Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biol Reprod ; 105(2): 543-553, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-33982061

RESUMEN

In mammalian cloning by somatic cell nuclear transfer (SCNT), the treatment of reconstructed embryos with histone deacetylase (HDAC) inhibitors improves efficiency. So far, most of those used for SCNT are hydroxamic acid derivatives-such as trichostatin A-characterized by their broad inhibitory spectrum. Here, we examined whether mouse SCNT efficiency could be improved using chlamydocin analogues, a family of newly designed agents that specifically inhibit class I and IIa HDACs. Development of SCNT-derived embryos in vitro and in vivo revealed that four out of five chlamydocin analogues tested could promote the development of cloned embryos. The highest pup rates (7.1-7.2%) were obtained with Ky-9, similar to those achieved with trichostatin A (7.2-7.3%). Thus, inhibition of class I and/or IIa HDACs in SCNT-derived embryos is enough for significant improvements in full-term development. In mouse SCNT, the exposure of reconstructed oocytes to HDAC inhibitors is limited to 8-10 h because longer inhibition with class I inhibitors causes a two-cell developmental block. Therefore, we used Ky-29, with higher selectivity for class IIa than class I HDACs for longer treatment of SCNT-derived embryos. As expected, 24-h treatment with Ky-29 up to the two-cell stage did not induce a developmental block, but the pup rate was not improved. This suggests that the one-cell stage is a critical period for improving SCNT cloning using HDAC inhibitors. Thus, chlamydocin analogues appear promising for understanding and improving the epigenetic status of mammalian SCNT-derived embryos through their specific inhibitory effects on HDACs.


Asunto(s)
Inhibidores de Histona Desacetilasas/química , Técnicas de Transferencia Nuclear/instrumentación , Oocitos/química , Animales , Inhibidores de Histona Desacetilasas/clasificación , Ratones , Péptidos Cíclicos/química
2.
J Reprod Dev ; 66(1): 67-73, 2020 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-31852860

RESUMEN

Improving artificial oocyte activation is essential for assisted reproduction or animal biotechnology that can obtain healthy offspring with a high success rate. Here, we examined whether intracytoplasmic injection of equine sperm-specific phospholipase C zeta (ePLCζ) mRNA, the PLCζ with the strongest oocyte activation potential in mammals, could improve the mouse oocyte activation rate and subsequent embryonic development using inactivated spermatozoa. mRNA of mouse PLCζ (mPLCζ) or ePLCζ were injected into mouse oocytes to determine the optimal mRNA concentration to maximize the oocyte activation rate and developmental rate of parthenogenetic embryos in vitro. Full-term development was examined using NaOH-treated inactive spermatozoa using the optimal activation method. We found that the most optimal ePLCζ mRNA concentration was 0.1 ng/µl for mouse oocyte activation, which was ten times stronger than mPLCζ mRNA. The concentration did not affect parthenogenetic embryo development in vitro. Relatively normal blastocysts were obtained with the same developmental rate (52-53% or 48-51%, respectively) when inactive spermatozoa were injected into activated oocytes using ePLCζ or mPLCζ mRNA injection. However, the birth rate after embryo transfer was slightly but significantly decreased in oocytes activated by ePLCζ mRNA (24%) compared to mPLCζ mRNA (37%) or strontium treatment (40%) activation. These results suggest that the higher activation rate does not always correlate the higher birth rate, and some mechanisms might exist in the oocyte activation process that could affect the later developmental stages like full-term development.


Asunto(s)
Desarrollo Embrionario/fisiología , Oocitos/metabolismo , Fosfoinositido Fosfolipasa C/metabolismo , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/metabolismo , Animales , Femenino , Caballos , Masculino , Ratones , Fosfoinositido Fosfolipasa C/genética , Inyecciones de Esperma Intracitoplasmáticas
3.
Proc Natl Acad Sci U S A ; 114(23): 5988-5993, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28533361

RESUMEN

If humans ever start to live permanently in space, assisted reproductive technology using preserved spermatozoa will be important for producing offspring; however, radiation on the International Space Station (ISS) is more than 100 times stronger than that on Earth, and irradiation causes DNA damage in cells and gametes. Here we examined the effect of space radiation on freeze-dried mouse spermatozoa held on the ISS for 9 mo at -95 °C, with launch and recovery at room temperature. DNA damage to the spermatozoa and male pronuclei was slightly increased, but the fertilization and birth rates were similar to those of controls. Next-generation sequencing showed only minor genomic differences between offspring derived from space-preserved spermatozoa and controls, and all offspring grew to adulthood and had normal fertility. Thus, we demonstrate that although space radiation can damage sperm DNA, it does not affect the production of viable offspring after at least 9 mo of storage on the ISS.


Asunto(s)
Daño del ADN/efectos de la radiación , Desarrollo Embrionario/efectos de la radiación , Espermatozoides/efectos de la radiación , Animales , Transferencia de Embrión/métodos , Transferencia de Embrión/mortalidad , Femenino , Liofilización/métodos , Células Germinativas/efectos de la radiación , Tamaño de la Camada/efectos de la radiación , Masculino , Ratones , Oocitos , Técnicas Reproductivas Asistidas , Vuelo Espacial , Inyecciones de Esperma Intracitoplasmáticas/métodos , Espermatozoides/fisiología
4.
EMBO Rep ; 18(6): 885-893, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28468955

RESUMEN

T-cell receptor (TCR)-transgenic mice have been employed for evaluating antigen-response mechanisms, but their non-endogenous TCR might induce immune response differently than the physiologically expressed TCR Nuclear transfer cloning produces animals that retain the donor genotype in all tissues including germline and immune systems. Taking advantage of this feature, we generated cloned mice that carry endogenously rearranged TCR genes from antigen-specific CD4+ T cells. We show that T cells of the cloned mice display distinct developmental pattern and antigen reactivity because of their endogenously pre-rearranged TCRα (rTα) and TCRß (rTß) alleles. These alleles were transmitted to the offspring, allowing us to establish a set of mouse lines that show chronic-type allergic phenotypes, that is, bronchial and nasal inflammation, upon local administrations of the corresponding antigens. Intriguingly, the existence of either rTα or rTß is sufficient to induce in vivo hypersensitivity. These cloned mice expressing intrinsic promoter-regulated antigen-specific TCR are a unique animal model with allergic predisposition for investigating CD4+ T-cell-mediated pathogenesis and cellular commitment in immune diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Hipersensibilidad/inmunología , Técnicas de Transferencia Nuclear , Receptores de Antígenos de Linfocitos T/genética , Alelos , Animales , Antígenos/administración & dosificación , Antígenos/inmunología , Clonación de Organismos , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología
5.
J Reprod Dev ; 65(4): 353-359, 2019 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-31118350

RESUMEN

Freeze-drying of spermatozoa is a convenient and safe method to preserve mammalian genetic material without the use of liquid nitrogen or a deep freezer. However, freeze-dried spermatozoa (FD sperm) are not frequently used because of the low success rate of offspring after intracytoplasmic spermatozoa injection (ICSI). In this study, we determined the optimal concentration and a point of action of trehalose as a protectant for the preservation of FD sperm from different mouse strains at room temperature (RT). Although trehalose demonstrated no potential to protect the FD sperm of ICR mice against the freeze-drying procedure itself, the blastocyst rate was significantly improved when FD sperm was preserved for more than 1 month at RT (56-63% vs. 29% without trehalose). The optimal concentration of trehalose was 0.5 M. Importantly, remarkable results were obtained when spermatozoa of inbred mouse strains (C57BL/6N, C3H/He, and 129/Sv) were used, and many offspring were obtained when FD sperm that was preserved for 3 months at RT (26-28% vs. 6-11% of without trehalose) was used. However, when DNA damage in FD sperm was examined by gamma-H2Ax assays, it was found that trehalose failed to protect the FD sperm from DNA damage. These results suggest that trehalose has the potential to protect other sperm factors rather than sperm DNA during preservation at RT for longer periods and trehalose is more effective for inbred mouse strains.


