological properties, including germ cell differentiation in vitro, independently of their origin. Although mouse induced pluripotent stem (iPS) cells can produce functional gametes in vivo, it is still unclear whether or not they have the ability to produce presumptive germ cells in vitro. Here, we show that mouse iPS cells derived from adult hepatocytes were able to differentiate into presumptive germ cells marked by mouse vasa homolog (Mvh) expression in feeder-free or suspension cultures. Embryoid body (EB) formation from iPS cells also induced the formation of round-shaped cells resembling immature oocytes. Mvh(+) cells formed clumps by co-aggregation with differentiation-supporting cells, and increased expression of germ cell markers was detected in these cell aggregates. Differentiation culture of presumptive germ cells from iPS cells could provide a conventional system for facilitating our understanding of the mechanisms underlying direct reprogramming and germline competency.


Asunto(s)
Células Madre Embrionarias/citología , Hepatocitos/citología , Células Madre Pluripotentes Inducidas/citología , Animales , Diferenciación Celular , ARN Helicasas DEAD-box/análisis , ARN Helicasas DEAD-box/genética , Cuerpos Embrioides/citología , Células Germinativas/citología , Masculino , Ratones , Ratones Transgénicos , Oocitos/citología , Retroviridae
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...