Updated on 2024/04/11

写真a

 
Ayano Kawaguchi
 
Organization
Faculty of Medicine, Dentistry and Pharmaceutical Sciences Professor
Position
Professor
Profile

頑強な脳発生に貢献する、神経前駆細胞の運命決定の仕組みに興味を持って研究を行っています。

External link

Degree

  • 博士(医学) ( 大阪大学 )

Research Interests

  • 発生

  • 神経前駆細胞

  • 神経発生

  • 非対称分裂

  • 分化

  • 神経幹細胞

  • 包括脳ネットワーク

Research Areas

  • Life Science / Neuroscience-general

  • Life Science / Anatomy and histopathology of nervous system

  • Life Science / Anatomy

  • Life Science / Developmental biology

Education

  • Osaka University   大学院医学系研究科   博士課程

    1998.4 - 2002.3

      More details

  • Osaka University   医学部   医学科

    1989.4 - 1995.3

      More details

    Country: Japan

    researchmap

Research History

  • 大阪大学医学系研究科 委託講師(兼任)

    2024.4 - 2025.3

      More details

  • Nagoya University   School of Medicine

    2022.12

      More details

  • Okayama University   学術研究院医歯薬学域 人体構成学分野   Professor

    2022.6

      More details

  • Nagoya University   Graduate School of Medicine   Associate Professor

    2008 - 2022

      More details

  • RIKEN   非対称細胞分裂研究グループ   Researcher

    2002 - 2008

      More details

  • Japan Society for Promotion of Science

    2001 - 2002

      More details

▼display all

Professional Memberships

Committee Memberships

  • 日本解剖学会   代議員  

    2022.6   

      More details

 

Papers

  • Calcium signals tune AMPK activity and mitochondrial homeostasis in dendrites of developing neurons Reviewed

    Akane Hatsuda, Junko Kurisu, Kazuto Fujishima, Ayano Kawaguchi, Nobuhiko Ohno, Mineko Kengaku

    Development   150 ( 21 )   2023.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    DOI: 10.1242/dev.201930

    researchmap

  • Advanced Techniques Using In Vivo Electroporation to Study the Molecular Mechanisms of Cerebral Development Disorders Invited Reviewed

    Chen Yang, Atsunori Shitamukai, Shucai Yang, Ayano Kawaguchi

    International Journal of Molecular Sciences   24 ( 18 )   14128 - 14128   2023.9

     More details

    Authorship:Last author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:MDPI AG  

    The mammalian cerebral cortex undergoes a strictly regulated developmental process. Detailed in situ visualizations, imaging of these dynamic processes, and in vivo functional gene studies significantly enhance our understanding of brain development and related disorders. This review introduces basic techniques and recent advancements in in vivo electroporation for investigating the molecular mechanisms underlying cerebral diseases. In utero electroporation (IUE) is extensively used to visualize and modify these processes, including the forced expression of pathological mutants in human diseases; thus, this method can be used to establish animal disease models. The advent of advanced techniques, such as genome editing, including de novo knockout, knock-in, epigenetic editing, and spatiotemporal gene regulation, has further expanded our list of investigative tools. These tools include the iON expression switch for the precise control of timing and copy numbers of exogenous genes and TEMPO for investigating the temporal effects of genes. We also introduce the iGONAD method, an improved genome editing via oviductal nucleic acid delivery approach, as a novel genome-editing technique that has accelerated brain development exploration. These advanced in vivo electroporation methods are expected to provide valuable insights into pathological conditions associated with human brain disorders.

    DOI: 10.3390/ijms241814128

    researchmap

  • CD206+ macrophages transventricularly infiltrate the early embryonic cerebral wall to differentiate into microglia Reviewed

    Yuki Hattori, Daisuke Kato, Futoshi Murayama, Sota Koike, Hisa Asai, Ayato Yamasaki, Yu Naito, Ayano Kawaguchi, Hiroyuki Konishi, Marco Prinz, Takahiro Masuda, Hiroaki Wake, Takaki Miyata

    Cell Reports   42 ( 2 )   112092 - 112092   2023.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Elsevier BV  

    DOI: 10.1016/j.celrep.2023.112092

    researchmap

  • Neuronal delamination and outer radial glia generation in neocortical development Reviewed

    Kawaguchi A

    Frontiers Cell and Developmental Biology   8   623573   2021.2

     More details

    Authorship:Last author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)  

    researchmap

  • Transient microglial absence assists postmigratory cortical neurons in proper differentiation. Reviewed International journal

    Yuki Hattori, Yu Naito, Yoji Tsugawa, Shigenori Nonaka, Hiroaki Wake, Takashi Nagasawa, Ayano Kawaguchi, Takaki Miyata

    Nature communications   11 ( 1 )   1631 - 1631   2020.4

     More details

    Language:English  

    In the developing cortex, postmigratory neurons accumulate in the cortical plate (CP) to properly differentiate consolidating subtype identities. Microglia, despite their extensive surveying activity, temporarily disappear from the midembryonic CP. However, the mechanism and significance of this absence are unknown. Here, we show that microglia bidirectionally migrate via attraction by CXCL12 released from the meninges and subventricular zone and thereby exit the midembryonic CP. Upon nonphysiological excessive exposure to microglia in vivo or in vitro, young postmigratory and in vitro-grown CP neurons showed abnormal differentiation with disturbed expression of the subtype-associated transcription factors and genes implicated in functional neuronal maturation. Notably, this effect is primarily attributed to interleukin 6 and type I interferon secreted by microglia. These results suggest that "sanctuarization" from microglia in the midembryonic CP is required for neurons to appropriately fine-tune the expression of molecules needed for proper differentiation, thus securing the establishment of functional cortical circuit.

    DOI: 10.1038/s41467-020-15409-3

    PubMed

    researchmap

  • Lzts1 controls both neuronal delamination and outer radial glial-like cell generation during mammalian cerebral development. Reviewed International journal

    Kawaue T, Shitamukai A, Nagasaka A, Tsunekawa Y, Shinoda T, Saito K, Terada R, Bilgic M, Miyata T, Matsuzaki F, Kawaguchi A

    Nature Communications   10 ( 1 )   2780 - 2780   2019.6

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)  

    In the developing central nervous system, cell departure from the apical surface is the initial and fundamental step to form the 3D, organized architecture. Both delamination of differentiating cells and repositioning of progenitors to generate outer radial glial cells (oRGs) contribute to mammalian neocortical expansion; however, a comprehensive understanding of their mechanisms is lacking. Here, we demonstrate that Lzts1, a molecule associated with microtubule components, promotes both cell departure events. In neuronally committed cells, Lzts1 functions in apical delamination by altering apical junctional organization. In apical RGs (aRGs), Lzts1 expression is variable, depending on Hes1 expression levels. According to its differential levels, Lzts1 induces diverse RG behaviors: planar division, oblique divisions of aRGs that generate oRGs, and their mitotic somal translocation. Loss-of-function of lzts1 impairs all these cell departure processes. Thus, Lzts1 functions as a master modulator of cellular dynamics, contributing to increasing complexity of the cerebral architecture during evolution.

    DOI: 10.1038/s41467-019-10730-y

    PubMed

    researchmap

  • Temporal patterning of neocortical progenitor cells: how do they know the right time? Reviewed

    Kawaguchi A

    Neuroscience Research   138   3 - 11   2019.1

     More details

    Authorship:Corresponding author  

    DOI: 10.1016/j.neures.2018.09.004

    researchmap

  • Neural Progenitor Cells Undergoing Yap/Tead-Mediated Enhanced Self-Renewal Form Heterotopias More Easily in the Diencephalon than in the Telencephalon Reviewed

    Kanako Saito, Ryotaro Kawasoe, Hiroshi Sasaki, Ayano Kawaguchi, Takaki Miyata

    Neurochemical Research   43 ( 1 )   171 - 180   2018.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Springer New York LLC  

    Spatiotemporally ordered production of cells is essential for brain development. Normally, most undifferentiated neural progenitor cells (NPCs) face the apical (ventricular) surface of embryonic brain walls. Pathological detachment of NPCs from the apical surface and their invasion of outer neuronal territories, i.e., formation of NPC heterotopias, can disrupt the overall structure of the brain. Although NPC heterotopias have previously been observed in a variety of experimental contexts, the underlying mechanisms remain largely unknown. Yes-associated protein 1 (Yap1) and the TEA domain (Tead) proteins, which act downstream of Hippo signaling, enhance the stem-like characteristics of NPCs. Elevated expression of Yap1 or Tead in the neural tube (future spinal cord) induces massive NPC heterotopias, but Yap/Tead-induced expansion of NPCs in the developing brain has not been previously reported to produce NPC heterotopias. To determine whether NPC heterotopias occur in a regionally characteristic manner, we introduced the Yap1-S112A or Tead-VP16 into NPCs of the telencephalon and diencephalon, two neighboring but distinct forebrain regions, of embryonic day 10 mice by in utero electroporation, and compared NPC heterotopia formation. Although NPCs in both regions exhibited enhanced stem-like behaviors, heterotopias were larger and more frequent in the diencephalon than in the telencephalon. This result, the first example of Yap/Tead-induced NPC heterotopia in the forebrain, reveals that Yap/Tead-induced NPC heterotopia is not specific to the neural tube, and also suggests that this phenomenon depends on regional factors such as the three-dimensional geometry and assembly of these cells.

    DOI: 10.1007/s11064-017-2390-x

    Scopus

    PubMed

    researchmap

  • Division modes and physical asymmetry in cerebral cortex progenitors Reviewed

    Delphine Delaunay, Ayano Kawaguchi, Colette Dehay, Fumio Matsuzaki

    CURRENT OPINION IN NEUROBIOLOGY   42   75 - 83   2017.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:CURRENT BIOLOGY LTD  

    Neural stem cells go through a sequence of timely regulated gene expression and pattern of division mode to generate diverse neurons during brain development. During vertebrate cerebral cortex development, neural stem cells begin with proliferative symmetric divisions, subsequently undergo neurogenic asymmetric divisions, and finally gliogenic divisions. In this review, we explore the relationship between stem cell versus neural fate specification and the division mode. Specifically, we discuss recent findings on the mechanisms of asymmetric divisions, division mode, and developmental progression of neural progenitor identity.

    DOI: 10.1016/j.conb.2016.11.009

    Web of Science

    PubMed

    researchmap

  • Differences in the Mechanical Properties of the Developing Cerebral Cortical Proliferative Zone between Mice and Ferrets at both the Tissue and Single-Cell Levels. Reviewed

    Nagasaka A, Shinoda T, Kawaue T, Suzuki M, Nagayama K, Matsumoto T, Ueno N, Kawaguchi A, Miyata T

    Frontiers in cell and developmental biology   4   139   2016.11

     More details

    Publishing type:Research paper (scientific journal)   Publisher:Frontiers Media SA  

    DOI: 10.3389/fcell.2016.00139

    PubMed

    researchmap

  • Cell-cycle-independent transitions in temporal identity of mammalian neural progenitor cells Reviewed

    Mayumi Okamoto, Takaki Miyata, Daijiro Konno, Hiroki R. Ueda, Takeya Kasukawa, Mitsuhiro Hashimoto, Fumio Matsuzaki, Ayano Kawaguchi

    NATURE COMMUNICATIONS   7   11349   2016.4

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:NATURE PUBLISHING GROUP  

    During cerebral development, many types of neurons are sequentially generated by self-renewing progenitor cells called apical progenitors (APs). Temporal changes in AP identity are thought to be responsible for neuronal diversity; however, the mechanisms underlying such changes remain largely unknown. Here we perform single-cell transcriptome analysis of individual progenitors at different developmental stages, and identify a subset of genes whose expression changes over time but is independent of differentiation status. Surprisingly, the pattern of changes in the expression of such temporal-axis genes in APs is unaffected by cell-cycle arrest. Consistent with this, transient cell-cycle arrest of APs in vivo does not prevent descendant neurons from acquiring their correct laminar fates. Analysis of cultured APs reveals that transitions in AP gene expression are driven by both cell-intrinsic and -extrinsic mechanisms. These results suggest that the timing mechanisms controlling AP temporal identity function independently of cell-cycle progression and Notch activation mode.

    DOI: 10.1038/ncomms11349

    Web of Science

    PubMed

    researchmap

  • Cell cycle-arrested cells know the right time Reviewed

    Ayano Kawaguchi, Fumio Matsuzaki

    CELL CYCLE   15 ( 20 )   2683 - 2684   2016

     More details

    Authorship:Corresponding author   Language:English   Publisher:TAYLOR & FRANCIS INC  

    DOI: 10.1080/15384101.2016.1204857

    Web of Science

    PubMed

    researchmap

  • Interkinetic nuclear migration generates and opposes ventricular-zone crowding: insight into tissue mechanics Reviewed

    Takaki Miyata, Mayumi Okamoto, Tomoyasu Shinoda, Ayano Kawaguchi

    FRONTIERS IN CELLULAR NEUROSCIENCE   8   473   2015.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:FRONTIERS MEDIA SA  

    The neuroepithelium (NE) or ventricular zone (VZ), from which multiple types of brain cells arise, is pseudostratified. In the NE/VZ, neural progenitor cells are elongated along the apicobasal axis, and their nuclei assume different apicobasal positions. These nuclei move in a cell cycledependent manner, i.e., apicalward during G2 phase and basalward during G1 phase, a process called interkinetic nuclear migration (INM). This review will summarize and discuss several topics: the nature of the INM exhibited by neural progenitor cells, the mechanical difficulties associated with INM in the developing cerebral cortex, the community-level mechanisms underlying collective and efficient INM, the impact on overall brain formation when NE/VZ is overcrowded due to loss of INM, and whether and how neural progenitor INM varies among mammalian species. These discussions will be based on recent findings obtained in live, three-dimensional specimens using quantitative and mechanical approaches. Experiments in which overcrowding was induced in mouse neocortical NE/VZ, as well as comparisons of neocortical INM between mice and ferrets, have revealed that the behavior of NE/VZ cells can be affected by cellular densification. A consideration of the physical aspects in the NE/VZ and the mechanical difficulties associated with high-degree pseudostratification (PS) is important for achieving a better understanding of neocortical development and evolution.

    DOI: 10.3389/fncel.2014.00473

    Web of Science

    PubMed

    researchmap

  • Neurogenin2-d4Venus and Gadd45g-d4Venus transgenic mice: Visualizing mitotic and migratory behaviors of cells committed to the neuronal lineage in the developing mammalian brain Reviewed

    Takumi Kawaue, Ken Sagou, Hiroshi Kiyonari, Kumiko Ota, Mayumi Okamoto, Tomoyasu Shinoda, Ayano Kawaguchi, Takaki Miyata

    DEVELOPMENT GROWTH & DIFFERENTIATION   56 ( 4 )   293 - 304   2014.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    To achieve highly sensitive and comprehensive assessment of the morphology and dynamics of cells committed to the neuronal lineage in mammalian brain primordia, we generated two transgenic mouse lines expressing a destabilized (d4) Venus controlled by regulatory elements of the Neurogenin2 (Neurog2) or Gadd45g gene. In mid-embryonic neocortical walls, expression of Neurog2-d4Venus mostly overlapped with that of Neurog2 protein, with a slightly (1h) delayed onset. Although Neurog2-d4Venus and Gadd45g-d4Venus mice exhibited very similar labeling patterns in the ventricular zone (VZ), in Gadd45g-d4Venus mice cells could be visualized in more basal areas containing fully differentiated neurons, where Neurog2-d4Venus fluorescence was absent. Time-lapse monitoring revealed that most d4Venus(+) cells in the VZ had processes extending to the apical surface; many of these cells eventually retracted their apical process and migrated basally to the subventricular zone, where neurons, as well as the intermediate neurogenic progenitors that undergo terminal neuron-producing division, could be live-monitored by d4Venus fluorescence. Some d4Venus(+) VZ cells instead underwent nuclear migration to the apical surface, where they divided to generate two d4Venus(+) daughter cells, suggesting that the symmetric terminal division that gives rise to neuron pairs at the apical surface can be reliably live-monitored. Similar lineage-committed cells were observed in other developing neural regions including retina, spinal cord, and cerebellum, as well as in regions of the peripheral nervous system such as dorsal root ganglia. These mouse lines will be useful for elucidating the cellular and molecular mechanisms underlying development of the mammalian nervous system.

    DOI: 10.1111/dgd.12131

    Web of Science

    PubMed

    researchmap

  • TAG-1-assisted progenitor elongation streamlines nuclear migration to optimize subapical crowding Reviewed

    Mayumi Okamoto, Takashi Namba, Tomoyasu Shinoda, Takefumi Kondo, Tadashi Watanabe, Yasuhiro Inoue, Kosei Takeuchi, Yukiko Enomoto, Kumiko Ota, Kanako Oda, Yoshino Wada, Ken Sagou, Kanako Saito, Akira Sakakibara, Ayano Kawaguchi, Kazunori Nakajima, Taiji Adachi, Toshihiko Fujimori, Masahiro Ueda, Shigeo Hayashi, Kozo Kaibuchi, Takaki Miyata

    NATURE NEUROSCIENCE   16 ( 11 )   1556 - 1566   2013.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:NATURE PUBLISHING GROUP  

    Neural progenitors exhibit cell cycle-dependent interkinetic nuclear migration (INM) along the apicobasal axis. Despite recent advances in understanding its underlying molecular mechanisms, the processes to which INM contributes mechanically and the regulation of INM by the apicobasally elongated morphology of progenitors remain unclear. We found that knockdown of the cell-surface molecule TAG-1 resulted in retraction of neocortical progenitors' basal processes. Highly shortened stem-like progenitors failed to undergo basalward INM and became overcrowded in the periventricular (subapical) space. Surprisingly, the overcrowded progenitors left the apical surface and migrated into basal neuronal territories. These observations, together with the results of in toto imaging and physical tests, suggest that progenitors may sense and respond to excessive mechanical stress. Although, unexpectedly, the heterotopic progenitors remained stem-like and continued to sequentially produce neurons until the late embryonic period, histogenesis was severely disrupted. Thus, INM is essential for preventing overcrowding of nuclei and their somata, thereby ensuring normal brain histogenesis.

    DOI: 10.1038/nn.3525

    Web of Science

    PubMed

    researchmap

  • Lunatic fringe potentiates Notch signaling in the developing brain Reviewed

    Tomoaki M. Kato, Ayano Kawaguchi, Yoichi Kosodo, Hitoshi Niwa, Fumio Matsuzaki

    MOLECULAR AND CELLULAR NEUROSCIENCE   45 ( 1 )   12 - 25   2010.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Notch signaling is essential for the self-renewal of mammalian neural progenitor cells. A variety of mechanisms modulate Notch signaling to balance the self-renewal and differentiation of progenitor cells. Fringe is a major Notch regulator and promotes or suppresses Notch signaling, depending on the Notch ligands. In the developing brain. Lunatic fringe (Lfng) is expressed in self-renewing progenitors, but its roles are unknown. In this study, in vivo mosaic analyses using in utero electroporation were developed to investigate the roles of Lfng in neural progenitor cells. We found that Lfng potentiates Notch signaling cell-autonomously. Its depletion did not affect the balance between neuronally committed cells and self-renewing progenitors, however, irrespective of the cell density of Lfng-depleted cells, and caused no obvious defects in brain development. In vivo overexpression experiments with Notch ligands suggest that Lfng strongly augments Notch signaling mediated by Delta-like 1 but not Jagged 1. (C) 2010 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.mcn.2010.05.004

    Web of Science

    PubMed

    researchmap

  • Migration, early axonogenesis, and Reelin-dependent layer-forming behavior of early/posterior-born Purkinje cells in the developing mouse lateral cerebellum Reviewed

    Takaki Miyata, Yuichi Ono, Mayumi Okamoto, Makoto Masaoka, Akira Sakakibara, Ayano Kawaguchi, Mitsuhiro Hashimoto, Masaharu Ogawa

    NEURAL DEVELOPMENT   5   23   2010.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:BIOMED CENTRAL LTD  

    Background: Cerebellar corticogenesis begins with the assembly of Purkinje cells into the Purkinje plate (PP) by embryonic day 14.5 (E14.5) in mice. Although the dependence of PP formation on the secreted protein Reelin is well known and a prevailing model suggests that Purkinje cells migrate along the 'radial glial' fibers connecting the ventricular and pial surfaces, it is not clear how Purkinje cells behave in response to Reelin to initiate the PP. Furthermore, it is not known what nascent Purkinje cells look like in vivo. When and how Purkinje cells start axonogenesis must also be elucidated.
    Results: We show that Purkinje cells generated on E10.5 in the posterior periventricular region of the lateral cerebellum migrate tangentially, after only transiently migrating radially, towards the anterior, exhibiting an elongated morphology consistent with axonogenesis at E12.5. After their somata reach the outer/dorsal region by E13.5, they change 'posture' by E14.5 through remodeling of non-axon (dendrite-like) processes and a switchback-like mode of somal movement towards a superficial Reelin-rich zone, while their axon-like fibers remain relatively deep, which demarcates the somata-packed portion as a plate. In reeler cerebella, the early born posterior lateral Purkinje cells are initially normal during migration with anteriorly extended axon-like fibers until E13.5, but then fail to form the PP due to lack of the posture-change step.
    Conclusions: Previously unknown behaviors are revealed for a subset of Purkinje cells born early in the posteior lateral cerebellum: tangential migration; early axonogenesis; and Reelin-dependent reorientation initiating PP formation. This study provides a solid basis for further elucidation of Reelin's function and the mechanisms underlying the cerebellar corticogenesis, and will contribute to the understanding of how polarization of individual cells drives overall brain morphogenesis.

    DOI: 10.1186/1749-8104-5-23

    Web of Science

    PubMed

    researchmap

  • Mechanisms that regulate the number of neurons during mouse neocortical development Reviewed

    Takaki Miyata, Daichi Kawaguchi, Ayano Kawaguchi, Yukiko Gotoh

    CURRENT OPINION IN NEUROBIOLOGY   20 ( 1 )   22 - 28   2010.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:CURRENT BIOLOGY LTD  

    Cortical development progresses through an early phase of progenitor expansion, a middle phase of neurogenesis, and a final phase of gliogenesis. During the middle phase, the neurogenic phase, the neocortical primordium balances the production of neurons against the maintenance of neural precursor cells (NPCs). The final number of neurons is determined by the duration of the neurogenic phase, the rate of NPC division, and the mode of NPC division, that is, whether a division gives rise to two NPCs, one NPC and one cell committed to the neuronal lineage, or two committed cells. We discuss here recent advances in understanding these key aspects that are fundamental for normal brain development.

    DOI: 10.1016/j.conb.2010.01.001

    Web of Science

    PubMed

    researchmap

  • Single-cell gene profiling defines differential progenitor subclasses in mammalian neurogenesis Reviewed

    Ayano Kawaguchi, Tomoko Ikawa, Takeya Kasukawa, Hiroki R. Ueda, Kazuki Kurimoto, Mitinori Saitou, Fumio Matsuzaki

    DEVELOPMENT   135 ( 18 )   3113 - 3124   2008.9

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:COMPANY OF BIOLOGISTS LTD  

    Cellular diversity of the brain is largely attributed to the spatial and temporal heterogeneity of progenitor cells. In mammalian cerebral development, it has been difficult to determine how heterogeneous the neural progenitor cells are, owing to dynamic changes in their nuclear position and gene expression. To address this issue, we systematically analyzed the cDNA profiles of a large number of single progenitor cells at the mid-embryonic stage in mouse. By cluster analysis and in situ hybridization, we have identified a set of genes that distinguishes between the apical and basal progenitors. Despite their relatively homogeneous global gene expression profiles, the apical progenitors exhibit highly variable expression patterns of Notch signaling components, raising the possibility that this causes the heterogeneous division patterns of these cells. Furthermore, we successfully captured the nascent state of basal progenitor cells. These cells are generated shortly after birth from the division of the apical progenitors, and show strong expression of the major Notch ligand delta-like 1, which soon fades away as the cells migrate in the ventricular zone. We also demonstrated that attenuation of Notch signals immediately induces differentiation of apical progenitors into nascent basal progenitors. Thus, a Notch-dependent feedback loop is likely to be in operation to maintain both progenitor populations.

    DOI: 10.1242/dev.022616

    Web of Science

    PubMed

    CiNii Article

    researchmap

  • Differential expression of Pax6 and Ngn2 between pair-generated cortical neurons Reviewed

    A Kawaguchi, M Ogawa, K Saito, F Matsuzaki, H Okano, T Miyata

    JOURNAL OF NEUROSCIENCE RESEARCH   78 ( 6 )   784 - 795   2004.12

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-LISS  

    Progenitor cells that generate neuron pairs ("pair progenitor cells") are implicated in mammalian cortical development, and their division has been thought to be "symmetric." However, asymmetric growth of two sister neurons generated by the division of a pair progenitor cell would lead to more efficient generation of neuronal diversity in the cortex. To explore mechanisms by which pair progenitor cells provide neuronal diversity, we examined molecular differences between a pair of neurons generated in clonal-density culture. Time-course analysis for the acquisition of neuronal markers and the disappearance of Pax6 and Neurogenin2 (Ngn2) demonstrated that 1) these transcription factors are expressed transiently in some but not all young neurons and 2) some neuron pairs showed uneven/asymmetric expression of Pax6 (19.5%) or Ngn2 (23.8%), whereas other pairs were either symmetrically positive or negative. Asymmetric Pax6 distribution in neuron pairs was not associated with asymmetric distribution of Numb, which raises an intriguing possibility, that Pax6 asymmetry in neuron pairs is produced by an alternative mode of the cell autonomous mechanisms. Stage-dependent changes were noted in the pattern of Ngn2 retention in daughter neurons, reflecting qualitative changes in the pair progenitor population. We suggest that pair progenitor cells contribute to the generation of neuronal diversity through cell-intrinsic heterogeneity and asymmetric division. (C) 2004 Wiley-Liss, Inc.

    DOI: 10.1002/jnr.20347

    Web of Science

    PubMed

    researchmap

  • Asymmetric production of surface-dividing and non-surface-dividing cortical progenitor cells Reviewed

    T Miyata, A Kawaguchi, K Saito, M Kawano, T Muto, M Ogawa

    DEVELOPMENT   131 ( 13 )   3133 - 3145   2004.7

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:COMPANY OF BIOLOGISTS LTD  

    Mature neocortical layers all derive from the cortical plate (CP), a transient zone in the dorsal telencephalon into which young neurons are continuously delivered. To understand cytogenetic and histogenetic events that trigger the emergence of the CP, we have used a slice culture technique. Most divisions at the ventricular surface generated paired cycling daughters (P/P divisions) and the majority of the P/P divisions were asymmetric in daughter cell behavior; they frequently sent one daughter cell to a non-surface (NS) position, the subventricular zone (SVZ), within a single cell-cycle length while keeping the other mitotic daughter for division at the surface. The NS-dividing cells were mostly Hu(+) and their daughters were also Hu+, suggesting their commitment to the neuronal lineage and supply of early neurons at a position much closer to their destiny than from the ventricular surface. The release of a cycling daughter cell to SVZ was achieved by collapse of the ventricular process of the cell, followed by its NS division. Neurogenin2 (Ngn2) was immunohistochemically detected in a certain cycling population during G1 phase and was further restricted during G2-M phases to the SVZ-directed population. Its retroviral introduction converted surface divisions to NS divisions. The asymmetric P/P division may therefore contribute to efficient neuron/progenitor segregation required for CP initiation through cell cycle-dependent and lineage-restricted expression of Ngn2.

    DOI: 10.1242/dev.01173

    Web of Science

    PubMed

    CiNii Article

    researchmap

  • Morphological asymmetry in dividing retinal progenitor cells Reviewed

    K Saito, A Kawaguchi, S Kashiwagi, S Yasugi, M Ogawa, T Miyata

    DEVELOPMENT GROWTH & DIFFERENTIATION   45 ( 3 )   219 - 229   2003.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:BLACKWELL PUBLISHING ASIA  

    For the understanding of histogenetic events in the 3-D retinal neuroepithelium, direct observation of the progenitor cells and their morphological changes is required. A slice culture method has been developed by which the behavior of single progenitor cells can be monitored. Although it has been believed that each retinal progenitor cell loses its basal process while it is in M phase, it is reported here that the process is retained throughout M phase and is inherited by one daughter cell, which can be a neuron or a progenitor cell. Daughter neurons used an inherited process for neuronal translocation and positioning. In divisions that produced two mitotic daughters, both of which subsequently divided to form four granddaughter cells, only one daughter cell inherited the original basal process while the other extended a new process. Interestingly, behavioral differences were often noted between such mitotic sisters in the trajectory of interkinetic nuclear movement, cell cycle length, and the composition of the granddaughter pair. Therefore, 'symmetric' (progenitor --> progenitor + progenitor) divisions are in fact morphologically asymmetric, and the behavior of the mitotic daughters can often be asymmetric, indicating the necessity for studying possible associations between the process inheritance and the cell fate choice.

    DOI: 10.1046/j.1524-4725.2003.690.x

    Web of Science

    PubMed

    researchmap

  • Flow cytometric analysis of neural stem cells in the developing and adult mouse brain Reviewed

    A Murayama, Y Matsuzaki, A Kawaguchi, T Shimazaki, H Okano

    JOURNAL OF NEUROSCIENCE RESEARCH   69 ( 6 )   837 - 847   2002.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Despite recent progress in the neural stem cell biology, their cellular characteristics have not been described well. We investigated various characteristics of neural stem cells (NSCs) in vivo during CNS development, using FACS to identify the NSCs. We first examined stage-dependent changes in the physical parameters, using forward scatter (FSC) and side scatter (SSC) profiles, of NSCs from the developing striatum, where they appear to be active throughout the life of mammals. NSCs were divided into several fractions according to their FSC/SSC profile. With development, their number decreased in the FSChigh fractions but increased in the FSClow/SSChigh fraction, whereas NSCs were significantly concentrated in the fraction containing the largest cells (about 20 mum in diameter) at any stage, which were mostly the cells with the highest nestin-enhancer activity. Furthermore, we demonstrated that, at all stages examined, the "side population" (SP), defined as the Hoechst 33342 low/negative fraction, which is known to be a stem cell-enriched population in bone marrow, was also enriched for Notch1-positive immature neural cells (about 60%) from the developing striatum. However, these immature SP cells were not detected in the large-cell fraction, however, but were concentrated instead in the FSClow/mid fractions. FACS analysis showed that SP cells from adults were included to some extent in the CD24(low)/ PNA(low) fraction, where NSCs were greatly concentrated. Collectively, the characteristics of NSCs were not uniform and changed developmentally. (C) 2002 Wiley-Liss, Inc.

    DOI: 10.1002/jnr.10339

    Web of Science

    PubMed

    researchmap

  • Visualization of cell cycling by an improvement in slice culture methods Reviewed

    T Miyata, A Kawaguchi, K Saito, H Kuramochi, M Ogawa

    JOURNAL OF NEUROSCIENCE RESEARCH   69 ( 6 )   861 - 868   2002.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-LISS  

    Slice culture combined with the use of fluorescent dyes and/or the introduction of fluorescent protein genes provides live and three-dimensional information on cytogenetic and histogenetic events at the level of the individual cell. Using slices prepared from midembryonic mouse cerebral wall tissue upon which fine Dil crystals were placed on the pial or ventricular surface, we recently found that dividing progenitor cells do not lose their pia-connected (basal) processes and that the processes are inherited by daughter cells, including neurons (Miyata et al. [2001] Neuron 31:727-741). To understand more fully the biological significance of this inheritance process, the fate of each daughter cell should be monitored over a culture period extended long enough to allow a neuron to migrate up to the cortex or for a progenitor to proceed to the next round of division. Exposure of slices to 40%, instead of 20%, O-2 significantly improved their overall thickening, cell production, and layer formation and also provided better spatial resolution by preventing the loss of transparency that accompanies cell death. (C) 2002 Wiley-Liss, Inc.

    DOI: 10.1002/jnr.10335

    Web of Science

    PubMed

    researchmap

  • Asymmetric inheritance of radial glial fibers by cortical neurons Reviewed

    T Miyata, A Kawaguchi, H Okano, M Ogawa

    NEURON   31 ( 5 )   727 - 741   2001.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:CELL PRESS  

    Recent studies demonstrated the neuronogenic role of radial glial cells (RGCs) in the rodent. To reveal the fate of radial glial processes, we intensively monitored divisions of RGCs in Dil-labeled slices from the embryonic day 14 mouse cortex. During RGC division, each pia-connected fiber becomes thin but is neither lost nor divided; it is inherited asymmetrically by one daughter cell. In divisions that produce a neuron and a progenitor, the neuron inherits the pial fiber, also grows a thick ventricular process for several hours, and is therefore indistinguishable from the progenitor RGC. The ventricular process in the radial glial-like neuron ("radial neuron") then collapses, leading to ascent of the neuron by using the "recycled" radial fiber.

    DOI: 10.1016/S0896-6273(01)00420-2

    Web of Science

    PubMed

    CiNii Article

    researchmap

  • Direct isolation of committed neuronal progenitor cells from transgenic mice coexpressing spectrally distinct fluorescent proteins regulated by stage-specific neural promoters Reviewed

    K Sawamoto, A Yamamoto, A Kawaguchi, M Yamaguchi, K Mori, SA Goldman, H Okano

    JOURNAL OF NEUROSCIENCE RESEARCH   65 ( 3 )   220 - 227   2001.8

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-LISS  

    Many tissues arise from pluripotent stem cells through cell-type specification and maturation. In the bone marrow, primitive stem cells generate all the different types of blood cells via the sequential differentiation of increasingly committed progenitor cells. Cell-surface markers that clearly distinguish stem cells, restricted progenitors, and differentiated progeny have enabled researchers to isolate these cells and to study the regulatory mechanisms of hematopoiesis. Neuronal differentiation appears to involve similar mechanisms. However, neural progenitor cells that are restricted to a neuronal fate have not been characterized in vivo, because specific cell-surface markers are not available. We have developed an alternative strategy to identify and isolate neuronal progenitor cells based on dual-color fluorescent proteins. To identify and isolate directly progenitor cells from brain tissue without the need for either transfection or intervening cell culture, we established lines of transgenic mice bearing fluorescent transgenes regulated by neural promoters. One set of transgenic lines expressed enhanced yellow fluorescent protein (EYFP) in neuronal progenitor cells and neurons under the control of the T alpha1 alpha -tubulin promoter. Another line expressed enhanced green fluorescent protein (EGFP) in immature neural cells under the control of the enhancer/promoter elements of the nestin gene. By crossing these lines we obtained mice expressing both transgenes. To isolate neuronal progenitor cells directly from the developing brain, we used flow cytometry, selecting cells that expressed EGFP and EYFP simultaneously. We expect this strategy to provide valuable material with which to study the mechanisms of neurogenesis and to develop cell-based therapies for neurological disorders. J. Neurosci. Res. 65:220-227, 2001. (C) 2001 Wiley-Liss, Inc.

    DOI: 10.1002/jnr.1145

    Web of Science

    PubMed

    researchmap

  • In vitro neurogenesis by progenitor cells isolated from the adult human hippocampus Reviewed

    NS Roy, S Wang, L Jiang, J Kang, A Benraiss, C Harrison-Restelli, R Fraser, WT Couldwell, A Kawaguchi, H Okano, M Nedergaard, SA Goldman

    NEUROLOGICAL SURGERY   29 ( 2 )   195 - 195   2001.2

     More details

    Language:Japanese   Publishing type:Research paper (scientific journal)   Publisher:IGAKU-SHOIN LTD  

    Web of Science

    researchmap

  • Nestin-EGFP transgenic mice: Visualization of the self-renewal and multipotency of CNS stem cells Reviewed

    A Kawaguchi, T Miyata, K Sawamoto, N Takashita, A Murayama, W Akamatsu, M Ogawa, M Okabe, Y Tano, SA Goldman, H Okano

    MOLECULAR AND CELLULAR NEUROSCIENCE   17 ( 2 )   259 - 273   2001.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    We generated transgenic mice carrying enhanced green fluorescent protein (EGFP) under the control of the nestin second-intronic enhancer (E/nestin:EGFP). Flow cytometry followed by in vitro assays revealed that in situ EGFP expression in the embryonic brain correlated with the mitotic index, the cogeneration of both neurons and glia, and the frequency of neurosphere formation in vitro. High-level EGFP expressors derived from embryos included a distinct subpopulation of cells that were self-renewable and multipotent, criteria that define neural stem cells (NSCs). Such cells were largely absent among lower-level or non-EGFP expressors, thereby permitting us to enrich for NSCs using EGFP expression level. In adults, although E/nestin:EGFP-positive cells included the NSC population, the frequency of neurosphere formation did not correlate directly with the level of EGFP expression. However, moderately EGFP-expressing cells in adults gained EGFP intensity when they formed neurospheres, suggesting embryonic and adult NSCs exist in different microenvironments in vivo.

    DOI: 10.1006/mcne.2000.0925

    Web of Science

    PubMed

    researchmap

  • In vitro neurogenesis by progenitor cells isolated from the adult human hippocampus Reviewed

    NS Roy, S Wang, L Jiang, J Kang, A Benraiss, C Harrison-Restelli, RAR Fraser, WT Couldwell, A Kawaguchi, H Okano, M Nedergaard, SA Goldman

    NATURE MEDICINE   6 ( 3 )   271 - 277   2000.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:NATURE AMERICA INC  

    Neurogenesis persists in the adult mammalian hippocampus. To identify and isolate neuronal progenitor cells of the adult human hippocampus, we transfected ventricular zone-free dissociates of surgically-excised dentate gyrus with DNA encoding humanized green fluorescent protein (hGFP), placed under the control of either the nestin enhancer (E/nestin) or the T alpha 1 tubulin promoter (P/T alpha 1), two regulatory regions that direct transcription in neural progenitor cells. The resultant P/T alpha 1:hGFP(+) and E/nestin:enhanced (E)GFP(+) cells expressed beta III-tubulin or microtubule-associated protein-2; many incorporated bromodeoxyuridine, indicating their genesis in vitro. Using fluorescence-activated cell sorting, the E/nestin:EGFP(+) and P/T alpha 1:hGFP(+) cells were isolated to near purity, and matured antigenically and physiologically as neurons. Thus, the adult human hippocampus contains mitotically competent neuronal progenitors that can be selectively extracted. The isolation of these cells may provide a cellular substrate for re-populating the damaged or degenerated adult hippocampus.

    DOI: 10.1038/73119

    Web of Science

    PubMed

    researchmap

  • Promoter-targeted selection and isolation of neural progenitor cells from the adult human ventricular zone Reviewed

    NS Roy, A Benraiss, S Wang, RAR Fraser, R Goodman, WT Couldwell, M Nedergaard, A Kawaguchi, H Okano, SA Goldman

    JOURNAL OF NEUROSCIENCE RESEARCH   59 ( 3 )   321 - 331   2000.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-LISS  

    Adult humans, like their nonhuman mammalian counterparts, harbor persistent neural progenitor cells in the forebrain ventricular lining. In the absence of adequate surface markers, however, these cells have proven difficult to isolate for study. We have previously identified and selected neural progenitor cells from both the fetal and adult rodent ventricular zone (VZ), by sorting forebrain cells transfected with plasmid DNA encoding the gene for green fluorescent protein driven by the early neuronal promoter for T alpha 1 tubulin (P/T alpha 1:hGFP). We have now extended this approach by purifying both P/T alpha 1:hGFP tubulin-defined neuronal progenitors, as well as potentially less committed E/nestin:hGFP-defined neural progenitor cells, from the adult human VZ. The ventricular wall of the temporal horn of the lateral ventricle was dissected from temporal lobes obtained from four adult patients undergoing therapeutic lobectomy. These samples were dissociated, and the cultured cells transduced with either PT alpha 1:hGFP or E/nestin:EGFP plasmid DNA. A week later, the cells were redissociated, selected via fluorescence-activated cell sorting (FACS) on the basis of neural promoter-driven GFP expression, and replated. The majority of these cells expressed the early neuronal protein beta III-tubulin upon FAGS; within the week thereafter, most matured as morphologically evident neurons that coexpressed beta III-tubulin and microtubule-associated protein (MAP)-2. Many of these neurons had incorporated bromodeoxyuridine in vitro in the days before FAGS, indicating their mitogenesis in vitro. Thus, the use of fluorescent transgenes under the control of early neural promoters permits the enrichment of neuronal progenitor cells from the adult human ventricular zone. The specific acquisition, in both purity and number, of residual neural progenitor cells from the adult human brain may now permit hitherto unfeasible studies of both their biology and practical application. (C) 2000 Wiley-Liss, Inc.

    DOI: 10.1002/(SICI)1097-4547(20000201)59:3<321::AID-JNR5>3.0.CO;2-9

    Web of Science

    PubMed

    researchmap

▼display all

MISC

  • Regulation of neural stem cell morphology in brain Invited

    55 ( 10 )   850 - 853   2023.8

     More details

    Authorship:Last author, Corresponding author   Language:Japanese   Publishing type:Article, review, commentary, editorial, etc. (trade magazine, newspaper, online media)  

    researchmap

  • Morphological perspectives on the fate determination of neural progenitor cells Invited

    Ayano Kawaguchi

    Journal of Okayama Medical Association   135 ( 1 )   12 - 17   2023.4

     More details

    Authorship:Lead author, Last author, Corresponding author   Language:Japanese   Publishing type:Article, review, commentary, editorial, etc. (bulletin of university, research institution)  

    DOI: 10.4044/joma.135.12

    researchmap

  • 細胞離脱の実行役分子Lzts1による大脳形成制御 Reviewed

    川口綾乃

    生化学   92 ( 6 )   817 - 821   2020.12

     More details

  • 非対称分裂

    川口綾乃, 松崎文雄

    分子細胞治療   3 ( 1 )   138 - 139   2004

     More details

  • 実験講座 胎生期大脳組織の三次元培養:複雑さへの回帰

    宮田 卓樹, 齋藤 加奈子, 川口 綾乃

    生体の科学   53 ( 3 )   243 - 249   2002.5

     More details

    Language:Japanese   Publisher:金原一郎記念医学医療振興財団  

    DOI: 10.11477/mf.2425902403

    CiNii Article

    researchmap

  • 発達脳における神経細胞の移動:新しいニューロン移動法とその原理 (特集 脳の発達に関与する分子機構)

    宮田 卓樹, 川口 綾乃, 岡野 栄之

    生体の科学   52 ( 3 )   224 - 229   2001.5

     More details

    Language:Japanese   Publisher:金原一郎記念医学医療振興財団  

    DOI: 10.11477/mf.2425902273

    CiNii Article

    researchmap

  • 神経系前駆細胞及び成熟ニューロンの非対称性に関する細胞生物学的解析

    岡野栄之, 飯島崇利, 川口綾乃, 宮田卓樹, 今井貴雄, 小川正晴

    日本分子生物学会年会プログラム・講演要旨集   23rd   292   2000.11

     More details

    Language:Japanese  

    J-GLOBAL

    researchmap

  • 神経幹細胞の同定--中枢神経系の再生を目指して (特集 幹細胞システムと再生医学)

    川口 綾乃, 岡野 栄之

    細胞工学   19 ( 3 )   392 - 397   2000.3

     More details

    Language:Japanese   Publisher:秀潤社  

    CiNii Article

    CiNii Books

    researchmap

  • FACSを用いた神経系前駆細胞の分離

    川口綾乃, 岡野栄之

    実験医学   18   1106 - 1108   2000

     More details

  • 神経幹細胞の同定とその中枢神経再生への応用の展望

    川口綾乃, 岡野栄之

    最新医学   54   1721 - 1729   1999

▼display all

Research Projects

  • 上皮構造からの細胞離脱による器官形成制御

    2022 - 2024

    科学技術振興機構(JST)  創発的研究支援事業 (フェーズ1) 

    川口綾乃

      More details

  • 上皮構造からの細胞の離脱・移動の新しい分子制御機構

    2022 - 2023

    上原記念生命科学財団  研究助成金 

    川口綾乃

      More details

  • 異所性灰白質病態と脳進化に関わる脳室下帯の形成メカニズムの解明

    2020 - 2023

    文部科学省  科学研究費補助金 基盤B 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 場の変化が明らかにする神経前駆細胞の時間個性獲得の機構

    2019 - 2020

    文部科学省  科学研究費補助金 新学術領域(公募研究) 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 大脳皮質の脳回形成に寄与する神経前駆細胞移動の分子機構

    2018

    堀科学芸術振興財団  第27回研究助成 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • Outer radial glia誕生がもたらす神経幹細胞の場と時間特性の変化

    2017 - 2018

    文部科学省  科学研究費補助金 新学術領域(公募研究) 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 脳室帯のシステム科学的研究

    2016 - 2020

    文部科学省  科学研究費補助金 基盤A 

    宮田卓樹

      More details

    Grant type:Competitive

    researchmap

  • 大脳発生過程における脳室面からの細胞離脱制御

    2016 - 2019

    文部科学省  科学研究費補助金 基盤C 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 神経幹細胞の分化に際し速やかに発現変動する遺伝子の脳組織形成への関与

    2013 - 2016

    文部科学省  科学研究費補助金 基盤C 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 発生時期による神経幹細胞の分裂パターンの変化を制御する機構の解明

    2011 - 2012

    文部科学省  科学研究費補助金 新学術領域(公募研究) 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • Neurogenesis regulated through three-dimensional cellular movement and cell-cell interactions within the neuroepithelium

    Grant number:22111006  2010.04 - 2016.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research  Grant-in-Aid for Scientific Research on Innovative Areas (Research in a proposed research area)

    Miyata Takaki, KAWAGUCHI Ayano, SAKAKIBARA Akira, HASHIMOTO Mitsuhiro, SHINODA Tomoyasu, OKAMOTO Mayumi

      More details

    Authorship:Collaborating Investigator(s) (not designated on Grant-in-Aid) 

    Grant amount:\190190000 ( Direct expense: \146300000 、 Indirect expense:\43890000 )

    Belonging to the "Cross-talk between moving cells and microenvironment as a basis of emerging order in multicellular system", this research project studied how movements of neural progenitor cells are coordinated to establish the safe and efficient "neurogenesis" (i.e. production of neurons to build a brain structure) without suffering from a "traffic jam" of cells in a narrow tissue-developing space. Using new techniques such as live imaging of all cells, quantitative analysis on trajectories of moving cells, and mechanical experiments, we found that cells are cleverly moving in a manner similar to "staggered commuting" (i.e. one cell goes first then the other follows). If this "crowd control" method does not work during development, brain structure cannot form normally (Nature Neuroscience, 2013). We further demonstrated brain cells' migration strategy is different between mice and ferret, suggesting that control of cellular movements may underlie brain evolution.

    researchmap

  • 発生期の脳組織中で神経幹細胞の維持と分化を制御する機構の解明

    2010 - 2012

    文部科学省  科学研究費補助金 基盤C 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 小脳発生の研究:胎生期のニューロン誕生・移動の機構

    2009 - 2012

    文部科学省  科学研究費補助金 基盤A 

    宮田卓樹

      More details

    Grant type:Competitive

    researchmap

  • 単一細胞マイクロアレイ法を用いた神経前駆細胞の時期特異的な分化制御機構の解析

    2008 - 2010

    名古屋大学学術振興基金  H20年度第2回学術研究助成 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 単一細胞遺伝子発現解析法を用いた、ほ乳類中枢神経系の前駆細胞の運命決定因子の探索

    2006 - 2007

    文部科学省  科学研究費補助金 若手(A) 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 哺乳類中枢神経系における非対称分裂を介した細胞運命決定機構の解析

    2003 - 2005

    文部科学省  科学研究費補助金 若手(B) 

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 中枢神経系幹細胞による細胞産生、組織構築のメカニズムの解明

    Grant number:01J01058  2001

    日本学術振興会  特別研究奨励費  特別研究員奨励費

    川口綾乃

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

▼display all

 

Class subject in charge

  • Primary Anatomy (2023academic year) special  - その他

  • Practicals: Human Morphology (2023academic year) special  - その他

  • Research Projects: Human Morphology (2023academic year) special  - その他

  • Research Projects and Practicals: Human Morphology I (2023academic year) special  - その他

  • Lecture and Research Projects: Human Morphology I (2023academic year) special  - その他

  • Research Projects and Practicals: Human Morphology II (2023academic year) special  - その他

  • Lecture and Research Projects: Human Morphology II (2023academic year) special  - その他

  • Human Anatomy (2023academic year) Concentration  - その他

  • Human Anatomy (2023academic year) special  - その他

  • Human Anatomy (2023academic year) Third semester  - 水4~5

  • Human Anatomy (2023academic year) Third semester  - 水4~5

  • Human Anatomy (2023academic year) Third semester  - 水4~5

  • Human Anatomy (2023academic year) Third semester  - 水4~5

  • Human Gross Anatomy (2023academic year) Concentration  - その他

  • Human Gross Anatomy (2023academic year) special  - その他

  • Research Presentation in Cell and Tissue Engineering (2023academic year) special  - その他

  • Primary Anatomy (2022academic year) special  - その他

  • Human Anatomy (2022academic year) special  - その他

  • Human Anatomy (2022academic year) Third semester  - 水4~5

  • Human Anatomy (2022academic year) Third semester  - 水4~5

  • Human Anatomy (2022academic year) Third semester  - 水4~5

  • Human Anatomy (2022academic year) Third semester  - 水4~5

  • Human Gross Anatomy (2022academic year) Concentration  - その他

  • Human Gross Anatomy (2022academic year) special  - その他

▼display all