Network


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

Hotspot


Dive into the research topics where Yuko Sekino is active.

Publication


Featured researches published by Yuko Sekino.


Neurochemistry International | 2007

Role of actin cytoskeleton in dendritic spine morphogenesis.

Yuko Sekino; Nobuhiko Kojima; Tomoaki Shirao

Dendritic spines are the postsynaptic receptive regions of most excitatory synapses, and their morphological plasticity play a pivotal role in higher brain functions, such as learning and memory. The dynamics of spine morphology is due to the actin cytoskeleton concentrated highly in spines. Filopodia, which are thin and headless protrusions, are thought to be precursors of dendritic spines. Drebrin, a spine-resident side-binding protein of filamentous actin (F-actin), is responsible for recruiting F-actin and PSD-95 into filopodia, and is suggested to govern spine morphogenesis. Interestingly, some recent studies on neurological disorders accompanied by cognitive deficits suggested that the loss of drebrin from dendritic spines is a common pathognomonic feature of synaptic dysfunction. In this review, to understand the importance of actin-binding proteins in spine morphogenesis, we first outline the well-established knowledge pertaining to the actin cytoskeleton in non-neuronal cells, such as the mechanism of regulation by small GTPases, the equilibrium between globular actin (G-actin) and F-actin, and the distinct roles of various actin-binding proteins. Then, we review the dynamic changes in the localization of drebrin during synaptogenesis and in response to glutamate receptor activation. Because side-binding proteins are located upstream of the regulatory pathway for actin organization via other actin-binding proteins, we discuss the significance of drebrin in the regulatory mechanism of spine morphology through the reorganization of the actin cytoskeleton. In addition, we discuss the possible involvement of an actin-myosin interaction in the morphological plasticity of spines.


The Journal of Neuroscience | 2014

Microglia Enhance Neurogenesis and Oligodendrogenesis in the Early Postnatal Subventricular Zone

Yukari Shigemoto-Mogami; Kazue Hoshikawa; James E. Goldman; Yuko Sekino; Kaoru Sato

Although microglia have long been considered as brain resident immune cells, increasing evidence suggests that they also have physiological roles in the development of the normal CNS. In this study, we found large numbers of activated microglia in the forebrain subventricular zone (SVZ) of the rat from P1 to P10. Pharmacological suppression of the activation, which produces a decrease in levels of a number of proinflammatory cytokines (i.e., IL-1β, IL-6, TNF-α, and IFN-γ) significantly inhibited neurogenesis and oligodendrogenesis in the SVZ. In vitro neurosphere assays reproduced the enhancement of neurogenesis and oligodendrogenesis by activated microglia and showed that the cytokines revealed the effects complementarily. These results suggest that activated microglia accumulate in the early postnatal SVZ and that they enhance neurogenesis and oligodendrogenesis via released cytokines.


The Journal of Comparative Neurology | 1999

Cellular localization of adenosine A1 receptors in rat forebrain: immunohistochemical analysis using adenosine A1 receptor-specific monoclonal antibody.

Tomoyo Ochiishi; Ling Chen; Aya Yukawa; Yoshiko Saitoh; Yuko Sekino; Takao Arai; Hiroyasu Nakata; Hiroshi Miyamoto

Monoclonal antibodies were generated against the adenosine A1 receptor (A1R) purified from rat brain. In immunoblot analyses of purified or partially purified A1R preparations from rat brain, these antibodies recognized a solitary band, the size of which corresponded to that expected for A1R. These antibodies recognized not only the native form of A1R but also the deglycosylated form of A1R. Immunocytochemical analysis of Chinese hamster ovarian cells that were transfected stably with rat A1R cDNA showed that their cell bodies were stained intensely by these antibodies, whereas nontransfected Chinese hamster ovarian cells were not. These antibodies detected the A1R naturally present in the DDT1 MF‐2 smooth muscle cells. One of these antibodies (the 511CA antibody) was then used to examine the immunohistochemical distribution of A1Rs in rat forebrain. On light microscopy, A1R immunoreactivity was observed in the cerebral cortex, septum, basal ganglia, hippocampal formation, and thalamus. However, in some regions of the forebrain, regional differences in staining intensity were found as follows: In the cerebral cortex, the strongest immunoreactivity was found in the large pyramidal neurons of layer V. This immunoreactivity was detected in the pyramidal cell bodies, dendrites, and axon initial segments. In the hippocampus, A1R immunoreactivity was detected mainly in the stratum pyramidale. The pyramidal cells in fields CA2–CA3 of the hippocampus were stained more intensely or more clearly than those in field CA1 or the dentate gyrus. More intense A1R immunoreactivity of the apical dendrites was detected in field CA2 compared with other hippocampal fields and the dentate gyrus. Many interneurons of the hippocampus were stained by the 511CA antibody. The subcellular distribution of A1Rs in the forebrain was examined by using a digital deconvolution system and electron microscopy. In the cerebral cortex, the view obtained by removing the background haze by deconvolution revealed that the immunofluoresence‐labeled A1Rs were distributed on the surfaces of the cell bodies and dendrites and in the cytoplasm of layer V neurons as small spots. In field CA1, immunoreactivity was detected in the areas surrounding pyramidal cells. Electron microscopy revealed the presence of A1R‐immunoreactive products in both the presynaptic terminals and the postsynaptic structures. The specific cellular distribution of A1Rs is consistent with the physiological premise that endogeneously released adenosine exerts control over the excitability of forebrain neurons at both presynaptic and postsynaptic sites through A1Rs. J. Comp. Neurol. 411:301–316, 1999.


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.


Neuroscience Research | 2001

Clustering and anchoring mechanisms of molecular constituents of postsynaptic scaffolds in dendritic spines

Tomoaki Shirao; Yuko Sekino

Recent technological progress has yielded great amounts of information about the molecular constituents of postsynaptic scaffolds in the dendritic spine. Actin filaments are major cytoskeletal elements in the dendritic spine, and they functionally interact with neurotransmitter receptors via regulatory actin-binding proteins. Drebrin A and alpha-actinin-2 are two major actin-binding proteins in dendritic spines. In adult brains, they are characteristically concentrated in spines, but not in dendritic shafts or cell bodies. Thus, they are part of a unique postsynaptic scaffold consisting of actin filaments, PSD protein family, and neurotransmitter receptors. Localization of NMDA receptors, actin filaments, and actin-binding proteins in spines changes in parallel with development, and in response to synaptic activity. This raises the possibility that clustering and anchoring of these characteristic molecular constituents at postsynaptic scaffolds play important roles in spine function. This article focuses on the clustering and anchoring mechanisms of NMDA receptors and actin filaments, and the involvement of actin-binding proteins, in dendritic spines, and the way in which characteristic postsynaptic scaffolds are built up.


Molecular and Cellular Neuroscience | 2005

Overexpression of drebrin A in immature neurons induces the accumulation of F-actin and PSD-95 into dendritic filopodia, and the formation of large abnormal protrusions

Toshiyuki Mizui; Hideto Takahashi; Yuko Sekino; Tomoaki Shirao

Drebrin A is a neuron-specific F-actin binding protein, and plays a pivotal role in the spine formation. In this study, we expressed drebrin A tagged with green fluorescent protein (GFP-DA) in hippocampal neurons at 7-9 days in vitro when presynaptic terminals are not fully maturated. GFP-DA was accumulated in dendritic protrusions and formed large abnormal structures. Since these structures were similar to filopodia in terms of lack of MAP2 immunostaining, we named them megapodia meaning large dendritic filopodia. F-actin and PSD-95 were also accumulated in megapodia, and their amounts were significantly correlated with that of GFP-DA. However, the expression of GFP-DA did not result in the promotion of the morphological change from filopodia into spines. These results demonstrate that drebrin A accumulates spine-resident proteins via protein-protein interaction in filopodia, and suggest that the spine formation requires the concurrence of the increase of drebrin-A expression and the functional presynaptic contact.


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.


Histochemistry and Cell Biology | 2007

Many faces of drebrin: from building dendritic spines and stabilizing gap junctions to shaping neurite-like cell processes

Irina Majoul; Tomoaki Shirao; Yuko Sekino; Rainer Duden

In this review we consider the multiple functions of developmentally regulated brain protein (drebrin), an actin-binding protein, in the formation of cellular polarity in different cell types. Drebrin has a well-established role in the morphogenesis, patterning and maintenance of dendritic spines in neurons. We have recently shown that drebrin also stabilizes Connexin-43 containing gap junctions at the plasma membrane. The latest literature and our own data suggest that drebrin may be broadly involved in shaping cell processes and in the formation of stabilized plasma membrane domains, an effect that is likely to be of crucial significance for formation of cell polarity in both neuronal and non-neuronal types.


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 Research | 2004

Antisense knockdown of drebrin A, a dendritic spine protein, causes stronger preference, impaired pre-pulse inhibition, and an increased sensitivity to psychostimulant

Rika Kobayashi; Yuko Sekino; Tomoaki Shirao; Satoshi Tanaka; Taichi Ogura; Ken Inada; Makoto Saji

Drebrin located in dendritic spines regulates their morphological changes and plays a role in the synaptic plasticity via spine function. Reduced drebrin has been found in the brain of patients with Alzheimers disease or Downs syndrome. To examine whether the down-regulation of drebrin protein levels causes deficits in higher brain function, such as memory or cognition, we performed antisense-induced knockdown of drebrin A expression in rat brain using an hemagglutinating virus of Japan (HVJ)-liposome gene transfer technique. We investigated the effects of drebrin in vivo knockdown on spatial memory in a water-maze task, sensorimotor gating in a pre-pulse-inhibition test, adaptive behaviors in an open-field test, and sensitivity to psychostimulant in an amphetamine-induced locomotor response. Rats with drebrin A in vivo knockdown displayed a stronger preference for a previous event due to perseverative behavior, impaired pre-pulse inhibition (PPI), increased locomotor activity, anxiety-like behavior, and an increased sensitivity to psychostimulant, suggesting behaviors related to schizophrenia. These findings indicated that decreased drebrin produces deficits in cognitive function but not in spatial memory, probably via hypofunction of dendritic spines.

Collaboration


Dive into the Yuko Sekino'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

Yasunari Kanda

Safety Pharmacology Society

View shared research outputs
Top Co-Authors

Avatar

Kenichi Kato

Tokyo University of Agriculture

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoichiro Kuroda

Uniformed Services University of the Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge