Updated on 2024/01/31

写真a

 
JUGE Narinobu
 
Organization
Advanced Science Research Center Assistant Professor
Position
Assistant Professor
External link

Research Interests

  • Transporter

  • PfCRT

Research Areas

  • Life Science / Functional biochemistry  / Biochemistry

  • Life Science / Pharmaceutical hygiene and biochemistry  / Biochemistry

Research History

  • Japan Science and Technology Agency

    2014.10 - 2018.3

      More details

  • 海外特別研究員

    2010.4 - 2012.3

      More details

Professional Memberships

 

Papers

  • Structures suggest a mechanism for energy coupling by a family of organic anion transporters. International journal

    Jonathan B Leano, Samir Batarni, Jacob Eriksen, Narinobu Juge, John E Pak, Tomomi Kimura-Someya, Yaneth Robles-Colmenares, Yoshinori Moriyama, Robert M Stroud, Robert H Edwards

    PLoS biology   17 ( 5 )   e3000260   2019.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Members of the solute carrier 17 (SLC17) family use divergent mechanisms to concentrate organic anions. Membrane potential drives uptake of the principal excitatory neurotransmitter glutamate into synaptic vesicles, whereas closely related proteins use proton cotransport to drive efflux from the lysosome. To delineate the divergent features of ionic coupling by the SLC17 family, we determined the structure of Escherichia coli D-galactonate/H+ symporter D-galactonate transporter (DgoT) in 2 states: one open to the cytoplasmic side and the other open to the periplasmic side with substrate bound. The structures suggest a mechanism that couples H+ flux to substrate recognition. A transition in the role of H+ from flux coupling to allostery may confer regulation by trafficking to and from the plasma membrane.

    DOI: 10.1371/journal.pbio.3000260

    PubMed

    researchmap

  • Purification and reconstitution of polyspecific H+/organic cation antiporter human MATE1. International journal

    Tatsuya Kawasaki, Takuya Matsumoto, Yuma Iwai, Mamiyo Kawakami, Narinobu Juge, Hiroshi Omote, Tomohiro Nabekura, Yoshinori Moriyama

    Biochimica et biophysica acta. Biomembranes   1860 ( 11 )   2456 - 2464   2018.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Human MATE1 (multidrug and toxin extrusion 1, hMATE1) is a H+/organic cation (OC) exchanger responsible for the final step of toxic organic cation excretion in the kidney and liver. To investigate the mechanism of transport, we have established an in vitro assay procedure that includes its expression in insect cells, solubilization with octyl glucoside, purification, and reconstitution into liposomes. The resultant proteoliposomes containing hMATE1 as the sole protein component took up radiolabeled tetraethylammonium (TEA) in a ∆pH-dependent and electroneutral fashion. Furthermore, lipid-detergent micelle containing hMATE1 showed ∆pH-dependent TEA binding similar to transport. Mutated hMATE1 with replacement E273Q completely lacked these TEA binding and transport. In the case of divalent substrates, transport was electrogenic. These observations indicate that the stoichiometry of OC/H+ exchange is independent of substrate charge. Purification and reconstitution of hMATE1 is considered to be suitable for understanding the detailed molecular mechanisms of hMATE1. The results suggest that Glu273 of hMATE1 plays essential roles in substrate binding and transport.

    DOI: 10.1016/j.bbamem.2018.07.005

    PubMed

    researchmap

  • Outward open conformation of a Major Facilitator Superfamily multidrug/H+ antiporter provides insights into switching mechanism. International journal

    Kumar Nagarathinam, Yoshiko Nakada-Nakura, Christoph Parthier, Tohru Terada, Narinobu Juge, Frank Jaenecke, Kehong Liu, Yunhon Hotta, Takaaki Miyaji, Hiroshi Omote, So Iwata, Norimichi Nomura, Milton T Stubbs, Mikio Tanabe

    Nature communications   9 ( 1 )   4005 - 4005   2018.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Multidrug resistance (MDR) poses a major challenge to medicine. A principle cause of MDR is through active efflux by MDR transporters situated in the bacterial membrane. Here we present the crystal structure of the major facilitator superfamily (MFS) drug/H+ antiporter MdfA from Escherichia coli in an outward open conformation. Comparison with the inward facing (drug binding) state shows that, in addition to the expected change in relative orientations of the N- and C-terminal lobes of the antiporter, the conformation of TM5 is kinked and twisted. In vitro reconstitution experiments demonstrate the importance of selected residues for transport and molecular dynamics simulations are used to gain insights into antiporter switching. With the availability of structures of alternative conformational states, we anticipate that MdfA will serve as a model system for understanding drug efflux in MFS MDR antiporters.

    DOI: 10.1038/s41467-018-06306-x

    PubMed

    researchmap

  • Efficient Mass Spectral Analysis of Active Transporters Overexpressed in Escherichia coli. International journal

    Mamiyo Kawakami, Narinobu Juge, Yuri Kato, Hiroshi Omote, Yoshinori Moriyama, Takaaki Miyaji

    Journal of proteome research   17 ( 3 )   1108 - 1119   2018.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Structural analysis of purified active membrane proteins can be performed by mass spectrometry (MS). However, no large-scale expression systems for active eukaryotic membrane proteins are available. Moreover, because membrane proteins cannot easily be digested by trypsin and ionized, they are difficult to analyze by MS. We developed a method for mass spectral analysis of eukaryotic membrane proteins combined with an overexpression system in Escherichia coli. Vesicular glutamate transporter 2 (VGLUT2/SLC17A6) with a soluble α-helical protein and histidine tag on the N- and C-terminus, respectively, was overexpressed in E. coli, solubilized with detergent, and purified by Ni-NTA affinity chromatography. Proteoliposomes containing VGLUT2 retained glutamate transport activity. For MS analysis, the detergent was removed from purified VGLUT2 by trichloroacetic acid precipitation, and VGLUT2 was then subjected to reductive alkylation and tryptic digestion. The resulting peptides were detected with 88% coverage by matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) MS with or without liquid chromatography. Vesicular excitatory amino acid transporter and vesicular acetylcholine transporter were also detected with similar coverage by the same method. Thus this methodology could be used to analyze purified eukaryotic active transporters. Structural analysis with chemical modifiers by MS could have applications in functional binding analysis for drug discovery.

    DOI: 10.1021/acs.jproteome.7b00777

    PubMed

    researchmap

  • Neuronal inhibition and seizure suppression by acetoacetate and its analog, 2-phenylbutyrate. International journal

    Atsushi Kadowaki, Nagisa Sada, Narinobu Juge, Ayaka Wakasa, Yoshinori Moriyama, Tsuyoshi Inoue

    Epilepsia   58 ( 5 )   845 - 857   2017.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    OBJECTIVE: The ketogenic diet is clinically used to treat drug-resistant epilepsy. The diet treatment markedly increases ketone bodies (acetoacetate and β-hydroxybutyrate), which work as energy metabolites in the brain. Here, we investigated effects of acetoacetate on voltage-dependent Ca2+ channels (VDCCs) in pyramidal cells of the hippocampus. We further explored an acetoacetate analog that inhibited VDCCs in pyramidal cells, reduced excitatory postsynaptic currents (EPSCs), and suppressed seizures in vivo. METHODS: The effects of acetoacetate and its analogs on VDCCs and EPSCs were evaluated using patch-clamp recordings from CA1 pyramidal cells of mouse hippocampal slices. The in vivo effects of these reagents were also evaluated using a chronic seizure model induced by intrahippocampal injection of kainate. RESULTS: Acetoacetate inhibited VDCCs in pyramidal cells of hippocampal slices, and reduced EPSCs in slices exhibiting epileptiform activity. More potent EPSC inhibitors were then explored by modifying the chemical structure of acetoacetate, and 2-phenylbutyrate was identified as an acetoacetate analog that inhibited VDCCs and EPSCs more potently. Although acetoacetate is known to inhibit vesicular glutamate transporters (VGLUTs), 2-phenylbutyrate did not inhibit VGLUTs, showing that 2-phenylbutyrate is an acetoacetate analog that preferably inhibits VDCCs. In addition, 2-phenylbutyrate markedly reduced EPSCs in slices exhibiting epileptiform activity, and suppressed hippocampal seizures in vivo in a mouse model of epilepsy. The in vivo antiseizure effects of 2-phenylbutyrate were more potent than those of acetoacetate. Finally, intraperitoneal 2-phenylbutyrate was delivered to the brain, and its brain concentration reached the level enough to reduce EPSCs. SIGNIFICANCE: These results demonstrate that 2-phenylbutyrate is an acetoacetate analog that inhibits VDCCs and EPSCs in pyramidal cells, suppresses hippocampal seizures in vivo, and has brain penetration ability. Thus 2-phenylbutyrate provides a useful chemical structure as a lead compound to develop new antiseizure drugs originating from ketone bodies.

    DOI: 10.1111/epi.13718

    PubMed

    researchmap

  • Structure, Function, and Drug Interactions of Neurotransmitter Transporters in the Postgenomic Era. International journal

    Hiroshi Omote, Takaaki Miyaji, Miki Hiasa, Narinobu Juge, Yoshinori Moriyama

    Annual review of pharmacology and toxicology   56   385 - 402   2016

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Vesicular neurotransmitter transporters are responsible for the accumulation of neurotransmitters in secretory vesicles and play essential roles in chemical transmission. The SLC17 family contributes to sequestration of anionic neurotransmitters such as glutamate, aspartate, and nucleotides. Identification and subsequent cellular and molecular biological studies of SLC17 transporters unveiled the principles underlying the actions of these transporters. Recent progress in reconstitution methods in combination with postgenomic approaches has advanced studies on neurotransmitter transporters. This review summarizes the molecular properties of SLC17-type transporters and recent findings regarding the novel SLC18 transporter.

    DOI: 10.1146/annurev-pharmtox-010814-124816

    PubMed

    researchmap

  • Wide expression of type I Na+-phosphate cotransporter 3 (NPT3/SLC17A2), a membrane potential-driven organic anion transporter. International journal

    Natsuko Togawa, Narinobu Juge, Takaaki Miyaji, Miki Hiasa, Hiroshi Omote, Yoshinori Moriyama

    American journal of physiology. Cell physiology   309 ( 2 )   C71-80   2015.7

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Membrane potential (Δψ)-driven and Cl(-)-dependent organic anion transport is a primary function of the solute carrier family 17 (SLC17) transporter family. Although the transport substrates and physiological relevance of the major members are well understood, SLC17A2 protein known to be Na(+)-phosphate cotransporter 3 (NPT3) is far less well characterized. In the present study, we investigated the transport properties and expression patterns of mouse SLC17A2 protein (mNPT3). Proteoliposomes containing the purified mNPT3 protein took up radiolabeled p-aminohippuric acid (PAH) in a Δψ- and Cl(-)-dependent manner. The mNPT3-mediated PAH uptake was inhibited by 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDs) and Evans blue, common inhibitors of SLC17 family members. The PAH uptake was also inhibited by various anionic compounds, such as hydrophilic nonsteroidal anti-inflammatory drugs (NSAIDs) and urate. Consistent with these observations, the proteoliposome took up radiolabeled urate in a Δψ- and Cl(-)-dependent manner. Immunohistochemistry with specific antibodies against mNPT3 combined with RT-PCR revealed that mNPT3 is present in various tissues, including the hepatic bile duct, luminal membranes of the renal urinary tubules, maternal side of syncytiotrophoblast in the placenta, apical membrane of follicle cells in the thyroid, bronchiole epithelial cells in the lungs, and astrocytes around blood vessels in the cerebrum. These results suggested that mNPT3 is a polyspecific organic anion transporter that is involved in circulation of urate throughout the body.

    DOI: 10.1152/ajpcell.00048.2015

    PubMed

    researchmap

  • Plasmodium falciparum chloroquine resistance transporter is a H+-coupled polyspecific nutrient and drug exporter. International journal

    Narinobu Juge, Sawako Moriyama, Takaaki Miyaji, Mamiyo Kawakami, Haruka Iwai, Tomoya Fukui, Nathan Nelson, Hiroshi Omote, Yoshinori Moriyama

    Proceedings of the National Academy of Sciences of the United States of America   112 ( 11 )   3356 - 61   2015.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Extrusion of chloroquine (CQ) from digestive vacuoles through the Plasmodium falciparum CQ resistance transporter (PfCRT) is essential to establish CQ resistance of the malaria parasite. However, the physiological relevance of PfCRT and how CQ-resistant PfCRT gains the ability to transport CQ remain unknown. We prepared proteoliposomes containing purified CQ-sensitive and CQ-resistant PfCRTs and measured their transport activities. All PfCRTs tested actively took up tetraethylammonium, verapamil, CQ, basic amino acids, polypeptides, and polyamines at the expense of an electrochemical proton gradient. CQ-resistant PfCRT exhibited decreased affinity for CQ, resulting in increased CQ uptake. Furthermore, CQ competitively inhibited amino acid transport. Thus, PfCRT is a H(+)-coupled polyspecific nutrient and drug exporter.

    DOI: 10.1073/pnas.1417102112

    PubMed

    researchmap

  • Vesicular GABA transporter (VGAT) transports β-alanine. International journal

    Narinobu Juge, Hiroshi Omote, Yoshinori Moriyama

    Journal of neurochemistry   127 ( 4 )   482 - 6   2013.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Vesicular GABA transporter (VGAT) is expressed in GABAergic and glycinergic neurons, and is responsible for vesicular storage and subsequent exocytosis of these inhibitory amino acids. In this study, we show that VGAT recognizes β-alanine as a substrate. Proteoliposomes containing purified VGAT transport β-alanine using Δψ but not ΔpH as a driving force. The Δψ-driven β-alanine uptake requires Cl(-). VGAT also facilitates Cl(-) uptake in the presence of β-alanine. A previously described VGAT mutant (Glu213Ala) that disrupts GABA and glycine transport similarly abrogates β-alanine uptake. These findings indicated that VGAT transports β-alanine through a mechanism similar to those for GABA and glycine, and functions as a vesicular β-alanine transporter. Vesicular GABA transporter (VGAT) is expressed in GABAergic and glycinergic neurons, and is responsible for vesicular storage and subsequent exocytosis of these inhibitory amino acids. In the present study, we showed that proteoliposomes containing purified VGAT transport β-alanine using Δψ as a driving force. VGAT also facilitates Cl(-) uptake. Our findings indicated that VGAT functions as a vesicular β-alanine transporter.

    DOI: 10.1111/jnc.12393

    PubMed

    researchmap

  • Fabs enable single particle cryoEM studies of small proteins. International journal

    Shenping Wu, Agustin Avila-Sakar, JungMin Kim, David S Booth, Charles H Greenberg, Andrea Rossi, Maofu Liao, Xueming Li, Akram Alian, Sarah L Griner, Narinobu Juge, Yadong Yu, Claudia M Mergel, Javier Chaparro-Riggers, Pavel Strop, Robert Tampé, Robert H Edwards, Robert M Stroud, Charles S Craik, Yifan Cheng

    Structure (London, England : 1993)   20 ( 4 )   582 - 92   2012.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    In spite of its recent achievements, the technique of single particle electron cryomicroscopy (cryoEM) has not been widely used to study proteins smaller than 100 kDa, although it is a highly desirable application of this technique. One fundamental limitation is that images of small proteins embedded in vitreous ice do not contain adequate features for accurate image alignment. We describe a general strategy to overcome this limitation by selecting a fragment antigen binding (Fab) to form a stable and rigid complex with a target protein, thus providing a defined feature for accurate image alignment. Using this approach, we determined a three-dimensional structure of an ∼65 kDa protein by single particle cryoEM. Because Fabs can be readily generated against a wide range of proteins by phage display, this approach is generally applicable to study many small proteins by single particle cryoEM.

    DOI: 10.1016/j.str.2012.02.017

    PubMed

    researchmap

  • Vesicular neurotransmitter transporter: bioenergetics and regulation of glutamate transport. International journal

    Hiroshi Omote, Takaaki Miyaji, Narinobu Juge, Yoshinori Moriyama

    Biochemistry   50 ( 25 )   5558 - 65   2011.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Glutamate plays essential roles in chemical transmission as a major excitatory neurotransmitter. The accumulation of glutamate in secretory vesicles is mediated by vesicular glutamate transporters (VGLUTs) that together with the driving electrochemical gradient of proteins influence the subsequent quantum release of glutamate and the function of higher-order neurons. The vesicular content of glutamate is well correlated with membrane potential (Δψ), which suggests that Δψ determines the vesicular glutamate concentration. The transport of glutamate into secretory vesicles is highly dependent on Cl(-). This anion stimulates glutamate transport but is inhibitory at higher concentrations. Accumulating evidence indicates that Cl(-) regulates glutamate transport through control of VGLUT activity and the H(+) electrochemical gradient. Recently, a comprehensive study demonstrated that Cl(-) regulation of VGLUT is competitively inhibited by metabolic intermediates such as ketone bodies. It also showed that ketone bodies are effective in controlling epilepsy. These results suggest a correlation between metabolic state and higher-order brain function. We propose a novel function for Cl(-) as a fundamental regulator for signal transmission.

    DOI: 10.1021/bi200567k

    PubMed

    researchmap

  • Metabolic control of vesicular glutamate transport and release. International journal

    Narinobu Juge, John A Gray, Hiroshi Omote, Takaaki Miyaji, Tsuyoshi Inoue, Chiaki Hara, Hisayuki Uneyama, Robert H Edwards, Roger A Nicoll, Yoshinori Moriyama

    Neuron   68 ( 1 )   99 - 112   2010.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Fasting has been used to control epilepsy since antiquity, but the mechanism of coupling between metabolic state and excitatory neurotransmission remains unknown. Previous work has shown that the vesicular glutamate transporters (VGLUTs) required for exocytotic release of glutamate undergo an unusual form of regulation by Cl(-). Using functional reconstitution of the purified VGLUTs into proteoliposomes, we now show that Cl(-) acts as an allosteric activator, and the ketone bodies that increase with fasting inhibit glutamate release by competing with Cl(-) at the site of allosteric regulation. Consistent with these observations, acetoacetate reduced quantal size at hippocampal synapses and suppresses glutamate release and seizures evoked with 4-aminopyridine in the brain. The results indicate an unsuspected link between metabolic state and excitatory neurotransmission through anion-dependent regulation of VGLUT activity.

    DOI: 10.1016/j.neuron.2010.09.002

    PubMed

    researchmap

  • Vesicular inhibitory amino acid transporter is a Cl-/gamma-aminobutyrate Co-transporter. International journal

    Narinobu Juge, Akiko Muroyama, Miki Hiasa, Hiroshi Omote, Yoshinori Moriyama

    The Journal of biological chemistry   284 ( 50 )   35073 - 8   2009.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    The vesicular inhibitory amino acid transporter (VIAAT) is a synaptic vesicle protein responsible for the vesicular storage of gamma-aminobutyrate (GABA) and glycine which plays an essential role in GABAergic and glycinergic neurotransmission. The transport mechanism of VIAAT remains largely unknown. Here, we show that proteoliposomes containing purified VIAAT actively took up GABA upon formation of membrane potential (Deltapsi) (positive inside) but not DeltapH. VIAAT-mediated GABA uptake had an absolute requirement for Cl(-) and actually accompanied Cl(-) movement. Kinetic analysis indicated that one GABA molecule and two Cl(-) equivalents were transported during one transport cycle. VIAAT in which Glu(213) was specifically mutated to alanine completely lost the ability to take up both GABA and Cl(-). Essentially the same results were obtained with glycine, another substrate of VIAAT. These results demonstrated that VIAAT is a vesicular Cl(-) transporter that co-transports Cl(-) with GABA or glycine in a Deltapsi dependent manner. It is concluded that Cl(-) plays an essential role in vesicular storage of GABA and glycine.

    DOI: 10.1074/jbc.M109.062414

    PubMed

    researchmap

  • Identification of a vesicular nucleotide transporter. International journal

    Keisuke Sawada, Noriko Echigo, Narinobu Juge, Takaaki Miyaji, Masato Otsuka, Hiroshi Omote, Akitsugu Yamamoto, Yoshinori Moriyama

    Proceedings of the National Academy of Sciences of the United States of America   105 ( 15 )   5683 - 6   2008.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    ATP is a major chemical transmitter in purinergic signal transmission. Before secretion, ATP is stored in secretory vesicles found in purinergic cells. Although the presence of active transport mechanisms for ATP has been postulated for a long time, the proteins responsible for its vesicular accumulation remains unknown. The transporter encoded by the human and mouse SLC17A9 gene, a novel member of an anion transporter family, was predominantly expressed in the brain and adrenal gland. The mouse and bovine counterparts were associated with adrenal chromaffin granules. Proteoliposomes containing purified transporter actively took up ATP, ADP, and GTP by using membrane potential as the driving force. The uptake properties of the reconstituted transporter were similar to that of the ATP uptake by synaptic vesicles and chromaffin granules. Suppression of endogenous SLC17A9 expression in PC12 cells decreased exocytosis of ATP. These findings strongly suggest that SLC17A9 protein is a vesicular nucleotide transporter and should lead to the elucidation of the molecular mechanism of ATP secretion in purinergic signal transmission.

    DOI: 10.1073/pnas.0800141105

    PubMed

    researchmap

  • [Molecular mechanism of vesicular glutamate transporter].

    Hiroshi Omote, Narinobu Juge

    Seikagaku. The Journal of Japanese Biochemical Society   79 ( 10 )   956 - 60   2007.10

     More details

    Language:Japanese   Publishing type:Research paper (scientific journal)  

    PubMed

    researchmap

  • Vesicular glutamate transporter contains two independent transport machineries. International journal

    Narinobu Juge, Yumi Yoshida, Shouki Yatsushiro, Hiroshi Omote, Yoshinori Moriyama

    The Journal of biological chemistry   281 ( 51 )   39499 - 506   2006.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Vesicular glutamate transporters (VGLUTs) are responsible for the vesicular storage of l-glutamate and play an essential role in glutamatergic signal transmission in the central nervous system. The molecular mechanism of the transport remains unknown. Here, we established a novel in vitro assay procedure, which includes purification of wild and mutant VGLUT2 and their reconstitution with purified bacterial F(o)F(1)-ATPase (F-ATPase) into liposomes. Upon the addition of ATP, the proteoliposomes facilitated l-glutamate uptake in a membrane potential (DeltaPsi)-dependent fashion. The ATP-dependent l-glutamate uptake exhibited an absolute requirement for approximately 4 mm Cl(-), was sensitive to Evans blue, but was insensitive to d,l-aspartate. VGLUT2s with mutations in the transmembrane-located residues Arg(184), His(128), and Glu(191) showed a dramatic loss in l-glutamate transport activity, whereas Na(+)-dependent inorganic phosphate (P(i)) uptake remained comparable to that of the wild type. Furthermore, P(i) transport did not require Cl(-) and was not inhibited by Evans blue. Thus, VGLUT2 appears to possess two intrinsic transport machineries that are independent of each other: a DeltaPsi-dependent l-glutamate uptake and a Na(+)-dependent P(i) uptake.

    PubMed

    researchmap

  • Secretion of L-glutamate from osteoclasts through transcytosis. International journal

    Riyo Morimoto, Shunsuke Uehara, Shouki Yatsushiro, Narinobu Juge, Zhaolin Hua, Shigenori Senoh, Noriko Echigo, Mitsuko Hayashi, Toshihide Mizoguchi, Tadashi Ninomiya, Nobuyuki Udagawa, Hiroshi Omote, Akitsugu Yamamoto, Robert H Edwards, Yoshinori Moriyama

    The EMBO journal   25 ( 18 )   4175 - 86   2006.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Osteoclasts are involved in the catabolism of the bone matrix and eliminate the resulting degradation products through transcytosis, but the molecular mechanism and regulation of transcytosis remain poorly understood. Upon differentiation, osteoclasts express vesicular glutamate transporter 1 (VGLUT1), which is essential for vesicular storage and subsequent exocytosis of glutamate in neurons. VGLUT1 is localized in transcytotic vesicles and accumulates L-glutamate. Osteoclasts secrete L-glutamate and the bone degradation products upon stimulation with KCl or ATP in a Ca2+-dependent manner. KCl- and ATP-dependent secretion of L-glutamate was absent in osteoclasts prepared from VGLUT1-/- knockout mice. Osteoclasts express mGluR8, a class III metabotropic glutamate receptor. Its stimulation by a specific agonist inhibits secretion of L-glutamate and bone degradation products, whereas its suppression by a specific antagonist stimulates bone resorption. Finally, it was found that VGLUT1-/- mice develop osteoporosis. Thus, in bone-resorbing osteoclasts, L-glutamate and bone degradation products are secreted through transcytosis and the released L-glutamate is involved in autoregulation of transcytosis. Glutamate signaling may play an important role in the bone homeostasis.

    PubMed

    researchmap

  • Vesicular storage and secretion of L-glutamate from glucagon-like peptide 1-secreting clonal intestinal L cells. International journal

    Shunsuke Uehara, Sun-Kyung Jung, Riyo Morimoto, Shigeo Arioka, Takaaki Miyaji, Narinobu Juge, Miki Hiasa, Kahori Shimizu, Akinori Ishimura, Masato Otsuka, Akitsugu Yamamoto, Pierre Maechler, Yoshinori Moriyama

    Journal of neurochemistry   96 ( 2 )   550 - 60   2006.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Vesicular glutamate transporter (VGLUT) is responsible for the vesicular storage of l-glutamate, and plays an essential role in glutamate-mediated intercellular signal transmission in the CNS and in some neuroendocrine cells. Intestinal L cells are the glucose-responsive neuroendocrine cells responsible for the secretion of glucagon-like peptide 1 (GLP-1). We have shown that intestinal L cells express VGLUT2, a VGLUT isoform, which suggests that L cells secrete L-glutamate. In the present study, we investigated this possibility using GLUTag mouse clonal L cells. RT-PCR and northern blot analyses revealed expression of the VGLUT1 and VGLUT2 genes, but not of the VGLUT3 gene. Western blot analysis revealed immunological counterparts for VGLUT2, whereas an immunological counterpart of VGLUT1 was not detected. Indirect immunofluorescence microscopy revealed a punctate distribution of VGLUT2 immunoreactivity throughout the cells, which co-localized with GLP-1. Double-labeling immunoelectronmicroscopy confirmed the association of VGLUT2 with GLP-1-containing secretory granules. The membrane fraction exhibited ATP-dependent L-glutamate uptake, which was sensitive to bafilomycin A1 (a vacuolar proton ATPase inhibitor) and Evans blue (a VGLUT inhibitor) but insensitive to D,L-aspartate. Upon depolarization with KCl, GLUTag cells secreted appreciable amounts of L-glutamate and GLP-1. D-Glucose and methyl-alpha-D-glucopyranoside, stimulators of exocytosis of GLP-1, also triggered the secretion of L-glutamate. The L-glutamate secretion was partially dependent on Ca2+ and sensitive to bafilomycin A1. These results demonstrated that GLUTag cells stored L-glutamate in secretory granules and secreted it with GLP-1 by exocytosis. As GLUTag cells and intestinal L cells express kainate receptors and plasma membrane glutamate transporters, these results support the concept of L-glutamate-mediated intercellular signaling in the vicinity of intestinal L cells.

    PubMed

    researchmap

▼display all

MISC

  • Identification and characterization of a vesicular nucleotide transporter

    Keisuke Sawada, Miki Hiasa, Noriko Echigo, Narinobu Juge, Takaaki Miyaji, Hiroshi Omote, Yoshinori Moriyama

    NEUROSCIENCE RESEARCH   65   S77 - S77   2009

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:ELSEVIER IRELAND LTD  

    DOI: 10.1016/j.neures.2009.09.280

    Web of Science

    researchmap

  • Characterization of transport properties of purified vesicular inhibitory amino acid transporter (VIAAT)

    Narinobu Juge, Akiko Muroyama, Miki Hiasa, Hiroshi Omote, Yoshinori Moriyama

    NEUROSCIENCE RESEARCH   65   S77 - S77   2009

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:ELSEVIER IRELAND LTD  

    DOI: 10.1016/j.neures.2009.09.281

    Web of Science

    researchmap

  • Identification and characterization of a vesicular nucleotide transporter

    Keisuke Sawada, Noriko Echigo, Narinobu Juge, Takaaki Miyaji, Miki Hiasa, Masato Otsuka, Hiroshi Omote, Yoshinori Moriyama

    YAKUGAKU ZASSHI-JOURNAL OF THE PHARMACEUTICAL SOCIETY OF JAPAN   128   65 - 66   2008

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:PHARMACEUTICAL SOC JAPAN  

    Web of Science

    researchmap

  • L-glutamate signaling regulates transcytotic vesicle function in osteoclasts.

    S. Uehara, R. Morimoto, S. Yatsushiro, N. Juge, M. Hayashi, S. Senoh, T. Mizoguchi, T. Ninomiya, N. Udagawa, Z. Hua, H. Omote, A. Yamamoto, R. H. Edwards, Y. Moriyama

    JOURNAL OF BONE AND MINERAL RESEARCH   21   S401 - S401   2006.9

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:AMER SOC BONE & MINERAL RES  

    Web of Science

    researchmap

Presentations

  • トランスポーターの生化学的・構造生物学的展開 Invited

    樹下成信

    GEヘルスケア・ジャパン「初めてのライフサイエンス基礎講座」  2015.5.15 

     More details

    Language:Japanese   Presentation type:Public lecture, seminar, tutorial, course, or other speech  

    researchmap

Research Projects

  • MFS型多剤排出輸送体の多剤認識と輸送機構の解明

    Grant number:19H03186  2019.04 - 2023.03

    日本学術振興会  科学研究費助成事業  基盤研究(B)

    田辺 幹雄, 野村 紀通, 樹下 成信

      More details

    Grant amount:\17290000 ( Direct expense: \13300000 、 Indirect expense:\3990000 )

    細菌が薬剤耐性を持つ原因の一つに多剤排出トランスポーターによる投与薬剤の排出が挙げられる。本課題ではグラム陰性菌の細胞質からペリプラズムに薬剤を排出するMajor facilitator superfamily(MFS)型の多剤排出トランスポーターの薬剤認識とその輸送機構の解明を目指しており、その目標達成のために特に2つの研究テーマを掲げている。
    1)MFS型-多剤排出トランスポーターのモデルタンパク質であるMdfAを使い生化学、構造解析、シミュレーションを用いて、薬剤分子の認識やその排出の輸送サイクルを明らかにする。
    2) 新規の病原菌由来の輸送体の構造解析を行い、薬剤輸送機構の解明を目指す。
    本年度の研究実績として、
    1)については、MdfAを認識する4つの抗体を用いてそれらがどのような構造状態を認識しているかをクライオ電顕を用いて確認することを目指した。前年度は分解能が10オングストローム程度で留まっていたデータについて、様々な条件で精製し直し、グリッドを作成したものの、分解能は10オングストロームから僅かに更新されたのみであった。MDシミュレーションを中心とした解析では現在も進行中であり、内開きから中間体を経て外開きの構造変化を順次追っている最中であるがその検証には時間を有する。MdfAと抗体のクロスリンク実験は進行中で、より安定な複合体の取得に取り組んでいる。
    2)前年度にタンパク精製に成功したS.aureusのトランスポーターにおいては阻害剤存在下での精製に成功し、結晶を得ることができた。回折能は10オングストローム程度であったため、こちらも阻害剤の存在下でのタンパク質の構造を認識する抗体の取得について実験が進んでいる。

    researchmap

  • 小胞型神経伝達物質トランスポーターの脂質制御機構の解明

    2018.04 - 2022.03

    KAKEN  JSPS 

    Narinobu Juge

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 小胞容量測定系の確立

    2016.04 - 2018.03

    KKEN  JSPS 

    Narinobu Juge

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • グルタミン酸のシナプス小胞充填機構の構造生物学的展開

    2014.10 - 2018.03

    JST  PREST 

    Narinobu Juge

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • 小胞型グルタミン酸トランスポーターの構造生物学

    2013.04 - 2015.03

    KAKEN  JSPS 

    Narinobu Juge

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

 

Class subject in charge

  • Life science for drug discovery (2023academic year) Third semester  - 木5~6

  • Drug/biomembrane interactions I (2023academic year) special  - その他

  • Drug/biomembrane interactions II (2023academic year) special  - その他

  • Experimental Molecular Biology (2023academic year) Second semester  - その他5~9

  • Experimental Molecular Biology (2023academic year) Second semester  - その他5~9

  • Statistics for Bioscience 1 (2023academic year) Third semester  - 火3~4

  • Statistics for Bioscience 1 (2023academic year) Third semester  - 火3~4

  • Statistics for Bioscience A (2023academic year) Third semester  - 火3~4

  • Statistics for Bioscience A (2023academic year) Third semester  - 火3~4

  • Cell Biology 1 (2023academic year) Third semester  - 火5~6

  • Cell Biology 1 (2023academic year) Third semester  - 火5~6

  • Cell Biology A (2023academic year) Third semester  - 火5~6

  • Cell Biology A (2023academic year) Third semester  - 火5~6

  • Pharmaceutical Laws and Administration 1 (2023academic year) 1st semester  - 火1~2

  • Pharmaceutical Laws and Administration 1 (2023academic year) 1st semester  - 火1~2

  • Pharmaceutical Laws and Administration 2 (2023academic year) Second semester  - 火1~2

  • Pharmaceutical Laws and Administration 2 (2023academic year) Second semester  - 火1~2

  • Practice in Fundamental Pharmaceutical Sciences III (2023academic year) Second semester  - その他5~9

  • Practice in Fundamental Pharmaceutical Sciences III (2023academic year) Second semester  - その他5~9

  • Practice in Fundamental Pharmaceutical Sciences III (2023academic year) Second semester  - その他5~9

  • Practice in Fundamental Pharmaceutical Sciences III (2023academic year) Second semester  - その他5~9

  • Life science for drug discovery (2022academic year) Third semester  - 木5~6

  • Drug/biomembrane interactions I (2022academic year) special  - その他

  • Drug/biomembrane interactions II (2022academic year) special  - その他

  • Cell Biology 1 (2022academic year) Third semester  - 火5~6

  • Cell Biology 1 (2022academic year) Third semester  - 火5~6

  • Cell Biology A (2022academic year) Third semester  - 火5~6

  • Cell Biology A (2022academic year) Third semester  - 火5~6

  • Pharmaceutical Laws and Administration (2022academic year) 1st and 2nd semester  - 火3~4

  • Pharmaceutical Laws and Administration 1 (2022academic year) 1st semester  - 火3~4

  • Pharmaceutical Laws and Administration 1 (2022academic year) 1st semester  - 火3~4

  • Pharmaceutical Laws and Administration 2 (2022academic year) Second semester  - 火3~4

  • Pharmaceutical Laws and Administration 2 (2022academic year) Second semester  - 火3~4

  • Practice in Fundamental Pharmaceutical Sciences III (2022academic year) Second semester  - その他5~9

  • Practice in Fundamental Pharmaceutical Sciences III (2022academic year) Second semester  - その他5~9

  • Practice in Fundamental Pharmaceutical Sciences III (2022academic year) Second semester  - その他5~9

  • Practice in Fundamental Pharmaceutical Sciences III (2022academic year) Second semester  - その他5~9

  • Life science for drug discovery (2021academic year) Third semester  - 木5~6

  • Drug/biomembrane interactions I (2021academic year) special  - その他

  • Drug/biomembrane interactions II (2021academic year) special  - その他

  • Cell Biology 1 (2021academic year) Third semester  - 火5,火6

  • Cell Biology 1 (2021academic year) Third semester  - 火5,火6

  • Cell Biology A (2021academic year) Third semester  - 火5,火6

  • Cell Biology A (2021academic year) Third semester  - 火5,火6

  • Practice in Fundamental Pharmaceutical Sciences III (2021academic year) Second semester  - その他6~9

  • Practice in Fundamental Pharmaceutical Sciences III (2021academic year) Second semester  - その他6~9

  • Practice in Fundamental Pharmaceutical Sciences III (2021academic year) Second semester  - その他6~9

  • Practice in Fundamental Pharmaceutical Sciences III (2021academic year) Second semester  - その他6~9

  • Life science for drug discovery (2020academic year) Third semester  - 木5,木6

  • Drug/biomembrane interactions I (2020academic year) special  - その他

  • Drug/biomembrane interactions II (2020academic year) special  - その他

  • Cell Biology 1 (2020academic year) Third semester  - 火5,火6

  • Cell Biology 1 (2020academic year) Third semester  - 火5,火6

  • Practice in Fundamental Pharmaceutical Sciences III (2020academic year) Second semester  - その他

  • Practice in Fundamental Pharmaceutical Sciences III (2020academic year) Second semester  - その他

▼display all