Network


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

Hotspot


Dive into the research topics where Yuki Kambe is active.

Publication


Featured researches published by Yuki Kambe.


Journal of Neuroscience Research | 2005

An increase in intracellular free calcium ions by nicotinic acetylcholine receptors in a single cultured rat cortical astrocyte

Hirotaka Oikawa; Noritaka Nakamichi; Yuki Kambe; Masato Ogura; Yukio Yoneda

Neuronal nicotinic acetylcholine receptors (nAChRs) are composed of an assembly between at least seven alpha (α2–α7, α9) and three beta (β2–β4) subunits in mammals. The addition of 50 mM KCl or 1 mM nicotine immediately increased the number of cells with high fluorescence intensity in rat cortical astrocytes on fluo‐3 fluorescence measurement. Nicotine was effective at increasing the fluorescence intensity in astrocytes cultured for 2 days after replating, but not in those used 1 or 5 days after replating, without markedly affecting the cellular viability irrespective of the exposure period. Nicotine markedly increased the fluorescence intensity in a concentration‐dependent manner at a concentration range of 10–100 μM in cultured astrocytes when analyzed on a responsive single cell. In these responsive single cells, the increase by nicotine was significantly prevented by the heteromeric α4/β2 subtype antagonist dihydro‐β‐erythroidine and the homomeric α7 subtype antagonist methyllycaconitine, as well as by nifedipine and EGTA but not thapsigargin. Methyllycaconitine failed to inhibit further the increase by nicotine in the presence of nifedipine, however, whereas the expression of mRNA was seen for all mammalian neuronal nAChR subunits in cultured rat cortical astrocytes as well as neurons. These results suggest that nicotine may increase intracellular free Ca2+ through the influx of extracellular Ca2+ across L‐type voltage‐gated Ca2+ channels rather than Ca2+ release from intracellular stores, in a manner related to the α4/β2 and/or α7 nAChR channels functionally expressed in cultured rat cortical astrocytes.


Journal of Neurochemistry | 2005

Protection by exogenous pyruvate through a mechanism related to monocarboxylate transporters against cell death induced by hydrogen peroxide in cultured rat cortical neurons.

Noritaka Nakamichi; Yuki Kambe; Hirotaka Oikawa; Masato Ogura; Katsura Takano; Keisuke Tamaki; Maki Inoue; Eiichi Hinoi; Yukio Yoneda

In cortical neurons cultured for 3 or 9 days in vitro (DIV), exposure to hydrogen peroxide (H2O2) led to a marked decrease in cell viability in a concentration‐dependent manner at a concentration range of 10 µm to 1 mm irrespective of the duration between 6 and 24 h. However, H2O2 was more potent in decreasing cellular viability in cortical neurons cultured for 9 DIV than in those for 3 DIV. Pyruvate was effective in preventing the neuronal cell death at 1 mm even when added 1–3 h after the addition of H2O2. Semi‐quantitative RT–PCR and western blotting analyses revealed significantly higher expression of both mRNA and protein for a particular monocarboxylate transporter (MCT) in neurons cultured for 9 DIV than in those for 3 DIV. A specific inhibitor of MCT significantly attenuated the neuroprotection by pyruvate in neurons cultured for 9 DIV, without markedly affecting that in neurons cultured for 3 DIV. These results suggest that vulnerability to H2O2 may at least in part involve expression of particular MCT isoforms responsible for the bi‐directional transport of pyruvate across cell surfaces in cultured rat cortical neurons.


Journal of Neuroscience Research | 2009

Possible Protection by Notoginsenoside R1 against Glutamate Neurotoxicity Mediated by N-methyl-D-aspartate Receptors Composed of an NR1/NR2B Subunit Assembly

Bin Gu; Noritaka Nakamichi; Wensheng Zhang; Yukary Nakamura; Yuki Kambe; Ryo Fukumori; Kazuhiro Takuma; Kiyofumi Yamada; Takeshi Takarada; Hideo Taniura; Yukio Yoneda

Notoginsenoside R1 (NTR1) is the main active ingredient in Panax notoginseng, a herbal medicine widely used in Asia for years. The purpose of this study was to investigate pharmacological properties of NTR1 on neurotoxicity of glutamate (Glu) in primary cultured mouse cortical neurons along with its possible mechanism of action. Wefound that NTR1 significantly protected neurons from the loss of cellular viability caused by brief exposure to 10 μM Glu for 1 hr in a dose‐dependent manner at concentrations from 0.1 to 10 μM, without affecting the viability alone. NTR1 significantly inhibited the increased number of cells positive to propidium iodide (PI) staining, increase of intracellular free Ca2+ ions, overproduction of intracellular reactive oxygen species, and depolarization of mitochondrial membrane potential in cultured neurons exposed to Glu, in addition to blocking decreased Bcl‐2 and increased Bax expression levels. We further evaluated the target site at which NTR1 protects neurons from Glu toxicity by using the acquired expression strategy of N‐methyl‐D‐aspartate (NMDA) receptor subunits in human embryonic kidney 293 cells. We found that 10 μM NTR1 protected NR1/NR2B subunit expressing cells from cell death by 100 μM NMDA, but not cells expressing NR1/NR2A subunits, when determined by PI staining. These results suggest that NTR1 may preferentially protect neurons from Glu excitotoxicity mediated by NMDA receptor composed of an NR1/NR2B subunit assembly in the brain.


Biochemical and Biophysical Research Communications | 2012

N-arachidonoyl glycine induces macrophage apoptosis via GPR18

Rina Takenouchi; Kazuhiko Inoue; Yuki Kambe; Atsuro Miyata

N-arachidonoyl glycine (NAGly), a member of lipoamino acids, was reported to exhibit anti-inflammatory effects in experimental ear edema or peritonitis. However the underlying mechanisms have not been clarified so far. In this study, we attempt to investigate the effects of NAGly on macrophages, including the relevant signaling pathways. NAGly potently induced apoptosis in mouse macrophage-derived cell line, RAW264.7. Pretreatment with inhibitors for MEK and p38 MAPK prevented the apoptosis induced by NAGly, although NAGly activated ERK1/2, p38 MAPK and JNK. Further, we focused on implication of GPR18, one of the orphan G protein-coupled receptors, because NAGly has been reported as a candidate ligand for GPR18. Pretreatment with pertussis toxin or siRNA to knock down the expression of GPR18 significantly attenuated the apoptosis induced by NAGly. In mouse peritoneal macrophages, the expression of GPR18 mRNA was elevated in proinflammatory stimulated macrophages but not in anti-inflammatory stimulated macrophages; consistently, NAGly remarkably reduced cell viability of the former, as compared to the latter. These results suggest that NAGly might be involved in function of macrophages through GPR18.


Journal of Neurochemistry | 2008

Insensitivity to glutamate neurotoxicity mediated by NMDA receptors in association with delayed mitochondrial membrane potential disruption in cultured rat cortical neurons

Yuki Kambe; Noritaka Nakamichi; Danko Georgiev; Nobuhiro Nakamura; Hideo Taniura; Yukio Yoneda

We have attempted to elucidate mechanisms underlying differential vulnerability to glutamate (Glu) using cultured neurons prepared from discrete structures of embryonic rat brains. Brief exposure to Glu led to a significant decrease in the mitochondrial activity in hippocampal neurons cultured for 9 or 12 days at 10 μM to 1 mM with an apoptosis‐like profile, without markedly affecting that in cortical neurons. Brief exposure to Glu also increased lactate dehydrogenase release along with a marked decrease in the number of cells immunoreactive for a neuronal marker protein in hippocampal, but not cortical, neurons. Similar insensitivity was seen to the cytotoxicity by NMDA, but not to that by tunicamycin, 2,4‐dinitrophenol, hydrogen peroxide or A23187, in cortical neurons. However, NMDA was more efficient in increasing intracellular free Ca2+ levels in cortical neurons than in hippocampal neurons. Antagonists for neuroprotective metabotropic Glu receptors failed to significantly affect the insensitivity to Glu, while NMDA was more effective in disrupting mitochondrial membrane potentials in hippocampal than cortical neurons. These results suggest that cortical neurons would be insensitive to the apoptotic neurotoxicity mediated by NMDA receptors through a mechanism related to mitochondrial membrane potentials, rather than intracellular free Ca2+ levels, in the rat brain.


Experimental Cell Research | 2008

A critical importance of polyamine site in NMDA receptors for neurite outgrowth and fasciculation at early stages of P19 neuronal differentiation

Danko Georgiev; Hideo Taniura; Yuki Kambe; Takeshi Takarada; Yukio Yoneda

We have investigated the role of N-methyl-d-aspartate receptors (NMDARs) and gamma-aminobutyric acid receptors type A (GABA(A)Rs) at an early stage of P19 neuronal differentiation. The subunit expression was profiled in 24-hour intervals with RT-PCR and functionality of the receptors was verified via fluo-3 imaging of Ca(2+) dynamics in the immature P19 neurons showing that both NMDA and GABA excite neuronal bodies, but only polyamine-site sensitive NMDAR stimulation leads to enhanced Ca(2+) signaling in the growth cones. Inhibition of NR1/NR2B NMDARs by 1 muM ifenprodil severely impaired P19 neurite extension and fasciculation, and this negative effect was fully reversible by polyamine addition. In contrast, GABA(A)R antagonism by a high dose of 200 microM bicuculline had no observable effect on P19 neuronal differentiation and fasciculation. Except for the differential NMDAR and GABA(A)R profiles of Ca(2+) signaling within the immature P19 neurons, we have also shown that inhibition of NR1/NR2B NMDARs strongly decreased mRNA level of NCAM-180, which has been previously implicated as a regulator of neuronal growth cone protrusion and neurite extension. Our data thus suggest a critical role of NR1/NR2B NMDARs during the process of neuritogenesis and fasciculation of P19 neurons via differential control of local growth cone Ca(2+) surges and NCAM-180 signaling.


Journal of Neuroscience Research | 2005

Counteraction by repetitive daily exposure to static magnetism against sustained blockade of N-methyl-D-aspartate receptor channels in cultured rat hippocampal neurons.

Takao Hirai; Hideo Taniura; Yasuaki Goto; Keisuke Tamaki; Hirotaka Oikawa; Yuki Kambe; Masato Ogura; Yu Ohno; Takeshi Takarada; Yukio Yoneda

In rat hippocampal neurons cultured with the antagonist for N‐methyl‐D‐aspartate (NMDA) receptors dizocilpine (MK‐801) for 8 days in vitro (DIV), a significant decrease was seen in the expression of microtubule‐associated protein‐2 (MAP‐2) as well as mRNA for both brain‐derived neurotrophic factor (BDNF) and growth‐associated protein‐43 (GAP‐43), in addition to decreased viability. MK‐801 not only decreased the expression of the NR1 subunit of NMDA receptors but also increased NR2A expression, without affecting NR2B expression. Repetitive daily exposure to static magnetic fields at 100 mT for 15 min led to a decrease in the expression of MAP‐2, without significantly affecting cell viability or the expression of neuronal nuclei (NeuN) and GAP‐43. However, the repetitive magnetism prevented decreases in both BDNF mRNA and MAP‐2 and additionally increased the expression of NR2A subunit, without altering NR1 expression in neurons cultured in the presence of MK‐801. Repetitive magnetism was also effective in preventing the decrease by MK‐801 in the ability of NMDA to increase intracellular free Ca2+ ions, without affecting the decrease in the maximal response. These results suggest that repetitive magnetism may at least in part counteract the neurotoxicity of MK‐801 through modulation of the expression of particular NMDA receptor subunits in cultured rat hippocampal neurons.


Current Neurovascular Research | 2004

Relevant Modulation by Ferrous Ions of N-Methyl-D-Aspartate Receptors in Ischemic Brain Injuries

Noritaka Nakamichi; Hirotaka Oikawa; Yuki Kambe; Yukio Yoneda

Activation of the N-methyl-D-aspartate (NMDA) receptor would induce rapid opening of an ion channel permeable to Ca2+ ions across cell membranes, followed by an increase in the concentration of free Ca2+ ions in the cytoplasm and subsequent signaling cascade from the cytoplasm to the nucleus for consolidation of a transient extracellular signal carried by L-glutamate in the central nervous system. Both neuronal plasticity and cell death have been shown to involve intracellular free Ca2+ ions incorporated through this receptor-operated cation channel in the brain. On the other hand, iron is also abundant in the brain, with an essential role in mechanisms underlying maintenance of cellular integrity and function. Ferrous ions are believed to participate in neuronal cell death through generation of reactive oxygen species in ischemic brain injuries, for instance, while ferrous but not ferric ions are shown to block the influx of Ca2+ ions across NMDA receptor channels in cultured neurons. In this review article, we will summarize the possible relationship between iron and NMDA receptor channels in mechanisms associated with neuronal cell death in brains with ischemia.


Journal of Cerebral Blood Flow and Metabolism | 2014

C-type natriuretic peptide modulates permeability of the blood-brain barrier

Manoj Bohara; Yuki Kambe; Tetsuya Nagayama; Hiroshi Tokimura; Kazunori Arita; Atsuro Miyata

C-type natriuretic peptide (CNP) is abundant in brain and is reported to exert autocrine function in vascular cells, but its effect on blood–brain barrier (BBB) permeability has not been clarified yet. Here, we examined this effect. Transendothelial electrical resistance (TEER) of in vitro BBB model, composed of bovine brain microvascular endothelial cells and astrocytes, was significantly dose dependently decreased by CNP (1, 10, and 100 nmol/L). C-type natriuretic peptide treatment reduced both the messenger RNA (mRNA) and protein expressions of tight junction (TJ) protein zonula occludens-1 (ZO-1). The effects on TEER, mRNA, and protein expressions of ZO-1 were mimicked by cyclic GMP (cGMP) analog 8-bromo-cGMP (1 μmol/L) and reversed by protein kinase G (PKG) inhibitor Rp-8-CPT-cGMPS (100 μmol/L), thus implying the role of PKG and cGMP signaling in BBB function. Transcription factor JunD knockdown by small interfering RNA resulted in no change of permeability by CNP. In vivo study of mouse brain by fluorimetric analysis with intravenous administration of sodium fluorescein (40 mg/kg) also showed a significant increase in BBB permeability by CNP (10 nmol/kg, intravenously). These findings suggest that CNP modulates the BBB permeability by altering ZO-1 expression.


Neurochemistry International | 2011

A possible pivotal role of mitochondrial free calcium in neurotoxicity mediated by N-methyl-d-aspartate receptors in cultured rat hippocampal neurons

Yuki Kambe; Noritaka Nakamichi; Takeshi Takarada; Ryo Fukumori; Ryota Nakazato; Eiichi Hinoi; Yukio Yoneda

We have previously shown that mitochondrial membrane potential disruption is involved in mechanisms underlying differential vulnerabilities to the excitotoxicity mediated by N-methyl-d-aspartate (NMDA) receptors between primary cultured neurons prepared from rat cortex and hippocampus. To further elucidate the role of mitochondria in the excitotoxicity after activation of NMDA receptors, neurons were loaded with the fluorescent dye calcein diffusible in the cytoplasm and organelles for determination of the activity of mitochondrial permeability transition pore (mPTP) responsible for the leakage of different mitochondrial molecules. The addition of CoCl(2) similarly quenched the intracellular fluorescence except mitochondria in both cultured neurons, while further addition of NMDA led to a leakage of the dye into the cytoplasm in hippocampal neurons only. An mPTP inhibitor prevented the NMDA-induced loss of viability in hippocampal neurons, while an activator of mPTP induced a similarly potent loss of viability in cortical and hippocampal neurons. Although NMDA was more effective in increasing rhodamine-2 fluorescence as a mitochondrial calcium indicator in hippocampal than cortical neurons, a mitochondrial calcium uniporter inhibitor significantly prevented the NMDA-induced loss of viability in hippocampal neurons. Expression of mRNA was significantly higher for the putative uniporter uncoupling protein-2 in hippocampal than cortical neurons. These results suggest that mitochondrial calcium uniporter would be at least in part responsible for the NMDA neurotoxicity through a mechanism relevant to promotion of mPTP orchestration in hippocampal neurons.

Collaboration


Dive into the Yuki Kambe'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