Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Chem Sci ; 15(17): 6349-6362, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38699251

ABSTRACT

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.

2.
Commun Biol ; 5(1): 699, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835981

ABSTRACT

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.


Subject(s)
Chromatin , Zygote , Animals , Cell Nucleus , Chromatin/genetics , Histones , Male , Mice , Semen
3.
Stem Cell Reports ; 16(10): 2503-2519, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34559999

ABSTRACT

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.


Subject(s)
Genetic Profile , INDEL Mutation , Induced Pluripotent Stem Cells/metabolism , Microsatellite Repeats , Animals , Cells, Cultured , Cellular Reprogramming , Cellular Reprogramming Techniques/methods , Humans , Mice , Mice, Inbred C57BL , Whole Genome Sequencing
4.
Sci Adv ; 7(24)2021 Jun.
Article in English | MEDLINE | ID: mdl-34117068

ABSTRACT

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.

5.
Biol Reprod ; 105(2): 543-553, 2021 08 03.
Article in English | MEDLINE | ID: mdl-33982061

ABSTRACT

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.


Subject(s)
Histone Deacetylase Inhibitors/chemistry , Nuclear Transfer Techniques/instrumentation , Oocytes/chemistry , Animals , Histone Deacetylase Inhibitors/classification , Mice , Peptides, Cyclic/chemistry
6.
Nat Commun ; 11(1): 2150, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358519

ABSTRACT

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.


Subject(s)
Histones/metabolism , MicroRNAs/genetics , Placenta/metabolism , Repressor Proteins/genetics , Animals , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Female , Genomic Imprinting , Mice , Multigene Family/genetics , Pregnancy , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
7.
Nat Commun ; 11(1): 197, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31924765

ABSTRACT

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.


Subject(s)
G1 Phase Cell Cycle Checkpoints/genetics , Induced Pluripotent Stem Cells/metabolism , S Phase Cell Cycle Checkpoints/genetics , Animals , Cell Division , Cellular Reprogramming , Erythroblasts , G1 Phase Cell Cycle Checkpoints/radiation effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/radiation effects , Neoplasms/genetics , Open Reading Frames , Point Mutation , S Phase Cell Cycle Checkpoints/radiation effects , X-Rays
8.
J Reprod Dev ; 66(1): 67-73, 2020 Feb 14.
Article in English | MEDLINE | ID: mdl-31852860

ABSTRACT

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.


Subject(s)
Embryonic Development/physiology , Oocytes/metabolism , Phosphoinositide Phospholipase C/metabolism , Sperm-Ovum Interactions/physiology , Spermatozoa/metabolism , Animals , Female , Horses , Male , Mice , Phosphoinositide Phospholipase C/genetics , Sperm Injections, Intracytoplasmic
9.
J Reprod Dev ; 65(4): 353-359, 2019 Aug 09.
Article in English | MEDLINE | ID: mdl-31118350

ABSTRACT

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.


Subject(s)
Semen Preservation/methods , Spermatozoa , Trehalose/pharmacology , Animals , Female , Freeze Drying/methods , Freeze Drying/veterinary , Male , Mice , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred ICR , Organ Preservation Solutions/pharmacology , Pregnancy , Pregnancy Rate , Semen Preservation/veterinary , Sperm Injections, Intracytoplasmic
10.
Cell Rep ; 27(6): 1742-1754.e6, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31067460

ABSTRACT

Placental development is a key event in mammalian reproduction and embryogenesis. However, the molecular basis underlying placental development is not fully understood. Here, we conduct a forward genetic screen to identify regulators for extraembryonic development and identify Zfp281 as a key factor. Zfp281 overexpression in mouse embryonic stem cells facilitates the induction of trophoblast stem-like cells. Zfp281 is preferentially expressed in the undifferentiated trophoblast stem cell population in an FGF-dependent manner, and disruption of Zfp281 in mice causes severe defects in early placental development. Consistently, Zfp281-depleted trophoblast stem cells exhibit defects in maintaining the transcriptome and differentiation capacity. Mechanistically, Zfp281 interacts with MLL or COMPASS subunits and occupies the promoters of its target genes. Importantly, ZNF281, the human ortholog of this factor, is required to stabilize the undifferentiated status of human trophoblast stem cells. Thus, we identify Zfp281 as a conserved factor for the maintenance of trophoblast stem cell plasticity.


Subject(s)
Placentation/genetics , Repressor Proteins/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Transcriptome/genetics , Trophoblasts/cytology , Animals , Base Sequence , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/drug effects , Female , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Genetic Loci , Genetic Testing , Haploidy , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mice, Knockout , Placentation/drug effects , Pregnancy , Stem Cells/drug effects , Transcription, Genetic
11.
Life Sci Alliance ; 2(2)2019 04.
Article in English | MEDLINE | ID: mdl-30940732

ABSTRACT

Reactive oxygen species (ROS) play critical roles in self-renewal division for various stem cell types. However, it remains unclear how ROS signals are integrated with self-renewal machinery. Here, we report that the MAPK14/MAPK7/BCL6B pathway creates a positive feedback loop to drive spermatogonial stem cell (SSC) self-renewal via ROS amplification. The activation of MAPK14 induced MAPK7 phosphorylation in cultured SSCs, and targeted deletion of Mapk14 or Mapk7 resulted in significant SSC deficiency after spermatogonial transplantation. The activation of this signaling pathway not only induced Nox1 but also increased ROS levels. Chemical screening of MAPK7 targets revealed many ROS-dependent spermatogonial transcription factors, of which BCL6B was found to initiate ROS production by increasing Nox1 expression via ETV5-induced nuclear translocation. Because hydrogen peroxide or Nox1 transfection also induced BCL6B nuclear translocation, our results suggest that BCL6B initiates and amplifies ROS signals to activate ROS-dependent spermatogonial transcription factors by forming a positive feedback loop.


Subject(s)
Adult Germline Stem Cells/physiology , Cell Self Renewal/physiology , Reactive Oxygen Species/metabolism , Animals , Benzodiazepinones/pharmacology , Cell Proliferation/drug effects , DNA-Binding Proteins/metabolism , Feedback, Physiological/physiology , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , NADPH Oxidase 1/genetics , NADPH Oxidase 1/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
12.
Sci Rep ; 8(1): 14922, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297864

ABSTRACT

Cloning animals using nuclear transfer (NT) provides the opportunity to preserve endangered species. However, there are risks associated with the collection of donor cells from a body, which may cause accidental death of the animal. Here, we tried to collect faeces-derived cells and examined the usability of those nuclei as a donor for NT. A relatively large number of cells could be collected from GFP-Tg mouse faeces by this method. After NT, only 4.2% of the reconstructed oocytes formed pseudo-pronucleus. This rate increased up to 25% when GFP and Hoechst were used as a marker to select better cells. However, the reconstructed oocytes/embryos showed several abnormalities, such as shrunken nuclear membranes and abnormal distribution of tubulin, and none of them developed beyond one-cell stage embryos. These developmental failures were caused by not only toxic substances derived from faeces but also intrinsic DNA damage of donor cell nuclei. However, when the serial NT was performed, some of the cloned embryos could develop to the two-cell stage. This method may remove toxic substances and enhance DNA repair in the oocyte cytoplasm. Thus, these results indicate that faeces cells might be useful for the conservation of endangered species when technical improvements are achieved.


Subject(s)
Cloning, Organism/methods , Feces/cytology , Mice/embryology , Nuclear Transfer Techniques , Animals , Cell Separation/methods , DNA Damage , Female , Male , Mice/genetics , Oocytes/cytology , Oocytes/ultrastructure
13.
Epigenetics ; 13(7): 693-703, 2018.
Article in English | MEDLINE | ID: mdl-30079806

ABSTRACT

Although phenotypic abnormalities frequently appear in the placenta following somatic cell nuclear transfer (SCNT), mouse trophoblast stem cells (TSCs) established from SCNT embryos reportedly show no distinct abnormalities compared with those derived from normal fertilization. In this study, we reexamined SCNT-TSCs to identify their imprinting statuses. Placenta-specific maternally imprinted genes (Gab1, Slc38a4, and Sfmbt2) consistently showed biallelic expression in SCNT-TSCs, suggesting their loss of imprinting (LOI). The LOI of Gab1 was associated with decreased DNA methylation, and that of Sfmbt2 was associated with decreased DNA methylation and histone H3K27 trimethylation. The maternal allele of the intergenic differentially methylated region (IG-DMR) was aberrantly hypermethylated following SCNT, even though this region was prone to demethylation in TSCs when established in a serum-free chemically defined medium. These findings indicate that the development of cloned embryos is associated with imprinting abnormalities specifically in the trophoblast lineage from its initial stage, which may affect subsequent placental development.


Subject(s)
Embryonic Stem Cells/pathology , Epigenesis, Genetic , Genomic Imprinting , Nuclear Transfer Techniques/adverse effects , Placenta/abnormalities , Trophoblasts/pathology , Adaptor Proteins, Signal Transducing , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Animals , Blastocyst/metabolism , Blastocyst/pathology , Cloning, Organism , DNA Methylation , Embryonic Stem Cells/metabolism , Female , Mice , Mice, Inbred C57BL , Phosphoproteins/genetics , Phosphoproteins/metabolism , Placenta/metabolism , Placenta/pathology , Placentation , Pregnancy , Repressor Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/metabolism
14.
Sci Rep ; 8(1): 10602, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30006561

ABSTRACT

Freeze-drying has been frequently used to preserve food and microorganisms at room temperature (RT) for extended periods of time; however, its application to mammalian species is difficult. Here, we developed a method to prolong the stability of freeze-dried (FD) mice spermatozoa at RT for more than one year without using any cryoprotectant agents. Our data showed that maintaining a vacuum in ampoules is critical to ensuring the viability of FD spermatozoa, as the stability of spermatozoa DNA increased when imperfectly vacuumed ampoules were detected using a non-destructive test and eliminated. Finally a large number of healthy offspring were obtained from mice oocytes fertilized with FD spermatozoa stored at RT for more than one year. Although the birth rate from three-month stored spermatozoa was lower than that from one-day stored spermatozoa, no further reduction was observed even in one-year stored spermatozoa. Therefore, FD spermatozoa preserved in this study were highly tolerant to warm temperatures. This method of storage shows a great potential for the preservation of genetic resources of mammalian species, such as genetically-modified mouse strains, without the use of electric power.


Subject(s)
Cell Survival/physiology , Semen Preservation/methods , Spermatozoa/physiology , Animals , Animals, Newborn , Embryo Transfer , Female , Freeze Drying/methods , Male , Mice , Mice, Inbred ICR , Oocytes , Sperm Injections, Intracytoplasmic , Temperature , Vacuum
15.
Reproduction ; 154(6): 859-866, 2017 12.
Article in English | MEDLINE | ID: mdl-28971892

ABSTRACT

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.


Subject(s)
Blastocyst/physiology , Cloning, Organism/methods , Nuclear Transfer Techniques , Oocytes/physiology , Animals , Crosses, Genetic , Embryo Culture Techniques , Embryo Implantation , Embryo Transfer , Female , Gestational Age , Live Birth , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred ICR , Pregnancy , Sperm Injections, Intracytoplasmic
16.
Cell Rep ; 20(12): 2756-2765, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930672

ABSTRACT

At fertilization, the paternal genome undergoes extensive reprogramming through protamine-histone exchange and active DNA demethylation, but only a few maternal factors have been defined in these processes. We identified maternal Mettl23 as a protein arginine methyltransferase (PRMT), which most likely catalyzes the asymmetric dimethylation of histone H3R17 (H3R17me2a), as indicated by in vitro assays and treatment with TBBD, an H3R17 PRMT inhibitor. Maternal histone H3.3, which is essential for paternal nucleosomal assembly, is unable to be incorporated into the male pronucleus when it lacks R17me2a. Mettl23 interacts with Tet3, a 5mC-oxidizing enzyme responsible for active DNA demethylation, by binding to another maternal factor, GSE (gonad-specific expression). Depletion of Mettl23 from oocytes resulted in impaired accumulation of GSE, Tet3, and 5hmC in the male pronucleus, suggesting that Mettl23 may recruit GSE-Tet3 to chromatin. Our findings establish H3R17me2a and its catalyzing enzyme Mettl23 as key regulators of paternal genome reprogramming.


Subject(s)
Arginine/metabolism , Cellular Reprogramming , Genome , Histones/metabolism , Zygote/metabolism , 5-Methylcytosine/metabolism , Amino Acid Sequence , Animals , Chromosomal Proteins, Non-Histone , DNA Demethylation , DNA-Binding Proteins/metabolism , Dioxygenases , Embryonic Development , Male , Methylation , Methyltransferases/chemistry , Methyltransferases/metabolism , Mice , Oxidation-Reduction , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 114(23): 5988-5993, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28533361

ABSTRACT

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.


Subject(s)
DNA Damage/radiation effects , Embryonic Development/radiation effects , Spermatozoa/radiation effects , Animals , Embryo Transfer/methods , Embryo Transfer/mortality , Female , Freeze Drying/methods , Germ Cells/radiation effects , Litter Size/radiation effects , Male , Mice , Oocytes , Reproductive Techniques, Assisted , Space Flight , Sperm Injections, Intracytoplasmic/methods , Spermatozoa/physiology
19.
EMBO Rep ; 18(6): 885-893, 2017 06.
Article in English | MEDLINE | ID: mdl-28468955

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hypersensitivity/immunology , Nuclear Transfer Techniques , Receptors, Antigen, T-Cell/genetics , Alleles , Animals , Antigens/administration & dosage , Antigens/immunology , Cloning, Organism , Disease Models, Animal , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology
20.
Sci Rep ; 6: 21472, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26892171

ABSTRACT

In contrast to cell cultures, particularly to cell lines, tissues or organs removed from the body cannot be maintained for long in any culture conditions. Although it is apparent that in vivo regional homeostasis is facilitated by the microvascular system, mimicking such a system ex vivo is difficult and has not been proved effective. Using the culture system of mouse spermatogenesis, we addressed this issue and devised a simple microfluidic device in which a porous membrane separates a tissue from the flowing medium, conceptually imitating the in vivo relationship between the microvascular flow and surrounding tissue. Testis tissues cultured in this device successfully maintained spermatogenesis for 6 months. The produced sperm were functional to generate healthy offspring with micro-insemination. In addition, the tissue kept producing testosterone and responded to stimulation by luteinizing hormone. These data suggest that the microfluidic device successfully created in vivo-like conditions, in which testis tissue maintained its physiologic functions and homeostasis. The present model of the device, therefore, would provide a valuable foundation of future improvement of culture conditions for various tissues and organs, and revolutionize the organ culture method as a whole.


Subject(s)
Lab-On-A-Chip Devices , Microfluidic Analytical Techniques , Spermatogenesis , Spermatozoa/cytology , Testis/cytology , Testis/physiology , Tissue Culture Techniques , Animals , Male , Mice , Testosterone/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...