Asunto(s)
Preservación de Semen/métodos , Espermatozoides , Trehalosa/farmacología , Animales , Femenino , Liofilización/métodos , Liofilización/veterinaria , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Soluciones Preservantes de Órganos/farmacología , Embarazo , Índice de Embarazo , Preservación de Semen/veterinaria , Inyecciones de Esperma Intracitoplasmáticas
6.
Proc Natl Acad Sci U S A ; 112(47): 14641-6, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26546670

RESUMEN

Substantial proportions of mammalian genomes comprise repetitive elements including endogenous retrotransposons. Although these play diverse roles during development, their appropriate silencing is critically important in maintaining genomic integrity in the host cells. The major mechanism for retrotransposon silencing is DNA methylation, but the wave of global DNA demethylation that occurs after fertilization renders preimplantation embryos exceptionally hypomethylated. Here, we show that hypomethylated preimplantation mouse embryos are protected from retrotransposons by repressive histone modifications mediated by the histone chaperone chromatin assembly factor 1 (CAF-1). We found that knockdown of CAF-1 with specific siRNA injections resulted in significant up-regulation of the retrotransposons long interspersed nuclear element 1, short interspersed nuclear element B2, and intracisternal A particle at the morula stage. Concomitantly, increased histone H2AX phosphorylation and developmental arrest of the majority (>95%) of embryos were observed. The latter was caused at least in part by derepression of retrotransposons, as treatment with reverse transcriptase inhibitors rescued some embryos. Importantly, ChIP analysis revealed that CAF-1 mediated the replacement of H3.3 with H3.1/3.2 at the retrotransposon regions. This replacement was associated with deposition of repressive histone marks, including trimethylation of histone H3 on lysine 9 (H3K9me3), H3K9me2, H3K27me3, and H4K20me3. Among them, H4K20me3 and H3K9me3 seemed to play predominant roles in retrotransposon silencing, as assessed by knockdown of specific histone methyltransferases and forced expression of unmethylatable mutants of H3.1K9 and H4K20. Our data thus indicate that CAF-1 is an essential guardian of the genome in preimplantation mouse embryos by deposition of repressive histone modifications via histone variant replacement.


Asunto(s)
Blastocisto/metabolismo , Factor 1 de Ensamblaje de la Cromatina/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Retroelementos/genética , Animales , Blastocisto/efectos de los fármacos , Núcleo Celular/metabolismo , Inmunoprecipitación de Cromatina , Femenino , Técnicas de Silenciamiento del Gen , Genes Dominantes , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Lisina/metabolismo , Masculino , Metilación/efectos de los fármacos , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Mutación/genética , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología
7.
Reproduction ; 154(6): 859-866, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28971892

RESUMEN

Recently, it has become possible to generate cloned mice using a somatic cell nucleus derived from not only F1 strains but also inbred strains. However, to date, all cloned mice have been generated using F1 mouse oocytes as the recipient cytoplasm. Here, we attempted to generate cloned mice from oocytes derived from the ICR-outbred mouse strain. Cumulus cell nuclei derived from BDF1 and ICR mouse strains were injected into enucleated oocytes of both strains to create four groups. Subsequently, the quality and developmental potential of the cloned embryos were examined. ICR oocytes were more susceptible to damage associated with nuclear injection than BDF1 oocytes, but their activation rate and several epigenetic markers of reconstructed cloned oocytes/embryos were similar to those of BDF1 oocytes. When cloned embryos were cultured for up to 4 days, those derived from ICR oocytes demonstrated a significantly decreased rate of development to the blastocyst stage, irrespective of the nuclear donor mouse strain. However, when cloned embryos derived from ICR oocytes were transferred to female recipients at the two-cell stage, healthy cloned offspring were obtained at a success rate similar to that using BDF1 oocytes. The ICR mouse strain is very popular for biological research and less expensive to establish than most other strains. Thus, the results of this study should promote the study of nuclear reprogramming not only by reducing the cost of experiments but also by allowing us to study the effect of oocyte cytoplasm by comparing it between strains.


Asunto(s)
Blastocisto/fisiología , Clonación de Organismos/métodos , Técnicas de Transferencia Nuclear , Oocitos/fisiología , Animales , Cruzamientos Genéticos , Técnicas de Cultivo de Embriones , Implantación del Embrión , Transferencia de Embrión , Femenino , Edad Gestacional , Nacimiento Vivo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Embarazo , Inyecciones de Esperma Intracitoplasmáticas
8.
Biol Reprod ; 92(3): 81, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25653280

RESUMEN

Whereas cloning mammals by direct somatic cell nuclear transfer has been successful using a wide range of donor cell types, neurons from adult brain remain "unclonable" for unknown reasons. Here, using a combination of two epigenetic approaches, we examined whether neurons from adult mice could be cloned. First, we used a specific antibody to discover cell types with reduced amounts of a repressive histone mark-dimethylated histone H3 lysine 9 (H3K9me2)-and identified CA1 pyramidal cells in the hippocampus and Purkinje cells in the cerebellum as candidates. Second, reconstructed embryos were treated with trichostatin A (TSA), a potent histone deacetylase inhibitor. Using CA1 cells, cloned offspring were obtained at high rates, reaching 10.2% and 4.6% (of embryos transferred) for male and female donors, respectively. Cerebellar Purkinje cell nuclei were too large to maintain their genetic integrity during nuclear transfer, leading to developmental arrest of embryos. However, gene expression analysis using cloned blastocysts corroborated a high rate of genomic reprogrammability of CA1 pyramidal and Purkinje cells. Neurons from the hippocampal dentate gyrus and cerebral cortex, which had higher amounts of H3K9me2, could also be used for producing cloned offspring, but the efficiencies were low. A more thorough analysis revealed that TSA treatment was essential for cloning adult neuronal cells. This study demonstrates, to our knowledge for the first time, that adult neurons can be cloned by nuclear transfer. Furthermore, our data imply that reduced amounts of H3K9me2 and increased histone acetylation appear to act synergistically to improve the development of cloned embryos.


Asunto(s)
Clonación de Organismos/métodos , Neuronas/citología , Técnicas de Transferencia Nuclear , Células de Purkinje/citología , Animales , Células Cultivadas , Desarrollo Embrionario , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Histona Demetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Modelos Animales , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo
9.
Hum Reprod ; 30(5): 1178-87, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25740879

RESUMEN

STUDY QUESTION: Is the poor development of embryos generated from round spermatid injection (ROSI) in humans and animals associated with abnormal active DNA demethylation? SUMMARY ANSWER: A significant proportion of ROSI-derived embryos failed to undergo active DNA demethylation. WHAT IS KNOWN ALREADY: Active DNA demethylation is initiated by the conversion of 5-methylcytosine (5mC) to 5-hydroxycytosine (5hmC) by the Tet3 enzyme. Active demethylation proceeds in a more pronounced manner in the male pronucleus than in the female one. PARTICIPANTS/MATERIALS, SETTING, METHODS: Mouse zygotes generated by ICSI or ROSI were analyzed for active DNA methylation by quantification of 5mC and 5hmC using specific antibodies. Some ROSI-derived embryos were subjected to time-lapse imaging for DNA methylation levels and were transferred into recipient pseudo-pregnant female mice. MAIN RESULTS AND THE ROLE OF CHANCE: In ICSI-derived embryos, the male:female pronucleus (M/F) ratio of 5mC immunostaining intensity was decreased while that of 5hmC was increased. However, a significant proportion of ROSI-derived embryos showed unchanged M/F ratios for 5mC and 5hmC even at the late zygotic period, indicating that they failed to undergo asymmetric active DNA demethylation. Consistent with this, some ROSI-derived embryos did not show preferential localization of Tet3 to the male pronucleus. ROSI-derived embryos were classified into 'demethylated' or 'non-demethylated' groups by time-lapse imaging and transferred into recipient female mice separately. More normal-sized fetuses were retrieved from the 'demethylated' group than 'non-demethylated' group at Day 11.5 of pregnancy. LIMITATIONS, REASONS FOR CAUTION: A causal relationship between impaired active DNA demethylation and the poor developmental ability of ROSI-derived embryos remains to be determined. WIDER IMPLICATIONS OF THE FINDINGS: We identified two types of ROSI-derived embryos in terms of the degree of active DNA demethylation. Induction of normal DNA demethylation at the zygotic stage might help in the technical improvement of ROSI. STUDY FUNDING/COMPETING INTERESTS: The work was supported by Grants-in-Aid from the Ministry of Education, Culture, Sports, Science and Technology of Japan and by the RIKEN Epigenetics Program. The authors have no competing interests to declare.


Asunto(s)
Núcleo Celular/metabolismo , Metilación de ADN , Espermátides/fisiología , Cigoto/metabolismo , 5-Metilcitosina/química , Animales , Gonadotropina Coriónica/química , Citosina/análogos & derivados , Citosina/química , Transferencia de Embrión , Femenino , Células Germinativas/citología , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Microscopía Fluorescente , Oocitos/citología , Embarazo , Preñez , Inyecciones de Esperma Intracitoplasmáticas/métodos
11.
Biol Reprod ; 91(5): 120, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25232016

RESUMEN

In mice, the establishment of paternal genomic imprinting in male germ cells starts at midgestation, as suggested by DNA methylation analyses of differentially methylated regions (DMRs). However, this information is based on averages from mixed populations of germ cells, and the DNA methylation pattern might not always provide a full representation of imprinting status. To obtain more detailed information on the establishment of paternal imprinting, single prospermatogonia at Embryonic Days 15.5 (E15.5), E16.5, and E17.5 and at Day 0.5 after birth were cloned using nuclear transfer; previous reports suggested that cloned embryos reflected the donor's genomic imprinting status. Then, the resultant fetuses (E9.5) were analyzed for the DNA methylation pattern of three paternal DMRs (IG-DMR, H19 DMR, and Rasgrf1 DMR) and the expression pattern of imprinted genes therein. The overall data indicated that establishment of genomic imprinting in all paternally imprinted regions was completed by E17.5, following a short intermediate period at E16.5. Furthermore, comparison between the methylation status of DMRs and the expression profiles of imprinted genes suggested that methylation of the IG-DMR, but not the H19 DMR, solely governed the control of its imprinted gene cluster. The Rasgrf1 DMR seemed to be imprinted later than the other two genes. We also found that the methylation status of the Gtl2 DMR, the secondary DMR that acquires DNA methylation after fertilization, was likely to follow the methylation status of the upstream IG-DMR. Thus, the systematic analyses of prospermatogonium-derived embryos provided additional important information on the establishment of paternal imprinting.


Asunto(s)
Células Madre Adultas/metabolismo , Padre , Impresión Genómica , Técnicas de Transferencia Nuclear , Células Madre Adultas/citología , Animales , Células Cultivadas , Clonación de Organismos/métodos , Metilación de ADN , Embrión de Mamíferos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR
12.
Nat Commun ; 15(1): 4946, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862540

RESUMEN

Genomic aberrations are a critical impediment for the safe medical use of iPSCs and their origin and developmental mechanisms remain unknown. Here we find through WGS analysis of human and mouse iPSC lines that genomic mutations are de novo events and that, in addition to unmodified cytosine base prone to deamination, the DNA methylation sequence CpG represents a significant mutation-prone site. CGI and TSS regions show increased mutations in iPSCs and elevated mutations are observed in retrotransposons, especially in the AluY subfamily. Furthermore, increased cytosine to thymine mutations are observed in differentially methylated regions. These results indicate that in addition to deamination of cytosine, demethylation of methylated cytosine, which plays a central role in genome reprogramming, may act mutagenically during iPSC generation.


Asunto(s)
Islas de CpG , Citosina , Metilación de ADN , Células Madre Pluripotentes Inducidas , Mutación Puntual , Células Madre Pluripotentes Inducidas/metabolismo , Citosina/metabolismo , Animales , Humanos , Ratones , Reprogramación Celular/genética , Retroelementos/genética , Línea Celular
13.
Chem Sci ; 15(17): 6349-6362, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38699251

RESUMEN

Careful control of electronic properties, structural order, and solubility of π-conjugated polymers is central to the improvement of organic photovoltaic (OPV) performance. In this work, we designed and synthesized a series of naphthobisthiadiazole-quaterthiophene copolymers by systematically replacing the alkyl groups with ester groups and changing the position of the fluorine groups in the quaterthiophene moiety. These alterations lowered the HOMO and LUMO energy levels and systematically varied the combination of intramolecular noncovalent interactions such as O⋯S and F⋯S interactions in the backbone. More importantly, although the introduction of such noncovalent interactions often lowers the solubility owing to the interlocking of backbone linkages, we found that careful design of the noncovalent interactions afforded polymers with relatively high solubility and high crystallinity at the same time. As a result, the power conversion efficiency of OPV cells that used fullerene (PC61BM) and nonfullerene (Y12) as the acceptor was improved. Our work offers important information for the development of high-performance π-conjugated polymers for OPVs.

14.
Biol Reprod ; 89(2): 24, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23803554

RESUMEN

Somatic cell nuclear transfer (SCNT) is a unique technology that produces cloned animals from single cells. It is desirable from a practical viewpoint that donor cells can be collected noninvasively and used readily for nuclear transfer. The present study was undertaken to determine whether peripheral blood cells freshly collected from living mice could be used for SCNT. We collected a drop of peripheral blood (15-45 µl) from the tail of a donor. A nucleated cell (leukocyte) suspension was prepared by lysing the red blood cells. Following SCNT using randomly selected leukocyte nuclei, cloned offspring were born at a 2.8% birth rate. Fluorescence-activated cell sorting revealed that granulocytes/monocytes and lymphocytes could be roughly distinguished by their sizes, the former being significantly larger. We then cloned putative granulocytes/monocytes and lymphocytes separately and obtained 2.1% and 1.7% birth rates, respectively (P > 0.05). Because the use of lymphocyte nuclei inevitably results in the birth of offspring with DNA rearrangements, we applied granulocyte/monocyte cloning to two genetically modified strains and two recombinant inbred strains. Normal-looking offspring were obtained from all four strains tested. The present study clearly indicated that genetic copies of mice could be produced using a drop of peripheral blood from living donors. This strategy will be applied to the rescue of infertile founder animals or a "last-of-line" animal possessing invaluable genetic resources.


Asunto(s)
Clonación de Organismos/métodos , Embrión de Mamíferos/citología , Técnicas de Transferencia Nuclear , Animales , Transferencia de Embrión , Fibroblastos/citología , Citometría de Flujo , Ratones
15.
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
16.
Commun Biol ; 5(1): 699, 2022 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-35835981

RESUMEN

The underlying mechanism for parental asymmetric chromatin dynamics is still unclear. To reveal this, we investigate chromatin dynamics in parthenogenetic, androgenic, and several types of male germ cells-fertilized zygotes. Here we illustrate that parental conflicting role mediates the regulation of chromatin dynamics. Sperm reduces chromatin dynamics in both parental pronuclei (PNs). During spermiogenesis, male germ cells acquire this reducing ability and its resistance. On the other hand, oocytes can increase chromatin dynamics. Notably, the oocytes-derived chromatin dynamics enhancing ability is dominant for the sperm-derived opposing one. This maternal enhancing ability is competed between parental pronuclei. Delayed fertilization timing is critical for this competition and compromises parental asymmetric chromatin dynamics and zygotic transcription. Together, parental competition for the maternal factor enhancing chromatin dynamics is a determinant to establish parental asymmetry, and paternal repressive effects have supporting roles to enhance asymmetry.


Asunto(s)
Cromatina , Cigoto , Animales , Núcleo Celular , Cromatina/genética , Histonas , Masculino , Ratones , Semen
17.
Stem Cell Reports ; 16(10): 2503-2519, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34559999

RESUMEN

We here demonstrate that microsatellite (MS) alterations are elevated in both mouse and human induced pluripotent stem cells (iPSCs), but importantly we have now identified a type of human iPSC in which these alterations are considerably reduced. We aimed in our present analyses to profile the InDels in iPSC/ntESC genomes, especially in MS regions. To detect somatic de novo mutations in particular, we generated 13 independent reprogramed stem cell lines (11 iPSC and 2 ntESC lines) from an identical parent somatic cell fraction of a C57BL/6 mouse. By using this cell set with an identical genetic background, we could comprehensively detect clone-specific alterations and, importantly, experimentally validate them. The effectiveness of employing sister clones for detecting somatic de novo mutations was thereby demonstrated. We then successfully applied this approach to human iPSCs. Our results require further careful genomic analysis but make an important inroad into solving the issue of genome abnormalities in iPSCs.


Asunto(s)
Perfil Genético , Mutación INDEL , Células Madre Pluripotentes Inducidas/metabolismo , Repeticiones de Microsatélite , Animales , Células Cultivadas , Reprogramación Celular , Técnicas de Reprogramación Celular/métodos , Humanos , Ratones , Ratones Endogámicos C57BL , Secuenciación Completa del Genoma
18.
Sci Adv ; 7(24)2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34117068

RESUMEN

Space radiation may cause DNA damage to cells and concern for the inheritance of mutations in offspring after deep space exploration. However, there is no way to study the long-term effects of space radiation using biological materials. Here, we developed a method to evaluate the biological effect of space radiation and examined the reproductive potential of mouse freeze-dried spermatozoa stored on the International Space Station (ISS) for the longest period in biological research. The space radiation did not affect sperm DNA or fertility after preservation on ISS, and many genetically normal offspring were obtained without reducing the success rate compared to the ground-preserved control. The results of ground x-ray experiments showed that sperm can be stored for more than 200 years in space. These results suggest that the effect of deep space radiation on mammalian reproduction can be evaluated using spermatozoa, even without being monitored by astronauts in Gateway.

19.
Nat Commun ; 11(1): 2150, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32358519

RESUMEN

Somatic cell nuclear transfer (SCNT) in mammals is an inefficient process that is frequently associated with abnormal phenotypes, especially in placentas. Recent studies demonstrated that mouse SCNT placentas completely lack histone methylation (H3K27me3)-dependent imprinting, but how it affects placental development remains unclear. Here, we provide evidence that the loss of H3K27me3 imprinting is responsible for abnormal placental enlargement and low birth rates following SCNT, through upregulation of imprinted miRNAs. When we restore the normal paternal expression of H3K27me3-dependent imprinted genes (Sfmbt2, Gab1, and Slc38a4) in SCNT placentas by maternal knockout, the placentas remain enlarged. Intriguingly, correcting the expression of clustered miRNAs within the Sfmbt2 gene ameliorates the placental phenotype. Importantly, their target genes, which are confirmed to cause SCNT-like placental histology, recover their expression level. The birth rates increase about twofold. Thus, we identify loss of H3K27me3 imprinting as an epigenetic error that compromises embryo development following SCNT.


Asunto(s)
Histonas/metabolismo , MicroARNs/genética , Placenta/metabolismo , Proteínas Represoras/genética , Animales , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Femenino , Impresión Genómica , Ratones , Familia de Multigenes/genética , Embarazo , ARN no Traducido/genética , ARN no Traducido/metabolismo
20.
Nat Commun ; 11(1): 197, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31924765

RESUMEN

A number of point mutations have been identified in reprogrammed pluripotent stem cells such as iPSCs and ntESCs. The molecular basis for these mutations has remained elusive however, which is a considerable impediment to their potential medical application. Here we report a specific stage at which iPSC generation is not reduced in response to ionizing radiation, i.e. radio-resistance. Quite intriguingly, a G1/S cell cycle checkpoint deficiency occurs in a transient fashion at the initial stage of the genome reprogramming process. These cancer-like phenomena, i.e. a cell cycle checkpoint deficiency resulting in the accumulation of point mutations, suggest a common developmental pathway between iPSC generation and tumorigenesis. This notion is supported by the identification of specific cancer mutational signatures in these cells. We describe efficient generation of human integration-free iPSCs using erythroblast cells, which have only a small number of point mutations and INDELs, none of which are in coding regions.


Asunto(s)
Puntos de Control de la Fase G1 del Ciclo Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/genética , Animales , División Celular , Reprogramación Celular , Eritroblastos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de la radiación , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de la radiación , Neoplasias/genética , Sistemas de Lectura Abierta , Mutación Puntual , Puntos de Control de la Fase S del Ciclo Celular/efectos de la radiación , Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA