Network


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

Hotspot


Dive into the research topics where Kenji Hanamura is active.

Publication


Featured researches published by Kenji Hanamura.


The Journal of Comparative Neurology | 2005

Drebrin A is a postsynaptic protein that localizes in vivo to the submembranous surface of dendritic sites forming excitatory synapses

Chiye Aoki; Yuko Sekino; Kenji Hanamura; Sho Fujisawa; Veeravan Mahadomrongkul; Yong Ren; Tomoaki Shirao

Drebrin A is a neuron‐specific, actin binding protein. Evidence to date is from in vitro studies, consistently supporting the involvement of drebrin A in spinogenesis and synaptogenesis. We sought to determine whether drebrin A arrives at the plasma membrane of neurons, in vivo, in time to orchestrate spinogenesis and synaptogenesis. To this end, a new antibody was used to locate drebrin A in relation to electron microscopically imaged synapses during early postnatal days. Western blotting showed that drebrin A emerges at postnatal day (PNd) 6 and becomes progressively more associated with F‐actin in the pellet fraction. Light microscopy showed high concentrations of drebrin A in the synaptic layers of the hippocampus and cortex. Electron microscopy revealed that drebrin A in these regions is located exclusively in dendrites both neonatally and in adulthood. In adulthood, nearly all of the synaptic drebrin A is within spines forming asymmetric excitatory synapses, verified by γ‐aminobutyric acid (GABA) negativity. At PNd7, patches of drebrin A immunoreactivity were discretely localized to the submembranous surfaces of dendrites forming slight protrusions—protospines. The drebrin A sites exhibited only thin postsynaptic densities and lacked axonal associations or were contacted by axons that contained only a few vesicles. Yet, because of their immunoreactivity to the NR2B subunit of N‐methyl‐D‐aspartate receptors and immunonegativity of axon terminals to GABA, these could be presumed to be nascent, excitatory synapses. Thus, drebrin A may be involved in organizing the dendritic pool of actin for the formation of spines and of axospinous excitatory synapses during early postnatal periods. J. Comp. Neurol. 483:383–402, 2005.


Journal of Neurochemistry | 2009

Drebrin E is involved in the regulation of axonal growth through actin–myosin interactions

Toshiyuki Mizui; Nobuhiko Kojima; Hiroyuki Yamazaki; Michihito Katayama; Kenji Hanamura; Tomoaki Shirao

Drebrin is a well‐known side‐binding protein of F‐actin in the brain. Immunohistochemical data suggest that the peripheral parts of growing axons are enriched in the drebrin E isoform and mature axons are not. It has also been observed that drebrin E is concentrated in the growth cones of PC12 cells. These data strongly suggest that drebrin E plays a role in axonal growth during development. In this study, we used primary hippocampal neuronal cultures to analyze the role of drebrin E. Immunocytochemistry showed that within axonal growth cones drebrin E specifically localized to the transitional zone, an area in which dense networks of F‐actins and microtubules overlapped. Over‐expression of drebrin E caused drebrin E and F‐actin to accumulate throughout the growth cone and facilitated axonal growth. In contrast, knockdown of drebrin E reduced drebrin E and F‐actin in the growth cone and prevented axonal growth. Furthermore, inhibition of myosin II ATPase masked the promoting effects of drebrin E over‐expression on axonal growth. These results suggest that drebrin E plays a role in axonal growth through actin–myosin interactions in the transitional zone of axonal growth cones.


Molecular and Cellular Neuroscience | 2006

Activation of N-methyl-D-aspartate receptor induces a shift of drebrin distribution : Disappearance from dendritic spines and appearance in dendritic shafts

Yuko Sekino; Satoshi Tanaka; Kenji Hanamura; Hiroyuki Yamazaki; Yoshio Sasagawa; Yinhuan Xue; Kensuke Hayashi; Tomoaki Shirao

Drebrin is a major actin-filament-binding protein localized in mature dendritic spines. A recent in vivo immunoelectron microscopic study suggests that drebrin content at each dendritic spine is regulated by some unknown mechanisms. In the present in vitro study, we examined whether glutamate stimulation alters drebrin content in dendritic spines. Glutamate stimulation induced disappearance of drebrin immunostaining from dendritic spines but led to appearance of drebrin immunostaining in dendritic shafts and somata. The glutamate-induced shift of drebrin immunostaining was blocked by an NMDA receptor antagonist. Immunoblot analyses showed that both the total and the cytosolic drebrin remained unchanged and revealed that the drebrin shift was not due to drebrin degradation. These findings indicate that NMDA receptor activation induces a shift in subcellular distribution of drebrin associated with actin filaments, and that the shift might be a molecular basis for actin reorganization accompanied with synaptic plasticity.


Journal of Cell Science | 2009

Activity of the AMPA receptor regulates drebrin stabilization in dendritic spine morphogenesis

Hideto Takahashi; Hiroyuki Yamazaki; Kenji Hanamura; Yuko Sekino; Tomoaki Shirao

Spine morphogenesis mainly occurs during development as a morphological shift from filopodia-like thin protrusions to bulbous ones. We have previously reported that synaptic clustering of the actin-binding protein drebrin in dendritic filopodia governs spine morphogenesis and synaptic PSD-95 clustering. Here, we report the activity-dependent cellular mechanisms for spine morphogenesis, in which the activity of AMPA receptors (AMPARs) regulates drebrin clustering in spines by promoting drebrin stabilization. In cultured developing hippocampal neurons, pharmacological blockade of AMPARs, but not of other glutamate receptors, suppressed postsynaptic drebrin clustering without affecting presynaptic clustering of synapsin I (synapsin-1). Conversely, the enhancement of the action of AMPARs promoted drebrin clustering in spines. When we explored drebrin dynamics by photobleaching individual spines, we found that AMPAR activity increased the fraction of stable drebrin without affecting the time constant of drebrin turnover. An increase in the fraction of stable drebrin corresponded with increased drebrin clustering. AMPAR blockade also suppressed normal morphological maturation of spines and synaptic PSD-95 clustering in spines. Together, these data suggest that AMPAR-mediated stabilization of drebrin in spines is an activity-dependent cellular mechanism for spine morphogenesis.


Neuroscience | 2010

Genetic disruption of the alternative splicing of drebrin gene impairs context-dependent fear learning in adulthood.

Nobuhiko Kojima; Kenji Hanamura; Hiroyuki Yamazaki; T. Ikeda; Shigeyoshi Itohara; Tomoaki Shirao

Dendritic spines are postsynaptic structures at excitatory synapses that play important roles in synaptic transmission and plasticity. Dendritic spine morphology and function are regulated by an actin-based cytoskeletal network. Drebrin A, an adult form of drebrin, is an actin-binding protein in dendritic spines, and its decrease is purportedly concerned with synaptic dysfunction in Alzheimers disease. Rapid conversion of drebrin E, an embryonic form of drebrin, to drebrin A occurs in parallel with synaptic maturation. To understand the physiological role of drebrin isoform conversion in vivo, we generated knockout mice in which a drebrin A-specific exon was deleted from the drebrin gene. Drebrin A-specific knockout (DAKO) mice expressed drebrin E, which substituted for drebrin A. Subcellular fractionation experiment indicated that cytosolic form of drebrin was increased in the brains of DAKO mice. Furthermore, drebrin accumulation in synaptosomes of DAKO mice was much higher than that of wild-type (WT) mice. DAKO mice were viable and showed no apparent abnormalities in their gross brain morphology and general behaviors. However, DAKO mice were impaired in a context-dependent freezing after fear conditioning. These data indicate that drebrin A plays an indispensable role in some processes of generating fear learning and memory.


Neuroscience | 2008

Expression of drebrin E in migrating neuroblasts in adult rat brain: coincidence between drebrin E disappearance from cell body and cessation of migration.

Mingqiao Song; Nobuhiko Kojima; Kenji Hanamura; Yuko Sekino; Hiroshi K. Inoue; Masahiko Mikuni; Tomoaki Shirao

Migrating neuroblasts in the adult brain form the rostral migratory stream (RMS) from the lateral ventricle to the olfactory bulb (OB) and then differentiate in the OB. In this study, we immunohistochemically analyzed drebrin expression in the RMS of the adult rat brain. Although drebrin is concentrated in dendritic spines of mature neurons, drebrin-immunopositive (DIP) cell bodies were observed in the RMS. The polysialated form of a neural cell adhesion molecule (PSA-NCAM) was detected in DIP cells. K(i)-67, a marker of proliferating cells, was also detected in a subset of DIP cells; however, neither glial fibrillary acidic protein, nestin nor vimentin was detected in DIP cells. These results indicate that DIP cells in the RMS are migrating neuroblasts. An image subtraction method, based on using anti-pan-drebrin and anti-drebrin A antibodies, demonstrated that DIP migrating neuroblasts are immunopositive for drebrin E but not for drebrin A (E+A-). Furthermore, olfactory bulbectomy increased the number of cells with drebrin E+A- signals in the RMS, indicating that these cells migrate along the RMS. Drebrin E+A- cells were also found in the subgranular layer of the dentate gyrus and in the piriform cortex. Thus, detection of drebrin E+A- signals is useful for identifying migrating neuroblasts in the adult brain. In the OB, drebrin E+A- signals were observed in the cell bodies of migrating neuroblasts in the core region; however, only fibrous and punctate drebrin E+A- signals were observed in postmigratory neuroblasts at the outer layers. These data demonstrate that the disappearance of drebrin E+A- signals from the cell body coincides with the cessation of neuronal migration. The disappearance of drebrin E from the cell body may be a molecular switch for the cessation of migration in newly generated neuroblasts.


Muscle & Nerve | 2011

Evidence for cell density affecting C2C12 myogenesis: possible regulation of myogenesis by cell-cell communication.

Kanako Tanaka; Kaori Sato; Tomomi Yoshida; Toshio Fukuda; Kenji Hanamura; Nobuhiko Kojima; Tomoaki Shirao; Takashi Yanagawa; Hideomi Watanabe

Introduction: Community effect is a phenomenon caused by cell–cell communication during myogenesis. In myogenic C2C12 cells in vitro, the confluent phase is needed for myogenesis induction. Methods: To examine the cell‐density effect, growth kinetics and myogenic differentiation were investigated in cells plated at four different cell densities. Results: We found that expression of a myogenic differentiation marker was high in a density‐dependent manner. At high density, where cell–cell contact was obvious, contact inhibition after the proliferation stage was accompanied by microarray findings demonstrating upregulation of negative regulating cell‐cycle markers, including CDKI p21 and the muscle differentiation markers MyoD and myogenin. Interestingly, developmentally regulated protein expression (drebrin) protein expression was also upregulated in a density‐dependent manner. Conclusions: These results suggest that contact inhibition after the proliferation stage may induce growth arrest via cell–cell communication through the expression of CDKI p21 and may be responsible for progressing cell fusion. Muscle Nerve 2011


Journal of Neurochemistry | 2014

Spikar, a novel drebrin-binding protein, regulates the formation and stabilization of dendritic spines.

Hiroyuki Yamazaki; Nobuhiko Kojima; Kenichi Kato; Eiji Hirose; Toshiharu Iwasaki; Toshiyuki Mizui; Hideto Takahashi; Kenji Hanamura; Reiko T. Roppongi; Noriyuki Koibuchi; Yuko Sekino; Nozomu Mori; Tomoaki Shirao

Dendritic spines are small, actin‐rich protrusions on dendrites, the development of which is fundamental for the formation of neural circuits. The actin cytoskeleton is central to dendritic spine morphogenesis. Drebrin is an actin‐binding protein that is thought to initiate spine formation through a unique drebrin‐actin complex at postsynaptic sites. However drebrin overexpression in neurons does not increase the final density of dendritic spines. In this study, we have identified and characterized a novel drebrin‐binding protein, spikar. Spikar is localized in cell nuclei and dendritic spines, and accumulation of spikar in dendritic spines directly correlates with spine density. A reporter gene assay demonstrated that spikar acts as a transcriptional co‐activator for nuclear receptors. We found that dendritic spine, but not nuclear, localization of spikar requires drebrin. RNA‐interference knockdown and overexpression experiments demonstrated that extranuclear spikar regulates dendritic spine density by modulating de novo spine formation and retraction of existing spines. Unlike drebrin, spikar does not affect either the morphology or function of dendritic spines. These findings indicate that drebrin‐mediated postsynaptic accumulation of spikar regulates spine density, but is not involved in regulation of spine morphology.


Neuroscience | 2016

Drebrin A regulates hippocampal LTP and hippocampus-dependent fear learning in adult mice.

Nobuhiko Kojima; Hiroki Yasuda; Kenji Hanamura; Yuta Ishizuka; Yuko Sekino; Tomoaki Shirao

Structural plasticity of dendritic spines, which underlies higher brain functions including learning and memory, is dynamically regulated by the actin cytoskeleton and its associated proteins. Drebrin A is an F-actin-binding protein preferentially expressed in the brain and localized in the dendritic spines of mature neurons. Isoform conversion from drebrin E to drebrin A and accumulation of the latter in dendritic spines occurs during synapse maturation. We have previously demonstrated that drebrin A plays a pivotal role in spine morphogenesis and plasticity. However, it is unclear whether drebrin A plays a specific role in processes required for structural plasticity, and whether drebrin E can substitute in this role. To answer these questions, we analyzed mutant mice (named DAKO mice), in which isoform conversion from drebrin E to drebrin A is disrupted. In DAKO mouse brain, drebrin E continues to be expressed throughout life instead of drebrin A. Electrophysiological studies using hippocampal slices revealed that long-term potentiation of CA1 synapses was impaired in adult DAKO mice, but not in adolescents. In parallel with this age-dependent impairment, DAKO mice exhibited impaired hippocampus-dependent fear learning in an age-dependent manner; the impairment was evident in adult mice, but not in adolescents. In addition, histological investigation revealed that the spine length of the apical dendrite of CA1 pyramidal cells was significantly longer in adult DAKO mice than in wild-type mice. Our data indicate that the roles of drebrin E and drebrin A in brain function are different from each other, that the isoform conversion of drebrin is critical, and that drebrin A is indispensable for normal synaptic plasticity and hippocampus-dependent fear memory in the adult brain.


Radiation Research | 2013

X Irradiation Changes Dendritic Spine Morphology and Density through Reduction of Cytoskeletal Proteins in Mature Neurons

Katsuyuki Shirai; Toshiyuki Mizui; Yoshiyuki Suzuki; Masahiko Okamoto; Kenji Hanamura; Yukari Yoshida; Mizuki Hino; Shin-ei Noda; Wael S. Al-Jahdari; Arnab Chakravarti; Tomoaki Shirao; Takashi Nakano

Neurons are essential components of neural circuits and provide brain function organization. We previously reported that X irradiation induces apoptosis in immature neurons. To the best of our knowledge, there have been few reports investigating the effects of X irradiation on mature neurons. We analyzed the effects of X irradiation on the morphology, density and cytoskeletal proteins in dendritic spines on mature neurons. We prepared developing hippocampal neurons from 18 days embryo by using Bankers method. Neurons at 21 days in vitro were X irradiated at several doses and were immediately fixed. To evaluate the dendritic spine morphology and density, the neurons were transfected with a reporter plasmid for enhanced green fluorescent protein (GFP). Changes in the dendritic spines as a result of X irradiation were evaluated using electron microscopy. To analyze the cytoskeletal proteins within the dendritic spines, we performed immunocytochemistry to detect filamentous actin (F-actin), drebrin and PSD-95. X irradiation immediately changed the dendritic spine morphology, and the irradiated spines were significantly thinner and longer than the nonirradiated spines. X irradiation decreased the dendritic spine density in a dose-dependent manner. Electron microscopy confirmed these changes of dendritic spines by X irradiation. Immunohistochemical studies showed that X irradiation decreased the accumulation of drebrin and F-actin, but not PSD-95, within the dendritic spines. These results suggest that X irradiation immediately decreases the dendritic spine density and changes the morphology of mature neurons by reducing the abundance of cytoskeletal proteins. The abnormal dendritic spines may be associated with acute adverse effects after X irradiation in a clinical setting, although further investigations are warranted to validate these findings.

Collaboration


Dive into the Kenji Hanamura'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