Network


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

Hotspot


Dive into the research topics where Takeshi Kanno is active.

Publication


Featured researches published by Takeshi Kanno.


Journal of Bone and Mineral Metabolism | 2002

Transgenic mice overexpressing soluble osteoclast differentiation factor (sODF) exhibit severe osteoporosis

Atsuko Mizuno; Takeshi Kanno; Masaki Hoshi; Osamu Shibata; Kazuki Yano; Nobuaki Fujise; Masahiko Kinosaki; Kyoji Yamaguchi; Eisuke Tsuda; Hisataka Yasuda; Kanji Higashio

Abstract. Osteoclast differentiation factor, ODF, also called RANKL, TRANCE, or OPGL, is a key molecule for osteoclast differentiation and activation, and is thought to act as a membrane-associated molecule in bone remodeling. Recent study suggested that soluble ODF (sODF) released from T cells also has some roles in bone resorption. To investigate the physiological and pathological function of sODF, we generated two types of transgenic mice overexpressing sODF. Mice overexpressing sODF ubiquitously from the early developmental stage died at the late fetal stage. The other type of mice, expressing sODF only in the liver after birth, grew to maturity with normal body size and weight. However, they exhibited a marked decrease in bone mineral density with aging compared with their non-transgenic littermates, and in addition, the strength of their femurs was extremely reduced. Histological analysis showed that the trabecular bone mass was decreased at 6 weeks of age and was sparse at age 3–4 months. The number of osteoclasts was significantly increased, while the number of osteoblasts was not altered on the surface of young trabecular bone. These results indicate that excessive production of sODF causes osteoporosis by accelerated osteoclastogenesis. The transgenic mouse overexpressing sODF in the liver could serve as a useful animal model for studying bone remodeling and evaluating therapeutic agents for osteoporosis.


Glia | 2002

A new neuromodulatory pathway with a glial contribution mediated via A2a adenosine receptors

Tomoyuki Nishizaki; Kaoru Nagai; Tamotsu Nomura; Hitoshi Tada; Takeshi Kanno; Hidetoshi Tozaki; X.X. Li; Takeshi Kondoh; Noriko Kodama; E. Takahashi; Norio Sakai; Kohichi Tanaka; Naoaki Saito

A low concentration (10 nM) of adenosine potentiated hippocampal neuronal activity via A2a adenosine receptors without affecting presynaptic glutamate release or postsynaptic glutamatergic conductance. Adenosine inhibited glutamate uptake through the glial glutamate transporter, GLT‐1, via A2a adenosine receptors. In addition, adenosine stimulated GLT‐1‐independent glutamate release from astrocytes, possibly in response to a rise in intracellular Ca2+, via A2a adenosine receptors involving PKA activation. Those adenosine actions could lead to an increase in synaptic glutamate concentrations responsible for the potentiation of hippocampal neuronal activity. The results of the present study thus represent a novel neuromodulatory pathway with a glial contribution, bearing both inhibition of GLT‐1 function and stimulation of glial glutamate release, as mediated via A2a adenosine receptors. GLIA 39:133–147, 2002.


Journal of Lipid Research | 2006

The linoleic acid derivative DCP-LA selectively activates PKC-ϵ, possibly binding to the phosphatidylserine binding site

Takeshi Kanno; Hideyuki Yamamoto; Takahiro Yaguchi; Rika Hi; Takeshi Mukasa; Hirokazu Fujikawa; Tetsu Nagata; Satoshi Yamamoto; Akito Tanaka; Tomoyuki Nishizaki

This study examined the effect of 8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid (DCP-LA), a newly synthesized linoleic acid derivative with cyclopropane rings instead of cis-double bonds, on protein kinase C (PKC) activity. In the in situ PKC assay with reverse-phase high-performance liquid chromatography, DCP-LA significantly activated PKC in PC-12 cells in a concentration-dependent (10 nM–100 μM) manner, with the maximal effect at 100 nM, and the DCP-LA effect was blocked by GF109203X, a PKC inhibitor, or a selective inhibitor peptide of the novel PKC isozyme PKC-ϵ. Furthermore, DCP-LA activated PKC in HEK-293 cells that was inhibited by the small, interfering RNA against PKC-ϵ. In the cell-free PKC assay, of the nine isozymes examined here, DCP-LA most strongly activated PKC-ϵ, with >7-fold potency over other PKC isozymes, in the absence of dioleoyl-phosphatidylserine and 1,2-dioleoyl-sn-glycerol; instead, the DCP-LA action was inhibited by dioleoyl-phosphatidylserine. DCP-LA also activated PKC-γ, a conventional PKC, but to a much lesser extent compared with that for PKC-ϵ, by a mechanism distinct from PKC-ϵ activation. Thus, DCP-LA serves as a selective activator of PKC-ϵ, possibly by binding to the phosphatidylserine binding site on PKC-ϵ. These results may provide fresh insight into lipid signaling in PKC activation.


Neuroscience | 2005

The linoleic acid derivative FR236924 facilitates hippocampal synaptic transmission by enhancing activity of presynaptic α7 acetylcholine receptors on the glutamatergic terminals

Satoshi Yamamoto; Takeshi Kanno; Tetsu Nagata; Takahiro Yaguchi; A. Tanaka; Tomoyuki Nishizaki

The present study aimed at understanding the effect of FR236924, a newly synthesized linoleic acid derivative with cyclopropane rings instead of cis-double bonds, on hippocampal synaptic transmission in both the in vitro and in vivo systems. FR236924 increased the rate of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor-mediated miniature excitatory postsynaptic currents, without affecting the amplitude, triggered by nicotine in CA1 pyramidal neurons of rat hippocampal slices, that is inhibited by GF109203X, a selective protein kinase C (PKC) inhibitor or alpha-bungarotoxin, an inhibitor of alpha7 acetylcholine (ACh) receptors. FR236924 stimulated glutamate release from rat hippocampal slices and in the hippocampus of freely behaving rats, and the effect was also inhibited by GF109203X or alpha-bungarotoxin. FR236924 induced a transient huge potentiation followed by a long-lasting potentiation in the slope of field excitatory postsynaptic potentials recorded from the CA1 region of rat hippocampal slices, and the latter effect was blocked by GF109203X or alpha-bungarotoxin. Likewise, the compound persistently facilitated hippocampal synaptic transmission in the CA1 region of the intact rat hippocampus. It is concluded from these results that FR236924 stimulates glutamate release by functionally targeting presynaptic alpha7 ACh receptors on the glutamatergic terminals under the influence of PKC, responsible for the facilitatory action on hippocampal synaptic transmission. This may provide evidence for a link between cis-unsaturated free fatty acids and presynaptic alpha7 ACh receptors in hippocampal synaptic plasticity.


Pain | 2006

Modulation of P2X receptors via adrenergic pathways in rat dorsal root ganglion neurons after sciatic nerve injury

Keishi Maruo; Hideyuki Yamamoto; Satoshi Yamamoto; Tetsu Nagata; Hirokazu Fujikawa; Takeshi Kanno; Takahiro Yaguchi; Soji Maruo; Shinichi Yoshiya; Tomoyuki Nishizaki

Abstract The present study examined noradrenaline‐induced modulation of ATP‐evoked currents in dorsal root ganglion (DRG) neurons after sciatic nerve injury (transection). ATP (10 μM) generated fast/mixed type of whole‐cell membrane currents, possibly as mediated via P2X3/P2X3‐like receptors, and slow type of the currents, possibly as mediated via P2X2/3 receptors, in acutely dissociated L4/5 DRG neurons, without significant difference between sham and injury group. For sham group, noradrenaline (10 μM) enhanced fast/mixed type of ATP‐evoked currents in ipsilateral DRG neurons, that is not inhibited by H‐7, a broad inhibitor of protein kinases, but otherwise it had no effect on slow type of the currents. For injury group, noradrenaline (10 μM) significantly potentiated slow type of ATP‐evoked currents in ipsilateral DRG neurons, that is abolished by H‐7 or GF109203X, a selective inhibitor of protein kinase C (PKC), while it depressed fast/mixed type of the currents. In the analysis of real‐time reverse transcription‐polymerase chain reaction, an increase in the mRNAs for α1b, α2a, α2d, and β2 adrenergic receptors was found with the ipsilateral DRGs after sciatic nerve injury. Collectively, the results of the present study suggest that noradrenaline potentiates P2X2/3 receptor currents by activating PKC via α1 adrenergic receptors linked to Gq protein, perhaps dominantly α1b adrenergic receptors, in DRG neurons after sciatic nerve injury. This may account for a nociceptive pathway in response to noradrenergic sprouting after peripheral nerve injury.


Biochemical and Biophysical Research Communications | 2002

The inhibitory and facilitatory actions of amyloid-β peptides on nicotinic ACh receptors and AMPA receptors

Hidetoshi Tozaki; Akira Matsumoto; Takeshi Kanno; Kaoru Nagai; Tetsu Nagata; Satoshi Yamamoto; Tomoykui Nishizaki

The present study investigated the effects of amyloid-beta peptides on nicotinic ACh receptors (Torpedo, alpha 4 beta 2, and alpha 7 receptors) and AMPA receptors expressed in Xenopus oocytes by monitoring whole-cell membrane currents. Ten-minutes treatment with amyloid-beta(1-42) (1 microM) inhibited Torpedo ACh receptor currents, reaching 53% of original levels 30 min after treatment. Amyloid-beta(1-40) inhibited the currents in a dose-dependent manner (0.1-10 microM) during treatment, gradually reversing after treatment. Amyloid-beta(1-40) and amyloid-beta(1-42) (0.1 microM) depressed alpha 4 beta 2 receptor currents to each 69% and 62% of original levels at 10-min treatment and lesser depression was obtained with alpha 7 receptors. Amyloid-beta(1-42) (0.1 microM) did not significantly inhibit AMPA receptor currents, but amyloid-beta(1-40) (0.1 microM) potentiated the currents to 145-191% of original levels. Amyloid-beta peptides, thus, exert their diverse actions on nicotinic ACh receptors and AMPA receptors, and the inhibitory actions on nicotinic ACh receptors may account for the deterioration of learning and memory in Alzheimers disease.


Brain Research | 2004

Interleukin-18 stimulates synaptically released glutamate and enhances postsynaptic AMPA receptor responses in the CA1 region of mouse hippocampal slices.

Takeshi Kanno; Tetsu Nagata; Satoshi Yamamoto; Haruki Okamura; Tomoyuki Nishizaki

The present study examined the effects of the proinflammatory cytokine interleukin-18 (IL-18) on mouse hippocampal synaptic transmission. IL-18 (100 ng/ml) significantly increased amplitude and frequency of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor-mediated miniature excitatory postsynaptic currents (AMPA-mEPSCs), that are monitored from CA1 pyramidal neurons of mouse hippocampal slices. IL-18 (100 ng/ml) enhanced slope of basal field excitatory postsynaptic potentials (fEPSPs) that are recorded from the CA1 region of mouse hippocampal slices. There was no significant difference in the expression of Schaffer collateral/CA1 long-term potentiation (LTP) between in the presence and absence of IL-18, although IL-18 tended to inhibit saturation levels of the potentiation induced by tetanic stimulation in a dose-dependent manner at concentrations ranged from 10 ng/ml to 1 microg/ml. Paired-pulse facilitation in the presence of IL-18 (100 ng/ml) was not influenced after tetanic stimulation, while that in the absence of IL-18 was depressed. The results of the present study, thus, suggest that IL-18 stimulates synaptically released glutamate and enhances postsynaptic AMPA receptor responses in CA1 pyramidal neurons of mouse hippocampal slices, thereby facilitating basal hippocampal synaptic transmission without affecting the LTP.


Journal of Endocrinology | 2013

PI3 kinase directly phosphorylates Akt1/2 at Ser473/474 in the insulin signal transduction pathway

Ayako Tsuchiya; Takeshi Kanno; Tomoyuki Nishizaki

Insulin stimulated translocation of the glucose transporter GLUT4 from the cytosol to the plasma membrane in a concentration (1 nM–1 μM)-dependent manner and increased glucose uptake in 3T3-L1 adipocytes. Insulin-induced GLUT4 translocation to the cell surface was prevented by the phosphoinositide 3 kinase (PI3K) inhibitor wortmannin, the 3-phosphoinositide-dependent protein kinase 1 (PDK1) inhibitor BX912 or the Akt1/2 inhibitor MK2206, and by knocking-down PI3K, PDK1 or Akt1/2. Insulin increased phosphorylation of Akt1/2 at Thr308/309 and Ser473/474, to activate Akt1/2, in the adipocytes. Insulin-induced phosphorylation of Akt1/2 was suppressed by wortmannin and knocking-down PI3K, while no significant inhibition of the phosphorylation was obtained with BX912 or knocking-down PDK1. In the cell-free Akt assay, PI3K phosphorylated Akt1 both at Thr308 and Ser473 and Akt2 at Ser474 alone. In contrast, PDK1 phosphorylates Akt1 at Thr308 and Akt2 at Thr309. The results of this study indicate that PI3K activates Akt1, independently of PDK1, and Akt2 by cooperating with PDK1 in the insulin signal transduction pathway linked to GLUT4 translocation.


Biochemical and Biophysical Research Communications | 2008

The NMDA receptor NR2A subunit regulates proliferation of MKN45 human gastric cancer cells

Kanako Watanabe; Takeshi Kanno; Tadayuki Oshima; Hiroto Miwa; Chikara Tashiro; Tomoyuki Nishizaki

The present study investigated proliferation of MKN28 and MKN45 human gastric cancer cells regulated by the N-methyl-d-aspartate (NMDA) receptor subunit. The NMDA receptor antagonist dl-2-amino-5-phosphonovaleric acid (AP5) inhibited proliferation of MKN45 cells, but not MKN28 cells. Of the NMDA subunits such as NR1, NR2 (2A, 2B, 2C, and 2D), and NR3 (3A and 3B), all the NMDA subunit mRNAs except for the NR2B subunit mRNA were expressed in both MKN28 and MKN45 cells. MKN45 cells were characterized by higher expression of the NR2A subunit mRNA and lower expression of the NR1 subunit mRNA, but MKN28 otherwise by higher expression of the NR1 subunit mRNA and lower expression of the NR2A subunit mRNA. MKN45 cell proliferation was also inhibited by silencing the NR2A subunit-targeted gene. For MKN45 cells, AP5 or knocking-down the NR2A subunit increased the proportion of cells in the G(1) phase of cell cycling and decreased the proportion in the S/G(2) phase. The results of the present study, thus, suggest that blockage of NMDA receptors including the NR2A subunit suppresses MKN45 cell proliferation due to cell cycle arrest at the G(1) phase; in other words, the NR2A subunit promotes MKN45 cell proliferation by accelerating cell cycling.


Journal of Cellular Physiology | 2011

Sphingosine induces apoptosis in hippocampal neurons and astrocytes by activating caspase-3/-9 via a mitochondrial pathway linked to SDK/14-3-3 protein/Bax/cytochrome c

Takeshi Kanno; Tomoyuki Nishizaki

The present study examined sphingosine‐induced apoptosis in cultured rat hippocampal neurons and astrocytes. Sphingosine induced apoptosis in a concentration (1–100 µM)‐dependent manner, that is inhibited by the PKC‐δ inhibitor rottlerin, and a similar effect was obtained with the sphingosine kinase inhibitors, to raise intracellular sphingosine concentrations. Sphingosine increased presence of sphingosine‐dependent protein kinase (SDK), and the effect was suppressed by rottlerin. Sphingosine increased phosphorylated 14‐3‐3 protein, thereby transforming the protein from a dimeric structure into a monomeric structure. Sphingosine accumulated Bax in the mitochondria and stimulated cytochrome c release into the cytosol, and those effects were inhibited by rottlerin. Sphingosine disrupted mitochondrial membrane potentials, that was abolished by silencing the PKC‐δ‐targeted gene. Moreover, sphingosine activated caspase‐9 and the effector caspase‐3 in a PKC‐δ‐dependent manner. Taken together, the results of the present study indicate that sphingosine activates SDK, produced through proteolytic processing of an active form of PKC‐δ, to phosphorylate 14‐3‐3 protein and transform into a monomeric structure, causing Bax dissociation from 14‐3‐3 protein and accumulation in the mitochondria, which perturbs mitochondrial membrane potentials allowing cytochrome c release into the cytosol, to activate caspase‐9 and the effector caspase‐3, responsible for apoptosis in hippocampal neurons and astrocytes. J. Cell. Physiol. 226: 2329–2337, 2011.

Collaboration


Dive into the Takeshi Kanno's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ayako Tsuchiya

Hyogo College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Akinobu Gotoh

Hyogo College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Akito Tanaka

Hyogo University of Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Takashi Nakano

Hyogo College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tadashi Shimizu

Hyogo University of Health Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tetsu Nagata

Hyogo College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hisao Nagaya

Hyogo College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge