Network


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

Hotspot


Dive into the research topics where Ken-ichi Mizutani is active.

Publication


Featured researches published by Ken-ichi Mizutani.


Development | 2005

Progenitors resume generating neurons after temporary inhibition of neurogenesis by Notch activation in the mammalian cerebral cortex.

Ken-ichi Mizutani; Tetsuichiro Saito

The mammalian cerebral cortex comprises six layers of neurons. Cortical progenitors in the ventricular zone generate neurons specific to each layer through successive cell divisions. Neurons of layer VI are generated at an early stage, whereas later-born neurons occupy progressively upper layers. The underlying molecular mechanisms of neurogenesis, however, are relatively unknown. In this study, we devised a system where the Notch pathway was activated spatiotemporally in the cortex by in vivo electroporation and Cre-mediated DNA recombination. Electroporation at E13.5 transferred DNA to early progenitors that gave rise to neurons of both low and upper layers. Forced expression of a constitutively active form of Notch (caNotch) at E13.5 inhibited progenitors from generating neurons and kept progenitors as proliferating radial glial cells. After subsequent transfection at E15.5 of a Cre expression vector to remove caNotch, double-transfected cells, in which caNotch was excised, migrated into the cortical plate and differentiated into neurons specific to upper layers. Bromodeoxyuridine-labeling experiments showed that the neurons were born after Cre transfection. These results indicate that cortical progenitors that had been temporarily subjected to Notch activation at an early stage generated neurons at later stages, but that the generation of low-layer neurons was skipped. Moreover, the double-transfected cells gave rise to upper-layer neurons, even after their transplantation into the E13.5 brain, indicating that the developmental state of progenitors is not halted by caNotch activity.


PLOS ONE | 2014

Prdm8 Regulates the Morphological Transition at Multipolar Phase during Neocortical Development

Mayuko Inoue; Takao Kuroda; Aya Honda; Mariko Komabayashi-Suzuki; Tae Komai; Yoichi Shinkai; Ken-ichi Mizutani

Here, we found that the PR domain protein Prdm8 serves as a key regulator of the length of the multipolar phase by controlling the timing of morphological transition. We used a mouse line with expression of Prdm8-mVenus reporter and found that Prdm8 is predominantly expressed in the middle and upper intermediate zone during both the late and terminal multipolar phases. Prdm8 expression was almost coincident with Unc5D expression, a marker for the late multipolar phase, although the expression of Unc5D was found to be gradually down-regulated to the point at which mVenus expression was gradually up-regulated. This expression pattern suggests the possible involvement of Prdm8 in the control of the late and terminal multipolar phases, which controls the timing for morphological transition. To test this hypothesis, we performed gain- and loss-of-function analysis of neocortical development by using in utero electroporation. We found that the knockdown of Prdm8 results in premature change from multipolar to bipolar morphology, whereas the overexpression of Prdm8 maintained the multipolar morphology. Additionally, the postnatal analysis showed that the Prdm8 knockdown stimulated the number of early born neurons, and differentiated neurons located more deeply in the neocortex, however, majority of those cells could not acquire molecular features consistent with laminar location. Furthermore, we found the candidate genes that were predominantly utilized in both the late and terminal multipolar phases, and these candidate genes included those encoding for guidance molecules. In addition, we also found that the expression level of these guidance molecules was inhibited by the introduction of the Prdm8 expression vector. These results indicate that the Prdm8-mediated regulation of morphological changes that normally occur during the late and terminal multipolar phases plays an important role in neocortical development.


Genes to Cells | 2015

Deletion of Prdm8 impairs development of upper-layer neocortical neurons

Mayuko Inoue; Ryota Iwai; Emiko Yamanishi; Kazuyuki Yamagata; Mariko Komabayashi-Suzuki; Aya Honda; Tae Komai; Hitoshi Miyachi; Satsuki Kitano; Chisato Watanabe; Waka Teshima; Ken-ichi Mizutani

Upper‐layer (UL) neocortical neurons are the most prominent distinguishing features of the mammalian neocortex compared with those of the avian dorsal cortex and are vastly expanded in primates. However, little is known about the identities of the genes that control the specification of UL neurons. Here, we found that Prdm8, a member of the PR (PRDI‐BF1 and RIZ homology) domain protein family, was specifically expressed in the postnatal UL neocortex, particular those in late‐born RORß‐positive layer IV neurons. We generated homozygous Prdm8 knockout (Prdm8 KO) mice and found that the deletion of Prdm8 causes growth retardation and a reduced brain weight, although the brain weight‐to‐body weight ratio is unchanged at postnatal day 8 (P8). Immunohistochemistry showed that the relative UL thickness, but not the thickness of the deep layer (DL), was significantly reduced in Prdm8 KO mice compared with wild‐type (WT) mice. In addition, we found that a number of late‐born Brn2‐positive UL neurons were significantly decreased in Prdm8 KO mice. To identify genes regulated by Prdm8 during neocortical development, we compared expression profiling analysis in Prdm8 KO and WT mice, and identified some candidate genes. These results suggest that the proper expression of Prdm8 is required for the normal development and construction of UL neurons in the mammalian neocortex.


Development | 2017

Prdm16 is crucial for progression of the multipolar phase during neural differentiation of the developing neocortex.

Mayuko Inoue; Ryota Iwai; Hidenori Tabata; Daijiro Konno; Mariko Komabayashi-Suzuki; Chisato Watanabe; Hiroko Iwanari; Yasuhiro Mochizuki; Takao Hamakubo; Fumio Matsuzaki; Koh-ichi Nagata; Ken-ichi Mizutani

ABSTRACT The precise control of neuronal migration and morphological changes during differentiation is essential for neocortical development. We hypothesized that the transition of progenitors through progressive stages of differentiation involves dynamic changes in levels of mitochondrial reactive oxygen species (mtROS), depending on cell requirements. We found that progenitors had higher levels of mtROS, but that these levels were significantly decreased with differentiation. The Prdm16 gene was identified as a candidate modulator of mtROS using microarray analysis, and was specifically expressed by progenitors in the ventricular zone. However, Prdm16 expression declined during the transition into NeuroD1-positive multipolar cells. Subsequently, repression of Prdm16 expression by NeuroD1 on the periphery of ventricular zone was crucial for appropriate progression of the multipolar phase and was required for normal cellular development. Furthermore, time-lapse imaging experiments revealed abnormal migration and morphological changes in Prdm16-overexpressing and -knockdown cells. Reporter assays and mtROS determinations demonstrated that PGC1α is a major downstream effector of Prdm16 and NeuroD1, and is required for regulation of the multipolar phase and characteristic modes of migration. Taken together, these data suggest that Prdm16 plays an important role in dynamic cellular redox changes in developing neocortex during neural differentiation. Summary: During mouse neurogenesis, levels of mitochondrial reactive oxygen species (mtROS) decline during differentiation. Prdm16 and PGC1α are candidate regulators of this metabolic shift.


Scientific Reports | 2017

Fluorescence and Bioluminescence Imaging of Angiogenesis in Flk1-Nano-lantern Transgenic Mice

Jun Matsushita; Shigenori Inagaki; Tomomi Nishie; Tomoki Sakasai; Junko Tanaka; Chisato Watanabe; Ken-ichi Mizutani; Yoshihiro Miwa; Ken Matsumoto; Kazuhiro Takara; Hisamichi Naito; Hiroyasu Kidoya; Nobuyuki Takakura; Takeharu Nagai; Satoru Takahashi; Masatsugu Ema

Angiogenesis is important for normal development as well as for tumour growth. However, the molecular and cellular mechanisms underlying angiogenesis are not fully understood, partly because of the lack of a good animal model for imaging. Here, we report the generation of a novel transgenic (Tg) mouse that expresses a bioluminescent reporter protein, Nano-lantern, under the control of Fetal liver kinase 1 (Flk1). Flk1-Nano-lantern BAC Tg mice recapitulated endogenous Flk1 expression in endothelial cells and lymphatic endothelial cells during development and tumour growth. Importantly, bioluminescence imaging of endothelial cells from the aortic rings of Flk1-Nano-lantern BAC Tg mice enabled us to observe endothelial sprouting for 18 hr without any detectable phototoxicity. Furthermore, Flk1-Nano-lantern BAC Tg mice achieved time-lapse luminescence imaging of tumour angiogenesis in freely moving mice with implanted tumours. Thus, this transgenic mouse line contributes a unique model to study angiogenesis within both physiological and pathological contexts.


Biochemical and Biophysical Research Communications | 2018

A Prdm8 target gene Ebf3 regulates multipolar-to-bipolar transition in migrating neocortical cells

Ryota Iwai; Hidenori Tabata; Mayuko Inoue; Kei-ichiro Nomura; Tadashi Okamoto; Masamitsu Ichihashi; Koh-ichi Nagata; Ken-ichi Mizutani

Precise control of neuronal migration is essential for the development of the neocortex. However, the molecular mechanisms underlying neuronal migration remain largely unknown. Here we identified helix-loop-helix transcription factor Ebf3 as a Prdm8 target gene, and found that Ebf3 is a key regulator of neuronal migration via multipolar-to-bipolar transition. Ebf3 knockdown cells exhibited severe defects in the formation of leading processes and an inhibited shift to the locomotion mode. Moreover, we found that Ebf3 knockdown represses NeuroD1 transcription, and NeuroD1 overexpression partially rescued migration defects in Ebf3 knockdown cells. Our findings highlight the critical role of Ebf3 in multipolar-to-bipolar transition via positive feedback regulation of NeuroD1 in the developing neocortex.


The Japanese Biochemical Society/The Molecular Biology Society of Japan | 2017

Prdm16 is crucial for progression of the multipolar phase during neural differentiation.

Ryota Iwai; Mayuko Inoue; Hidenori Tabata; Daijiro Konno; Mariko Suzuki; Takao Hamakubo; Fumio Matsuzaki; Koh-ichi Nagata; Ken-ichi Mizutani


The Molecular Biology Society of Japan | 2016

Avascular region in the developing neocortex regulates contact between dividing neural progenitors and vascular endothelial cells.

Ken-ichi Mizutani; Ryota Iwai; Mariko Suzuki; Emiko Yamanishi; Hidenori Tabata; Chisato Watanabe; Waka Teshima; Kazuo Kinoshita; Hiroshi Harada; Koh-ichi Nagata; Masatsugu Ema


The Molecular Biology Society of Japan | 2016

Prdm16, a modulator of mtROS is critical for progression of multipolar phase during neural differentiation of the developing neocortex.

Ryota Iwai; Mayuko Inoue; Hidenori Tabata; Mariko Suzuki; Hiroko Iwanari; Yasuhiro Mochizuki; Takao Hamakubo; Daijiro Konno; Fumio Matsuzaki; Koh-ichi Nagata; Ken-ichi Mizutani


The Molecular Biology Society of Japan | 2016

Patterning of angiogenesis in the developing neocortex and its physiological role

Chisato Watanabe; Chiaki Ohtaka-Maruyama; Mariko Suzuki; Ryota Iwai; Emiko Yamanishi; Syoko Yamamoto; Masatsugu Ema; Ken-ichi Mizutani

Collaboration


Dive into the Ken-ichi Mizutani'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge