Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Takafusa Hikichi is active.

Publication


Featured researches published by Takafusa Hikichi.


Stem Cells | 2006

Equivalency of Nuclear Transfer‐Derived Embryonic Stem Cells to Those Derived from Fertilized Mouse Blastocysts

Sayaka Wakayama; Martin Jakt; Masako Suzuki; Ryoko Araki; Takafusa Hikichi; Satoshi Kishigami; Hiroshi Ohta; Nguyen Van Thuan; Eiji Mizutani; Yuko Sakaide; Sho Senda; Satoshi Tanaka; Mitsuhiro Okada; Masashi Miyake; Masumi Abe; Shin-Ichi Nishikawa; Kunio Shiota; Teruhiko Wakayama

Therapeutic cloning, whereby nuclear transfer (NT) is used to generate embryonic stem cells (ESCs) from blastocysts, has been demonstrated successfully in mice and cattle. However, if NT‐ESCs have abnormalities, such as those associated with the offspring produced by reproductive cloning, their scientific and medical utilities might prove limited. To evaluate the characteristics of NT‐ESCs, we established more than 150 NT‐ESC lines from adult somatic cells of several mouse strains. Here, we show that these NT‐ESCs were able to differentiate into all functional embryonic tissues in vivo. Moreover, they were identical to blastocyst‐derived ESCs in terms of their expression of pluripotency markers in the presence of tissue‐dependent differentially DNA methylated regions, in DNA microarray profiles, and in high‐coverage gene expression profiling. Importantly, the NT procedure did not cause irreversible damage to the nuclei. These similarities of NT‐ESCs and ESCs indicate that murine therapeutic cloning by somatic cell NT can provide a reliable model for preclinical stem cell research.


Reproduction | 2009

The histone deacetylase inhibitor scriptaid enhances nascent mRNA production and rescues full-term development in cloned inbred mice

Nguyen Van Thuan; Hong-Thuy Bui; Jin-Hoi Kim; Takafusa Hikichi; Sayaka Wakayama; Satoshi Kishigami; Eiji Mizutani; Teruhiko Wakayama

Since the birth of Cumulina, the first mouse clone produced by somatic cell nuclear transfer (SCNT), the success rate of cloning in mice has been extremely low compared with other species and most of the inbred mouse strains have never been cloned. Recently, our laboratory has found that treatment of SCNT mouse embryos with trichostatin A, a histone deacetylase inhibitor (HDACi), improved the full-term development of B6D2F1 mouse clones significantly. However, this was not effective for the inbred strains. Here, we show for the first time that by treating SCNT embryos with another HDACi, scriptaid, all the important inbred mouse strains can be cloned, such as C57BL/6, C3H/He, DBA/2, and 129/Sv. Moreover, the success of somatic nuclear reprogramming and cloning efficiency via nuclear transfer technique is clearly linked to the competent de novo synthesis of nascent mRNA in cloned mouse embryos.


Biology of Reproduction | 2005

Establishment of Male and Female Nuclear Transfer Embryonic Stem Cell Lines from Different Mouse Strains and Tissues

Sayaka Wakayama; Hiroshi Ohta; Satoshi Kishigami; Nguyen Van Thuan; Takafusa Hikichi; Eiji Mizutani; Masashi Miyake; Teruhiko Wakayama

Abstract Nuclear transfer can be used to generate embryonic stem cell lines from somatic cells, and these have great potential in regenerative medicine. However, it is still unclear whether any individual or cell type can be used to generate such lines. Here, we tested seven different male and female mouse genotypes and three cell types as sources of nuclei to determine the efficiency of establishing nuclear transfer embryonic stem cell lines. Lines were successfully established from all sources. Cumulus cell nuclei from F1 mouse genotypes showed a significantly higher cumulative establishment rate from reconstructed oocytes than from other cells; however, there were no genotype differences in success rates from cloned blastocysts. Thus, the overall success depends on preimplantation development, and, once embryos have reached the blastocyst stage, the genotype differences disappear. All mouse genotypes that were tested demonstrated at least one cell line that subsequently contributed to germline transmission in chimeric mice, so these cell lines clearly possess the same potential as embryonic stem cells derived from fertilized embryos. Thus, nuclear transfer embryonic stem cells can be generated relatively easily from a variety of inbred mouse genotypes and cell types of both sexes, even though it may be more difficult to generate clones directly.


Nature Protocols | 2006

Production of cloned mice by somatic cellnuclear transfer

Satoshi Kishigami; Sayaka Wakayama; Nguyen Van Thuan; Hiroshi Ohta; Eiji Mizutani; Takafusa Hikichi; Hong-Thuy Bui; Sebastian T. Balbach; Atsuo Ogura; Michele Boiani; Teruhiko Wakayama

Although it has now been 10 years since the first cloned mammals were generated from somatic cells using nuclear transfer (NT), the success rate for producing live offspring by cloning remains <5%. Nevertheless, the techniques have potential as important tools for future research in basic biology. We have been able to develop a stable NT method in the mouse, in which donor nuclei are directly injected into the oocyte using a piezo-actuated micromanipulator. Although manipulation of the piezo unit is complex, once mastered it is of great help not only in NT experiments but also in almost all other forms of micromanipulation. In addition to this technique, embryonic stem (ES) cell lines established from somatic cell nuclei by NT can be generated relatively easily from a variety of mouse genotypes and cell types. Such NT-ES cells can be used not only for experimental models of human therapeutic cloning but also as a backup of the donor cells genome. Our most recent protocols for mouse cloning, as described here, will allow the production of cloned mice in ≥3 months.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Production of healthy cloned mice from bodies frozen at −20°C for 16 years

Sayaka Wakayama; Hiroshi Ohta; Takafusa Hikichi; Eiji Mizutani; Takamasa Iwaki; Osami Kanagawa; Teruhiko Wakayama

Cloning animals by nuclear transfer provides an opportunity to preserve endangered mammalian species. However, it has been suggested that the “resurrection” of frozen extinct species (such as the woolly mammoth) is impracticable, as no live cells are available, and the genomic material that remains is inevitably degraded. Here we report production of cloned mice from bodies kept frozen at −20 °C for up to 16 years without any cryoprotection. As all of the cells were ruptured after thawing, we used a modified cloning method and examined nuclei from several organs for use in nuclear transfer attempts. Using brain nuclei as nuclear donors, we established embryonic stem cell lines from the cloned embryos. Healthy cloned mice were then produced from these nuclear transferred embryonic stem cells by serial nuclear transfer. Thus, nuclear transfer techniques could be used to “resurrect” animals or maintain valuable genomic stocks from tissues frozen for prolonged periods without any cryopreservation.


Stem Cells | 2007

Efficient establishment of mouse embryonic stem cell lines from single blastomeres and polar bodies.

Sayaka Wakayama; Takafusa Hikichi; Rinako Suetsugu; Yuko Sakaide; Hong-Thuy Bui; Eiji Mizutani; Teruhiko Wakayama

Recently, ES cell lines were established from single blastomeres taken from eight‐cell embryos in mice and humans with success rates of 4% and 2%, respectively, which suggests that the method could be used in regenerative medicine to reduce ethical concerns over harm to embryos. However, those studies used other ES cells as supporting cells. Here, we report a simple and highly efficient method of establishing mouse ES cell lines from single blastomeres, in which single blastomeres are simply plated onto a feeder layer of mouse embryonic fibroblasts with modified ES cell medium. A total of 112 ES cell lines were established from two‐cell (establishment rate, 50%–69%), early four‐cell (28%–40%), late four‐cell (22%), and eight‐cell (14%–16%) stage embryos. We also successfully established 18 parthenogenetic ES cell lines from first (36%–40%) and second polar bodies (33%), the nuclei of which were reconstructed to embryos by nuclear transfer. Most cell lines examined maintained normal karyotypes and expressed markers of pluripotency, including germline transmission in chimeric mice. Our results suggest that the single cells of all early‐stage embryos or polar bodies have the potential to be converted into ES cells without any special treatment.


Genes to Cells | 2001

Involvement of RNase G in in vivo mRNA metabolism in Escherichia coli

Genryou Umitsuki; Masaaki Wachi; Ayako Takada; Takafusa Hikichi; Kazuo Nagai

Escherichia coli rng gene (previously called cafA) encodes a novel RNase, named RNase G, which is involved in the 5′ end‐processing of 16S rRNA. In rng mutant cells, a precursor form of 16S rRNA, 16.3S rRNA, is accumulated. Here we report a role of RNase G in the in vivo mRNA metabolism.


Reproduction | 2007

Regulation of chromatin and chromosome morphology by histone H3 modifications in pig oocytes

Hong-Thuy Bui; Nguyen Van Thuan; Satoshi Kishigami; Sayaka Wakayama; Takafusa Hikichi; Hiroshi Ohta; Eiji Mizutani; Emi Yamaoka; Teruhiko Wakayama; Takashi Miyano

Oocyte growth, maturation, and activation are complex processes that include transcription, heterochromatin formation, chromosome condensation and decondensation, two consecutive chromosome separations, and genomic imprinting. The objective of this study was to investigate changes in histone H3 modifications in relation to chromatin/chromosome morphology in pig oocytes during their growth, maturation, and activation. During the growth phase, histone H3 was acetylated at lysines 9, 14, and 18 (K9, K14, and K18), and became methylated at K9 when the follicles developed to the antral stage (oocyte diameter, 90 mum). When the fully grown oocytes (diameter, 120 mum) started their maturation, histone H3 became phosphorylated at serine 28 (S28) and then at S10, and deacetylated at K9, K14, and K18 as the chromosomes condensed. After the electroactivation of mature oocytes, histone H3 was reacetylated and dephosphorylated concomitant with the decondensation of the chromosomes. Histone H3 kinase activity increased over a similar time course to that of the phosphorylation of histone H3-S28 during oocyte maturation, and this activity decreased as histone H3-S10 and H3-S28 began to be dephosphorylated after the activation of the mature oocytes. These results suggest that the chromatin morphology of pig oocytes is regulated by the acetylation/deacetylation and the phosphorylation/dephosphorylation of histone H3, and the phosphorylation of histone H3 is the key event in meiotic chromosome condensation in oocytes. The inhibition of histone deacetylase with trichostatin A (TSA) inhibited the deacetylation and phosphorylation of histone H3, and chromosome condensation. Therefore, the deacetylation of histone H3 is thought to be required for its phosphorylation in meiosis. Although histone H3 acetylation and phosphorylation were reversible, the histone methylation that was established during the oocyte growth phase was stable throughout the course of oocyte maturation and activation.


Human Molecular Genetics | 2009

Paternal deletion of Meg1/Grb10 DMR causes maternalization of the Meg1/Grb10 cluster in mouse proximal Chromosome 11 leading to severe pre- and postnatal growth retardation

Hirosuke Shiura; Kenji Nakamura; Takafusa Hikichi; Toshiaki Hino; Kanako Oda; Rika Suzuki-Migishima; Takashi Kohda; Tomoko Kaneko-Ishino; Fumitoshi Ishino

Mice with maternal duplication of proximal Chromosome 11 (MatDp(prox11)), where Meg1/Grb10 is located, exhibit pre- and postnatal growth retardation. To elucidate the responsible imprinted gene for the growth abnormality, we examined the precise structure and regulatory mechanism of this imprinted region and generated novel model mice mimicking the pattern of imprinted gene expression observed in the MatDp(prox11) by deleting differentially methylated region of Meg1/Grb10 (Meg1-DMR). It was found that Cobl and Ddc, the neighboring genes of Meg1/Grb10, also comprise the imprinted region. We also found that the mouse-specific repeat sequence consisting of several CTCF-binding motifs in the Meg1-DMR functions as a silencer, suggesting that the Meg1/Grb10 imprinted region adopted a different regulatory mechanism from the H19/Igf2 region. Paternal deletion of the Meg1-DMR (+/DeltaDMR) caused both upregulation of the maternally expressed Meg1/Grb10 Type I in the whole body and Cobl in the yolk sac and loss of paternally expressed Meg1/Grb10 Type II and Ddc in the neonatal brain and heart, respectively, demonstrating maternalization of the entire Meg1/Grb10 imprinted region. We confirmed that the +/DeltaDMR mice exhibited the same growth abnormalities as the MatDp(prox11) mice. Fetal and neonatal growth was very sensitive to the expression level of Meg1/Grb10 Type I, indicating that the 2-fold increment of the Meg1/Grb10 Type I is one of the major causes of the growth retardation observed in the MatDp(prox11) and +/DeltaDMR mice. This suggests that the corresponding human GRB10 Type I plays an important role in the etiology of Silver-Russell syndrome caused by partial trisomy of 7p11-p13.


FEBS Letters | 2006

Normal specification of the extraembryonic lineage after somatic nuclear transfer

Satoshi Kishigami; Takafusa Hikichi; Nguyen Van Thuan; Hiroshi Ohta; Sayaka Wakayama; Hong-Thuy Bui; Eiji Mizutani; Teruhiko Wakayama

To examine the establishment and maintenance of trophectoderm (TE) lineage in somatic cloned blastocysts, the expression of Cdx2, a key molecule for specification of TE fate, was immunohistochemically examined simultaneously with Oct4 expression. Cloned mouse embryos were made by nuclear transfer using cumulus cells, tail‐tip fibroblasts, and embryonic stem cells. After 96 h of culture, the rates of Oct4‐expressing blastocysts were as low as 50% and 60% for cumulus and fibroblast clones, respectively. However, regardless of Oct4 expression, the majority of those cloned blastocysts (>90%) normally expressed Cdx2. Thus, even though somatic cloned embryos have reduced potential to produce the inner cell mass lineage, the TE lineage can be established and maintained.

Collaboration


Dive into the Takafusa Hikichi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Koji Kitazawa

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge