九州大学 研究者情報
論文一覧
神野 尚三(じんの しようぞう) データ更新日:2024.04.25

教授 /  医学研究院 基礎医学部門 生体制御学


原著論文
1. Jun Yamada, Shoichiro Maeda, Miori Tojo, Miyuki Hayashida, Kyoko M Iinuma, Shozo Jinno, Altered regulation of oligodendrocytes associated with parvalbumin neurons in the ventral hippocampus underlies fear generalization in male mice, Neuropsychopharmacology , 10.1038/s41386-023-01611-6, 48, 11, 1668-1679, 2023.10.
2. Risako Fujikawa, Shozo Jinno, Identification of hyper-ramified microglia in the CA1 region of the mouse hippocampus potentially associated with stress resilience, Eur J Neurosci, 10.1111/ejn.15812, 56, 8, 5137-5153, 2022.10.
3. Shoichiro Maeda, Jun Yamada, Kyoko M Iinuma, Satomi Nadanaka, Hiroshi Kitagawa, Shozo Jinno, Chondroitin sulfate proteoglycan is a potential target of memantine to improve cognitive function via the promotion of adult neurogenesis, Br J Pharmacol, 10.1111/bph.15920, 179, 20, 4857-4877, 2022.10.
4. Jun Yamada, Shoichiro Maeda, Mariko Soya, Hidefumi Nishida, Kyoko M Iinuma, Shozo Jinno, Alleviation of cognitive deficits via upregulation of chondroitin sulfate biosynthesis by lignan sesamin in a mouse model of neuroinflammation, J Nutr Biochem, 10.1016/j.jnutbio.2022.109093, 108, 109093, 2022.10.
5. Risako Fujikawa, Jun Yamada, Kyoko M Iinuma, Shozo Jinno, Phytoestrogen genistein modulates neuron-microglia signaling in a mouse model of chronic social defeat stress., Neuropharmacology, 10.1016/j.neuropharm.2021.108941, 206, 108941-108941, 2022.03, Microglia, resident immune cells in the brain, are shown to mediate the crosstalk between psychological stress and depression. Interestingly, increasing evidence indicates that sex hormones, particularly estrogen, are involved in the regulation of immune system. In this study, we aimed to understand the potential effects of chronic social defeat stress (CSDS) and genistein (GEN), an estrogenic compound of the plant origin, on neuron-microglia interactions in the mouse hippocampus. The time spent in the avoidance zone in the social interaction test was increased by CSDS 1 day after the exposure, while the avoidance behavior returned to control levels 14 days after the CSDS exposure. Similar results were obtained from the elevated plus-maze test. However, the immobility time in the forced swim test was increased by CSDS 14 days after the exposure, and the depression-related behavior was in part alleviated by GEN. The numerical densities of microglia in the hippocampus were increased by CSDS, and they were decreased by GEN. The voxel densities of synaptic structures and synaptic puncta colocalized with microglia were decreased by CSDS, and they were increased by GEN. Neither CSDS nor GEN affected the gene expressions of major pro-inflammatory cytokines. Conversely, the expression levels of genes related to neurotrophic factors were decreased by CSDS, and they were partially reversed by GEN. These findings show that GEN may in part alleviate stress-related symptoms, and the effects of GEN may be associated with the modulation of neuron-microglia signaling via chemokines and neurotrophic factors in the hippocampus..
6. Tomohiro Ohgomori, Shozo Jinno, Signal Transducer and Activator of Transcription 3 Activation in Hippocampal Neural Stem Cells and Cognitive Deficits in Mice Following Short-term Cuprizone Exposure., Neuroscience, 10.1016/j.neuroscience.2021.07.031, 472, 90-102, 2021.09, Recent studies have emphasized that adult hippocampal neurogenesis impairment may be associated with cognitive problems. Because cuprizone (CPZ), a copper-chelating reagent, was shown to decrease the production of new neurons, we aimed to further understand the involvement of adult hippocampal neurogenesis impairment in cognitive function by using a short-term (2-week) CPZ exposure paradigm. The CPZ-fed mice showed cognitive deficits, i.e., impaired sensorimotor gating and reduced social novelty preference, compared to normal-fed mice. Although a long-term (e.g., 5-week) CPZ exposure paradigm was found to cause demyelination, we encountered that the labeling for myelin in the hippocampus was unaffected by 2-week CPZ exposure. The densities of neuronal progenitor cells (NPCs) and newborn granule cells (NGCs) were lower in CPZ-fed mice than in normal-fed mice, while those of neural stem cells (NSCs) were comparable between groups. We then examined whether short-term CPZ exposure might induce activation of signal transducer and activator of transcription 3 (STAT3), which plays a major role in cytokine receptor signaling. The densities of phosphorylated STAT3-positive (pSTAT3+) NSCs were higher in CPZ-fed mice than in normal-fed mice, while those of pSTAT3+ NPCs/NGCs were very low in both groups. Interestingly, the densities of bromodeoxyuridine-positive (BrdU+) NSCs were higher in CPZ-fed mice than in normal-fed mice 2 weeks after BrdU injection, while those of BrdU+ NPCs/NGCs were lower in CPZ-fed mice than in normal-fed mice. These findings suggest that short-term CPZ exposure inhibits differentiation of NSCs into NPCs via activation of STAT3, which may in part underlie cognitive deficits..
7. Tomohiro Ohgomori, Kyoko Iinuma, Jun Yamada, Shozo Jinno, A unique subtype of ramified microglia associated with synapses in the rat hippocampus., The European journal of neuroscience, 10.1111/ejn.15330, 54, 3, 4740-4754, 2021.08, To date, a number of studies have reported the heterogeneity of activated microglia. However, there is increasing evidence suggests that ramified, so-called resting, microglia may also be heterogeneous, and they may play diverse roles in normal brain homeostasis. Here, we found that both 5D4 keratan sulfate epitope-positive (5D4+ ) and 5D4-negative (5D4- ) microglia coexisted in the hippocampus of normal rats, while all microglia were negative for the 5D4 epitope in the hippocampus of normal mice. We thus aimed to determine the potential heterogeneity of microglia related to the 5D4 epitope in the normal rat hippocampus. The optical disector analysis showed that the densities of 5D4+ microglia were higher in the stratum oriens of the CA3 region than in other layers and regions. Although both 5D4+ and 5D4- microglia exhibited a ramified morphology, the three-dimensional reconstruction analysis showed that the node numbers, end numbers, and complexity of processes were higher in 5D4+ than in 5D4- microglia. The linear discriminant analysis showed that 5D4+ and 5D4- microglia can be classified into distinct morphometric subtypes. The ratios of contact between synaptic boutons and microglial processes were higher in 5D4+ than in 5D4- microglia. The gene expressions of pro-inflammatory cytokine interleukin-1β and purinergic receptor P2Y12 (P2Y12 R) were higher in 5D4+ than in 5D4- microglia. Together, these results indicate that at least two different subtypes of ramified microglia coexist in the normal rat hippocampus and also suggest that 5D4+ microglia may represent a unique subtype associated with synapses..
8. Risako Fujikawa, Jun Yamada, Shozo Jinno, Subclass imbalance of parvalbumin-expressing GABAergic neurons in the hippocampus of a mouse ketamine model for schizophrenia, with reference to perineuronal nets, Schizophrenia Research, 10.1016/j.schres.2020.11.016, 2020.11.
9. Tomohiro Ohgomori, Shozo Jinno, Modulation of neuropathology and cognitive deficits by lipopolysaccharide preconditioning in a mouse pilocarpine model of status epilepticus, Neuropharmacology, 10.1016/j.neuropharm.2020.108227, 176, 108227-108227, 2020.10.
10. Jun Yamada, Chihiro Sato, Kohtarou Konno, Masahiko Watanabe, Shozo Jinno, PSA-NCAM Colocalized with Cholecystokinin-Expressing Cells in the Hippocampus Is Involved in Mediating Antidepressant Efficacy, The Journal of Neuroscience, 10.1523/jneurosci.1779-19.2019, 40, 4, 825-842, 2020.01, The extracellular glycan polysialic acid linked to neural cell adhesion molecule (PSA-NCAM) is principally expressed in the developing brain and the adult neurogenic regions. Although colocalization of PSA-NCAM with cholecystokinin (CCK) was found in the adult brain, the role of PSA-NCAM remains unclear. In this study, we aimed to elucidate the functional significance of PSA-NCAM in the CA1 region of the male mouse hippocampus. Combined fluorescence in situ hybridization and immunohistochemistry showed that few vesicular glutamate transporter 3-negative/CCK-positive (VGluT3-/CCK+) cells were colocalized with PSA-NCAM, but most of the VGluT3+/CCK+ cells were colocalized with PSA-NCAM. The somata of PSA-NCAM+/CCK+ cells were highly innervated by serotonergic boutons than those of PSA-NCAM-/CCK+ cells. The expression ratios of 5-HT3A receptors and p11, a serotonin receptor-interacting protein, were higher in PSA-NCAM+/CCK+ cells than in PSA-NCAM-/CCK+ cells. Pharmacological digestion of PSA-NCAM impaired the efficacy of antidepressant fluoxetine (FLX), a selective serotonin reuptake inhibitor, but not the efficacy of benzodiazepine anxiolytic diazepam. A Western blot showed that restraint stress decreased the expressions of p11 and mature brain-derived neurotrophic factor (BDNF), and FLX increased them. Interestingly, the FLX-induced elevation of expression of p11, but not mature BDNF, was impaired by the digestion of PSA-NCAM. Quantitative reverse transcription-polymerase chain reaction showed that restraint stress reduced the expression of polysialyltransferase ST8Sia IV and FLX elevated it. Collectively, PSA-NCAM colocalized with VGluT3+/CCK+ cells in the CA1 region of the hippocampus may play a unique role in the regulation of antidepressant efficacy via the serotonergic pathway.SIGNIFICANCE STATEMENT Polysialic acid (PSA) is composed of eight or more α2,8-linked sialic acids. Here, we examined the functional significance of polysialic acid linked to the neural cell adhesion molecule (PSA-NCAM) in the adult mouse hippocampus. Few vesicular glutamate transporter 3-negative/cholecystokinin-positive (VGluT3-/CCK+) cells were colocalized with PSA-NCAM, but most of the VGluT3+/CCK+ cells were colocalized with PSA-NCAM. The expression ratios of 5-HT3A receptors and p11, a serotonin receptor-interacting protein, were higher in PSA-NCAM+/CCK+ cells than in PSA-NCAM-/CCK+ cells. The efficacy of antidepressants, but not anxiolytics, was impaired by the digestion of PSA-NCAM. The antidepressant-induced increase in p11 expression was inhibited following PSA-NCAM digestion. We hence hypothesize that PSA-NCAM colocalized with VGluT3+/CCK+ cells may play a unique role in regulating antidepressant efficacy..
11. Tomohiro Ohgomori, Shozo Jinno, The expression of keratan sulfate reveals a unique subset of microglia in the mouse hippocampus after pilocarpine‐induced status epileptics, The Journal of Comparative Neurology, 10.1002/cne.24734, 528, 1, 18-35, 2020.01.
12. Jun Yamada, Shozo Jinno, Promotion of synaptogenesis and neural circuit development by exosomes, Annals of Translational Medicine, 10.21037/atm.2019.09.154, 7, S8, S323-S323, 2019.12.
13. Jun Yamada, Shozo Jinno, Potential link between antidepressant-like effects of ketamine and promotion of adult neurogenesis in the ventral hippocampus of mice, Neuropharmacology, 10.1016/j.neuropharm.2019.107710, 158, 107710-107710, 2019.11.
14. Tomohiro Ohgomori, Shozo Jinno, Cuprizone-induced demyelination in the mouse hippocampus is alleviated by phytoestrogen genistein, Toxicology and Applied Pharmacology, 10.1016/j.taap.2018.11.009, 363, 98-110, 2019.01.
15. Jun Yamada, Satomi Nadanaka, Hiroshi Kitagawa, Kosei Takeuchi, Shozo Jinno, Increased Synthesis of Chondroitin Sulfate Proteoglycan Promotes Adult Hippocampal Neurogenesis in Response to Enriched Environment, The Journal of Neuroscience, 10.1523/jneurosci.0632-18.2018, 38, 39, 8496-8513, 2018.09.
16. Tomohiro Ohgomori, Ryo Yamasaki, Jun-ichi Kira, Shozo Jinno, Upregulation of Vesicular Glutamate Transporter 2 and STAT3 Activation in the Spinal Cord of Mice Receiving 3,3′-Iminodipropionitrile, Neurotoxicity Research, 10.1007/s12640-017-9822-x, 33, 4, 768-780, 2018.05.
17. Takayasu Mishima, Manami Deshimaru, Takuya Watanabe, Kaori Kubota, Mariko Kinoshita-Kawada, Junichi Yuasa-Kawada, Kotaro Takasaki, Yoshinari Uehara, Shozo Jinno, Katsunori Iwasaki, Yoshio Tsuboi, Behavioral defects in a DCTN1G71A transgenic mouse model of Perry syndrome, Neuroscience Letters, 10.1016/j.neulet.2017.12.038, 666, 98-103, 2018.02.
18. Jun Yamada, Tomohiro Ohgomori, Shozo Jinno, Alterations in expression of Cat‐315 epitope of perineuronal nets during normal ageing, and its modulation by an open‐channel NMDA receptor blocker, memantine, The Journal of Comparative Neurology, 10.1002/cne.24198, 525, 9, 2035-2049, 2017.06.
19. Jun Yamada, Shozo Jinno, Molecular heterogeneity of aggrecan-based perineuronal nets around five subclasses of parvalbumin-expressing neurons in the mouse hippocampus, The Journal of Comparative Neurology, 10.1002/cne.24132, 525, 5, 1234-1249, 2017.04.
20. Hisataka Fujimoto, Kotaro Konno, Masahiko Watanabe, Shozo Jinno, Late postnatal shifts of parvalbumin and nitric oxide synthase expression within the GABAergic and glutamatergic phenotypes of inferior colliculus neurons, The Journal of Comparative Neurology, 10.1002/cne.24104, 525, 4, 868-884, 2017.03.
21. Jun Yamada, Shozo Jinno, Aging of hippocampal neurogenesis and soy isoflavone, Oncotarget, 10.18632/oncotarget.13534, 7, 51, 83835-83836, 2016.12.
22. Jun Yamada, Jun Hatabe, Kaori Tankyo, Shozo Jinno, Cell type- and region-specific enhancement of adult hippocampal neurogenesis by daidzein in middle-aged female mice, Neuropharmacology, 10.1016/j.neuropharm.2016.08.036, 111, 92-106, 2016.12.
23. Shozo Jinno, Aging affects new cell production in the adult hippocampus: A quantitative anatomic review, Journal of Chemical Neuroanatomy, 10.1016/j.jchemneu.2015.10.009, 76, 64-72, 2016.10.
24. Tomohiro Ohgomori, Jun Yamada, Hideyuki Takeuchi, Kenji Kadomatsu, Shozo Jinno, Comparative morphometric analysis of microglia in the spinal cord of SOD1G93Atransgenic mouse model of amyotrophic lateral sclerosis, European Journal of Neuroscience, 10.1111/ejn.13227, 43, 10, 1340-1351, 2016.05.
25. Hisataka Fujimoto, Tomohiro Ohgomori, Kentaro Abe, Kenji Uchimura, Kenji Kadomatsu, Shozo Jinno, Time-dependent localization of high- and low-sulfated keratan sulfates in the song nuclei of developing zebra finches, European Journal of Neuroscience, 10.1111/ejn.13073, 42, 9, 2716-2725, 2015.11.
26. Jun Yamada, Shozo Jinno, Subclass-specific formation of perineuronal nets around parvalbumin-expressing GABAergic neurons in Ammon's horn of the mouse hippocampus, The Journal of Comparative Neurology, 10.1002/cne.23712, 523, 5, 790-804, 2015.04.
27. K Kubo, S Jinno, K Nariyama, S Komune, Stability of the synaptic structure in the hippocampus of BALB/c mice with allergic rhinitis, The Journal of Laryngology & Otology, 10.1017/s0022215114002400, 129, S2, S56-S61, 2015.03, AbstractObjective:The aim of this study was to determine whether allergic rhinitis can induce structural changes in the synapse formation in the hippocampus of BALB/c mice immunocytochemically.

Methods:Allergic rhinitis was induced in mice by two intra-peritoneal injections of ovalbumin administered with a one-week interval. After two weeks, the sensitised mice were challenged with an intra-nasal injection of ovalbumin for two weeks. To analyse the hippocampal synaptic structures, sections were immunostained with antibodies against glutamic acid decarboxylase 65 and glutamic acid decarboxylase 67 (for γ-aminobutyric acid-ergic terminals), synaptophysin (for glutamatergic and γ-aminobutyric acid-ergic terminals) and spinophilin (for dendritic spines). The number of nasal rubbing movements was significantly greater in the allergic rhinitis mice than in the control mice. However, the expression patterns of the four above-mentioned synaptic markers in the hippocampus showed no detectable difference between the allergic rhinitis and control mice.

Results and Conclusion:These data indicate that the synaptic structure in the hippocampus might remain unaltered in allergic rhinitis patients.

.
28. J. Yamada, T. Ohgomori, S. Jinno, Perineuronal nets affect parvalbumin expression in GABAergic neurons of the mouse hippocampus, European Journal of Neuroscience, 10.1111/ejn.12792, 41, 3, 368-378, 2015.02.
29. Jun Yamada, Shozo Jinno, Age-related differences in oligodendrogenesis across the dorsal-ventral axis of the mouse hippocampus, Hippocampus, 10.1002/hipo.22287, 24, 8, 1017-1029, 2014.08.
30. Jun Yamada, Shozo Jinno, S100A6 (calcyclin) is a novel marker of neural stem cells and astrocyte precursors in the subgranular zone of the adult mouse hippocampus, Hippocampus, 10.1002/hipo.22207, 24, 1, 89-101, 2014.01.
31. J. Yamada, S. Jinno, Spatio-temporal differences in perineuronal net expression in the mouse hippocampus, with reference to parvalbumin, Neuroscience, 10.1016/j.neuroscience.2013.08.061, 253, 368-379, 2013.12.
32. Shozo Jinno, Quantitative Assessment of Hippocampus Architecture Using the Optical Disector, Neurostereology, 10.1002/9781118444177.ch9, 113-127, 2013.11.
33. Jun Yamada, Shozo Jinno, Novel objective classification of reactive microglia following hypoglossal axotomy using hierarchical cluster analysis, The Journal of Comparative Neurology, 10.1002/cne.23228, 521, 5, 1184-1201, 2013.04.
34. J. Yamada, S. Jinno, Upregulation of calcium binding protein, S100A6, in activated astrocytes is linked to glutamate toxicity, Neuroscience, 10.1016/j.neuroscience.2012.08.068, 226, 119-129, 2012.12.
35. Jinno S, 海馬の長軸に沿った機能的分化とその神経回路基盤, 福岡医学雑誌, 103, 6, 111-119, 2012.06, 海馬は大脳辺縁系に属し、哺乳類の中枢神経系の中では最もよく研究されてきた領域の一つである。今日迄に、記憶や情動などの高次脳機能に海馬が深く関わっていることが明らかにされている。近年研究が急速に進展している海馬の長軸方向に沿った構造的・機能的な分化について概説し、更に、海馬の構造的分化と可塑性に関する研究成果をトピックとして紹介し、海馬研究の進展を示した。.
36. Jun Yamada, Shozo Jinno, Alterations in neuronal survival and glial reactions after axotomy by ceftriaxone and minocycline in the mouse hypoglossal nucleus, Neuroscience Letters, 10.1016/j.neulet.2011.09.051, 504, 3, 295-300, 2011.10.
37. J. Yamada, H. Nakanishi, S. Jinno, Differential involvement of perineuronal astrocytes and microglia in synaptic stripping after hypoglossal axotomy, Neuroscience, 10.1016/j.neuroscience.2011.03.030, 182, 1-10, 2011.05.
38. Shozo Jinno, Topographic differences in adult neurogenesis in the mouse hippocampus: A stereology-based study using endogenous markers, Hippocampus, 10.1002/hipo.20762, 21, 5, 467-480, 2011.05.
39. S. Jinno, Regional and laminar differences in antigen profiles and spatial distributions of astrocytes in the mouse hippocampus, with reference to aging, Neuroscience, 10.1016/j.neuroscience.2011.02.013, 180, 41-52, 2011.04.
40. Shozo Jinno, Decline in adult neurogenesis during aging follows a topographic pattern in the mouse hippocampus, The Journal of Comparative Neurology, 10.1002/cne.22527, 519, 3, 451-466, 2011.02.
41. Shozo Jinno, Jun Yamada, Using comparative anatomy in the axotomy model to identify distinct roles for microglia and astrocytes in synaptic stripping, Neuron Glia Biology, 10.1017/s1740925x11000135, 7, 1, 55-66, 2011.02, The synaptic terminals' withdrawal from the somata and proximal dendrites of injured motoneuron by the processes of glial cells following facial nerve axotomy has been the subject of research for many years. This phenomenon is referred to as synaptic stripping, which is assumed to help survival and regeneration of neurons via reduction of synaptic inputs. Because there is no disruption of the blood–brain barrier or infiltration of macrophages, the axotomy paradigm has the advantage of being able to selectively investigate the roles of resident glial cells in the brain. Although there have been numerous studies of synaptic stripping, the detailed mechanisms are still under debate. Here we suggest that the species and strain differences that are often present in previous work might be related to the current controversies of axotomy studies. For instance, the survival ratios of axotomized neurons were generally found to be higher in rats than in mice. However, some studies have used the axotomy paradigm to follow the glial reactions and did not assess variations in neuronal viability. In the first part of this article, we summarize and discuss the current knowledge on species and strain differences in neuronal survival, glial augmentation and synaptic stripping. In the second part, we focus on our recent findings, which show the differential involvement of microglia and astrocytes in synaptic stripping and neuronal survival. This article suggests that the comparative study of the axotomy paradigm across various species and strains may provide many important and unexpected discoveries on the multifaceted roles of microglia and astrocytes in injury and repair..
42. Shozo Jinno, Toshio Kosaka, Stereological estimation of numerical densities of glutamatergic principal neurons in the mouse hippocampus, Hippocampus, 10.1002/hipo.20685, 20, 7, 829-840, 2010.07.
43. Shozo Jinno, Toshio Kosaka, Neuronal circuit-dependent alterations in expression of two isoforms of glutamic acid decarboxylase in the hippocampus following electroconvulsive shock: A stereology-based study, Hippocampus, 10.1002/hipo.20576, 19, 11, 1130-1141, 2009.11.
44. Shozo Jinno, Structural organization of long-range GABAergic projection system of the hippocampus, Frontiers in Neuroanatomy, 10.3389/neuro.05.013.2009, 3, 13, 2009.07.
45. H. Wake, A. J. Moorhouse, S. Jinno, S. Kohsaka, J. Nabekura, Resting Microglia Directly Monitor the Functional State of Synapses In Vivo and Determine the Fate of Ischemic Terminals, The Journal of Neuroscience, 10.1523/jneurosci.4363-08.2009, 29, 13, 3974-3980, 2009.04.
46. Shozo Jinno, Kumiko Araki, Yuji Matsumoto, Yoo-Hun Suh, Tsuneyuki Yamamoto, Selective apoptosis induction in the hippocampal mossy fiber pathway by exposure to CT105, the C-terminal fragment of Alzheimer's amyloid precursor protein, Brain Research, 10.1016/j.brainres.2008.10.052, 1249, 68-78, 2009.01.
47. Jun Yamada, Yoshinori Hayashi, Shozo Jinno, Zhou Wu, Kazuhide Inoue, Shinichi Kohsaka, Hiroshi Nakanishi, Reduced synaptic activity precedes synaptic stripping in vagal motoneurons after axotomy, Glia, 10.1002/glia.20711, 56, 13, 1448-1462, 2008.10.
48. Shozo Jinno, Toshio Kosaka, Reduction of Iba1-expressing microglial process density in the hippocampus following electroconvulsive shock, Experimental Neurology, 10.1016/j.expneurol.2008.04.028, 212, 2, 440-447, 2008.08.
49. Jinno S, Stereologyによる新たな神経科学研究基盤の確立, 福岡医学雑誌, 99, 8, 169-174, 2008.08.
50. Pablo Fuentealba, Rahima Begum, Marco Capogna, Shozo Jinno, László F. Márton, Jozsef Csicsvari, Alex Thomson, Peter Somogyi, Thomas Klausberger, Ivy Cells: A Population of Nitric-Oxide-Producing, Slow-Spiking GABAergic Neurons and Their Involvement in Hippocampal Network Activity, Neuron, 10.1016/j.neuron.2008.01.034, 57, 6, 917-929, 2008.03.
51. Shozo Jinno, Frank Fleischer, Stefanie Eckel, Volker Schmidt, Toshio Kosaka, Spatial arrangement of microglia in the mouse hippocampus: A stereological study in comparison with astrocytes, Glia, 10.1002/glia.20552, 55, 13, 1334-1347, 2007.10.
52. S. Jinno, T. Klausberger, L. F. Marton, Y. Dalezios, J. D. B. Roberts, P. Fuentealba, E. A. Bushong, D. Henze, G. Buzsaki, P. Somogyi, Neuronal Diversity in GABAergic Long-Range Projections from the Hippocampus, The Journal of Neuroscience, 10.1523/jneurosci.1847-07.2007, 27, 33, 8790-8804, 2007.08.
53. Shozo Jinno, Toshio Kosaka, Cellular architecture of the mouse hippocampus: A quantitative aspect of chemically defined GABAergic neurons with stereology, Neuroscience Research, 10.1016/j.neures.2006.07.007, 56, 3, 229-245, 2006.11.
54. S. Jinno, A. Jeromin, T. Kosaka, Postsynaptic and extrasynaptic localization of Kv4.2 channels in the mouse hippocampal region, with special reference to targeted clustering at gabaergic synapses, Neuroscience, 10.1016/j.neuroscience.2005.04.065, 134, 2, 483-494, 2005.01.
55. Shozo Jinno, Toshio Kosaka, Parvalbumin is expressed in glutamatergic and GABAergic corticostriatal pathway in mice, The Journal of Comparative Neurology, 10.1002/cne.20246, 477, 2, 188-201, 2004.09.
56. Shozo Jinno, Andreas Jeromin, Toshio Kosaka, Expression and possible role of neuronal calcium sensor-1 in the cerebellum, The Cerebellum, 10.1080/14734220310025187, 3, 2, 83-88, 2004.06.
57. Junichi Nabekura, Shutaro Katsurabayashi, Yasuhiro Kakazu, Shumei Shibata, Atsushi Matsubara, Shozo Jinno, Yoshito Mizoguchi, Akira Sasaki, Hitoshi Ishibashi, Developmental switch from GABA to glycine release in single central synaptic terminals, Nature Neuroscience, 10.1038/nn1170, 7, 1, 17-23, 2004.01.
58. S. Jinno, T. Kosaka, Patterns of colocalization of neuronal nitric oxide synthase and somatostatin-like immunoreactivity in the mouse hippocampus: quantitative analysis with optical disector, Neuroscience, 10.1016/j.neuroscience.2004.01.027, 124, 4, 797-808, 2004.01.
59. Shozo Jinno, Andreas Jeromin, John Roder, Toshio Kosaka, Compartmentation of the mouse cerebellar cortex by neuronal calcium sensor-1, The Journal of Comparative Neurology, 10.1002/cne.10585, 458, 4, 412-424, 2003.04.
60. Shozo Jinno, Satoru Ishizuka, Toshio Kosaka, Ionic currents underlying rhythmic bursting of ventral mossy cells in the developing mouse dentate gyrus, European Journal of Neuroscience, 10.1046/j.1460-9568.2003.02569.x, 17, 7, 1338-1354, 2003.04.
61. S Jinno, T Kosaka, Heterogeneous expression of the cholecystokinin-like immunoreactivity in the mouse hippocampus, with special reference to the dorsoventral difference, Neuroscience, 10.1016/j.neuroscience.2003.08.039, 122, 4, 869-884, 2003.01.
62. Shozo Jinno, Toshio Kosaka, Immunocytochemical characterization of hippocamposeptal projecting GABAergic nonprincipal neurons in the mouse brain: a retrograde labeling study, Brain Research, 10.1016/s0006-8993(02)02804-4, 945, 2, 219-231, 2002.08.
63. S Jinno, A Jeromin, J Roder, T Kosaka, Immunocytochemical localization of neuronal calcium sensor-1 in the hippocampus and cerebellum of the mouse, with special reference to presynaptic terminals, Neuroscience, 10.1016/s0306-4522(02)00172-0, 113, 2, 449-461, 2002.08.
64. Shozo Jinno, Toshio Kosaka, Patterns of expression of calcium binding proteins and neuronal nitric oxide synthase in different populations of hippocampal GABAergic neurons in mice, The Journal of Comparative Neurology, 10.1002/cne.10251, 449, 1, 1-25, 2002.07.
65. A Doi, H Ishibashi, S Jinno, T Kosaka, N Akaike, Presynaptic inhibition of GABAergic miniature currents by metabotropic glutamate receptor in the rat CNS, Neuroscience, 10.1016/s0306-4522(01)00484-5, 109, 2, 299-311, 2002.01.
66. Shozo Jinno, Naoko Kinukawa, Toshio Kosaka, Morphometric multivariate analysis of GABAergic neurons containing calretinin and neuronal nitric oxide synthase in the mouse hippocampus, Brain Research, 10.1016/s0006-8993(01)02292-2, 900, 2, 195-204, 2001.05.
67. S Jinno, T Kosaka, Colocalization of parvalbumin and somatostatin-like immunoreactivity in the mouse hippocampus: Quantitative analysis with optical disector, The Journal of Comparative Neurology, 428, 3, 377-388, 2000.12, The colocalization of parvalbumin (PV) and somatostatin (SS)-like immunoreactivity was studied quantitatively in the mouse hippocampus, with particular reference to their areal and dorsoventral differences. The optical disector method was applied by using a confocal laser scanning microscope with immunofluorescent double-labeling. In the present study, we found a particular subpopulation of hippocampal nonprincipal neurons that contained both PV and SS-like immunoreactivity, i.e., PV-immunoreactive (IR)/SS-like immunoreactive (LIR) neurons. In the CA1 region, PV-IR/SS-LIR, neurons were restricted to the stratum oriens (SO). In the CA3 region, they were scattered in the SO, stratum pyramidale (SP), and stratum radiatum (SR). However, they were rarely seen in the dentate gyrus (DG). The proportion of PV-IR/SS-LIR neurons in the PV-IR neurons or SS-LIR neurons was about 10% in the CA1 region, 15-30% in the CA3 region, 0-5% in the DG, and 10-20% in total. Laminar analysis revealed that the proportions of PV-IR/SS-LIR neurons in the PV-IR neurons were high in the SO (about 25%) of the CA1 region, and in the SO (about 50%) and SR (30-45%) of the CA3 region. The proportion of PV-IR/SS-LIR neurons in the SS-LIR neurons was low in the SO of the CA1 region (about 10%), but high in the SO (35-65%) and SR (35-45%) of the CA3 region. Morphologically, medium-sized horizontal fusiform and multipolar PV-IR/SS-LIR neurons were frequently observed, and they showed weak immunoreactivity for PV. Large-sized vertical bitufted and triangular PV-ER neurons lacked SS-like immunoreactivity, and most of them showed moderate to intense immunoreactivity for PV. Tn addition, we provide direct Evidence that some PV-IR/SS-LIR neurons projected to the medial septum by using retrograde labeling with Fluoro-Gold injection. These observations indicate that PV-IR/SS-LIR neurons constitute a particular subpopulation of hippocampal nonprincipal neurons. (C) 2000 Wiley-Liss, Inc..
68. S Jinno, Y Aika, T Fukuda, T Kosaka, Quantitative analysis of neuronal nitric oxide synthase-immunoreactive neurons in the mouse hippocampus with optical disector, The Journal of Comparative Neurology, 410, 3, 398-412, 1999.08, A detailed quantitative analysis of immunocytochemically identified nonprincipal neurons containing neuronal nitric oxide synthase (nNOS) was performed on the mouse hippocampus, with particular reference to the dorsoventral gradient. The present study applied two variations of a stereologic technique, the optical disector-one that; used confocal laser-scanning microscope optical sections to examine colocalization of nNOS and glutamic acid decarboxylase 67 (GAD67), and the other that used conventional thick sections to examine numerical densities (NDs) and cell sizes of nNOS-immunoreactive (IR) neurons. Colocalization analysis indicated that practically all nNOS-IR neurons (97.6%) were GAD67-IR, whereas a part of the GAD67-IR neurons (about 30%) were nNOS-IR in the whole hippocampus at both dorsal and ventral levels. The percentages of CAD67-IR neurons containing nNOS were higher in the dentate gyrus (DG, about; 50%), and lower in the Ammon's horn (about 20%). Laminar analysis revealed that the majority of GAD67-IR neurons contained nNOS in the stratum lacunosum-moleculare of the CA3 region (about 60%) and in the molecular layer of the DG (about 80%). The NDs of nNOS-IR neurons in the whole hippocampus showed a dorsoventral gradient, which increased from dorsal(1.6 x 10(3)/mm(3)) to ventral (2.2 x 10(3)/mm(3)) levels. The NDs were relatively higher in the principal cell layers, where about 40% of nNOS-IR neurons were situated both in the Ammon's horn and DG. The mean cell sizes of nNOS-IR neurons showed no remarkable laminar differences or dorsoventral gradient in the Ammon's horn, but; they were extensively larger in the hilus of the DG than in other layers. These results indicate that nNOS-IR neurons in the mouse hippocampus represent a subpopulation of gamma-aminobutyric acid (GABA)ergic neurons and suggest that the laminar distributions of nNOS-IR neurons related to possible functional heterogeneity of GABAergic neurons in each hippocampal layer. (C) 1999 Wiley-Liss, Inc..
69. Shozo Jinno, Yusuke Aika, Takaichi Fukuda, Toshio Kosaka, Quantitative analysis of GABAergic neurons in the mouse hippocampus, with optical disector using confocal laser scanning microscope, Brain Research, 10.1016/s0006-8993(98)01075-0, 814, 1-2, 55-70, 1998.12.

九大関連コンテンツ

pure2017年10月2日から、「九州大学研究者情報」を補完するデータベースとして、Elsevier社の「Pure」による研究業績の公開を開始しました。