Network


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

Hotspot


Dive into the research topics where Yoshifumi Sonobe is active.

Publication


Featured researches published by Yoshifumi Sonobe.


Journal of Biological Chemistry | 2006

Tumor Necrosis Factor-α Induces Neurotoxicity via Glutamate Release from Hemichannels of Activated Microglia in an Autocrine Manner

Hideyuki Takeuchi; Shijie Jin; Jinyan Wang; Guiqin Zhang; Jun Kawanokuchi; Reiko Kuno; Yoshifumi Sonobe; Tetsuya Mizuno; Akio Suzumura

Glutamate released by activated microglia induces excitoneurotoxicity and may contribute to neuronal damage in neurodegenerative diseases, including Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, and multiple sclerosis. In addition, tumor necrosis factor-α (TNF-α) secreted from activated microglia may elicit neurodegeneration through caspase-dependent cascades and silencing cell survival signals. However, direct neurotoxicity of TNF-α is relatively weak, because TNF-α also increases production of neuroprotective factors. Accordingly, it is still controversial how TNF-α exerts neurotoxicity in neurodegenerative diseases. Here we have shown that TNF-α is the key cytokine that stimulates extensive microglial glutamate release in an autocrine manner by up-regulating glutaminase to cause excitoneurotoxicity. Further, we have demonstrated that the connexin 32 hemichannel of the gap junction is another main source of glutamate release from microglia besides glutamate transporters. Although pharmacological blockade of glutamate receptors is a promising therapeutic candidate for neurodegenerative diseases, the associated perturbation of physiological glutamate signals has severe adverse side effects. The unique mechanism of microglial glutamate release that we describe here is another potential therapeutic target. We rescued neuronal cell death in vitro by using a glutaminase inhibitor or hemichannel blockers to diminish microglial glutamate release without perturbing the physiological glutamate level. These drugs may give us a new therapeutic strategy against neurodegenerative diseases with minimum adverse side effects.


Brain Research | 2011

Production and functions of IL-33 in the central nervous system

Satoko Yasuoka; Jun Kawanokuchi; Bijay Parajuli; Shijie Jin; Yukiko Doi; Mariko Noda; Yoshifumi Sonobe; Hideyuki Takeuchi; Tetsuya Mizuno; Akio Suzumura

Interleukin-33 (IL-33) is a novel multifunctional IL-1 family cytokine. IL-33 signals via a heterodimer composed of IL-1 receptor-related protein ST2 and IL-1 receptor accessory protein (IL-1RAcP). IL-33 has been shown to activate T helper 2 cells (Th2), mast cells and basophils to produce a variety of Th2 cytokines and mediate allergic-type immune responses. Recent studies have revealed that glial cells are induced to express IL-33 mRNA and protein. However, the functions of IL-33 and its producing cells in the central nervous system (CNS) are still uncertain. In this study, we investigated the expression and function of IL-33 in the CNS. IL-33 is produced by endothelial cells and astrocytes but not by microglia or neurons. The IL-33 receptors are expressed mainly in microglia and astrocytes. IL-33 dose-dependently induces the proliferation of microglia and enhances the production of pro-inflammatory cytokines, such as IL-1β and TNFα, as well as the anti-inflammatory cytokine IL-10. It also enhances chemokines and nitric oxide production and phagocytosis by microglia. Thus, IL-33 produced in the CNS activates microglia and may function as a pro-inflammatory mediator in the pathophysiology of the CNS.


The FASEB Journal | 2008

Interferon-γ directly induces neurotoxicity through a neuron specific, calcium-permeable complex of IFN-γ receptor and AMPA GluR1 receptor

Tetsuya Mizuno; Guiqin Zhang; Hideyuki Takeuchi; Jun Kawanokuchi; Jinyan Wang; Yoshifumi Sonobe; Shijie Jin; Naoki Takada; Yukio Komatsu; Akio Suzumura

Interferon‐γ (IFN‐γ) is a proinflamma tory cytokine that plays a pivotal role in pathology of diseases in the central nervous system (CNS), such as multiple sclerosis. However, the direct effect of IFN‐γ on neuronal cells has yet to be elucidated. We show here that IFN‐γ directly induces neuronal dysfunction, which appears as dendritic bead formation in mouse cortical neurons and enhances glutamate neurotoxicity mediated via alpha‐amino‐3‐hydroxy‐5‐methyl‐4‐isox‐ azolepropionic (AMPA) receptors but not N‐methyl‐D‐ aspartate receptors. In the CNS, IFN‐γ receptor forms a unique, neuron‐specific, calcium‐permeable receptor complex with AMPA receptor subunit GluRl. Through this receptor complex, IFN‐γ phosphorylates GluRl at serine 845 position by JAKT2/STAT1 pathway, in creases Ca2+ influx and following nitric oxide production, and subsequently decreases ATP production, leading to the dendritic bead formation. These findings provide novel mechanisms of neuronal excitotoxicity, which may occur in both inflammatory and neurodegen erative diseases in the CNS.— Mizuno, T., Zhang, G., Takeuchi, H., Kawanokuchi, J., Wang, J., Sonobe, Y., Jin, S., Takada, N., Komatsu, Y., Suzumura, A. Inter feron‐γ directly induces neurotoxicity through a neu ron specific, calcium‐ permeable complex of IFN‐γ receptor and AMPA GluRl receptor. FASEB J. 22, 1797–1806 (2008)


PLOS ONE | 2011

Blockade of Gap Junction Hemichannel Suppresses Disease Progression in Mouse Models of Amyotrophic Lateral Sclerosis and Alzheimer's Disease

Hideyuki Takeuchi; Hiroyuki Mizoguchi; Yukiko Doi; Shijie Jin; Mariko Noda; Jianfeng Liang; Hua Li; Yan Zhou; Rarami Mori; Satoko Yasuoka; Endong Li; Bijay Parajuli; Jun Kawanokuchi; Yoshifumi Sonobe; Jun Sato; Koji Yamanaka; Gen Sobue; Tetsuya Mizuno; Akio Suzumura

BACKGROUND Glutamate released by activated microglia induces excitotoxic neuronal death, which likely contributes to non-cell autonomous neuronal death in neurodegenerative diseases, including amyotrophic lateral sclerosis and Alzheimers disease. Although both blockade of glutamate receptors and inhibition of microglial activation are the therapeutic candidates for these neurodegenerative diseases, glutamate receptor blockers also perturbed physiological and essential glutamate signals, and inhibitors of microglial activation suppressed both neurotoxic/neuroprotective roles of microglia and hardly affected disease progression. We previously demonstrated that activated microglia release a large amount of glutamate specifically through gap junction hemichannel. Hence, blockade of gap junction hemichannel may be potentially beneficial in treatment of neurodegenerative diseases. METHODS AND FINDINGS In this study, we generated a novel blood-brain barrier permeable gap junction hemichannel blocker based on glycyrrhetinic acid. We found that pharmacologic blockade of gap junction hemichannel inhibited excessive glutamate release from activated microglia in vitro and in vivo without producing notable toxicity. Blocking gap junction hemichannel significantly suppressed neuronal loss of the spinal cord and extended survival in transgenic mice carrying human superoxide dismutase 1 with G93A or G37R mutation as an amyotrophic lateral sclerosis mouse model. Moreover, blockade of gap junction hemichannel also significantly improved memory impairments without altering amyloid β deposition in double transgenic mice expressing human amyloid precursor protein with K595N and M596L mutations and presenilin 1 with A264E mutation as an Alzheimers disease mouse model. CONCLUSIONS Our results suggest that gap junction hemichannel blockers may represent a new therapeutic strategy to target neurotoxic microglia specifically and prevent microglia-mediated neuronal death in various neurodegenerative diseases.


Journal of Biological Chemistry | 2011

Fractalkine Attenuates Excito-neurotoxicity via Microglial Clearance of Damaged Neurons and Antioxidant Enzyme Heme Oxygenase-1 Expression

Mariko Noda; Yukiko Doi; Jianfeng Liang; Jun Kawanokuchi; Yoshifumi Sonobe; Hideyuki Takeuchi; Tetsuya Mizuno; Akio Suzumura

Glutamate-induced excito-neurotoxicity likely contributes to non-cell autonomous neuronal death in neurodegenerative diseases. Microglial clearance of dying neurons and associated debris is essential to maintain healthy neural networks in the central nervous system. In fact, the functions of microglia are regulated by various signaling molecules that are produced as neurons degenerate. Here, we show that the soluble CX3C chemokine fractalkine (sFKN), which is secreted from neurons that have been damaged by glutamate, promotes microglial phagocytosis of neuronal debris through release of milk fat globule-EGF factor 8, a mediator of apoptotic cell clearance. In addition, sFKN induces the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) in microglia in the absence of neurotoxic molecule production, including NO, TNF, and glutamate. sFKN treatment of primary neuron-microglia co-cultures significantly attenuated glutamate-induced neuronal cell death. Using several specific MAPK inhibitors, we found that sFKN-induced heme oxygenase-1 expression was primarily mediated by activation of JNK and nuclear factor erythroid 2-related factor 2. These results suggest that sFKN secreted from glutamate-damaged neurons provides both phagocytotic and neuroprotective signals.


Brain Research | 2006

The role of TNF-alpha and its receptors in the production of NGF and GDNF by astrocytes.

Reiko Kuno; Yusuke Yoshida; Atsumi Nitta; Toshitaka Nabeshima; Jinyan Wang; Yoshifumi Sonobe; Jun Kawanokuchi; Hideyuki Takeuchi; Tetsuya Mizuno; Akio Suzumura

The neurotrophic factors, nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF), are produced by astrocytes, and are induced by inflammatory stimuli including bacterial lipopolysaccharide and pro-inflammatory cytokines. In this study, we examined the regulatory mechanisms of tumor necrosis factor-alpha (TNF-alpha)-induced production of neurotrophic factors. We show here that cultured astrocytes express both TNF-alpha receptor 1 (TNFR1) and TNFR2, and that activation of these receptors by TNF-alpha promotes expression of both NGF and GDNF. In addition, we observe that not only exogenous TNF-alpha but also TNF-alpha produced by astrocytes induce NGF and GDNF production in astrocytes. These results suggest that an autocrine loop involving TNF-alpha contributes to the production of neurotrophic factors in response to inflammation.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Inhibition of midkine alleviates experimental autoimmune encephalomyelitis through the expansion of regulatory T cell population

Jinyan Wang; Hideyuki Takeuchi; Yoshifumi Sonobe; Shijie Jin; Tetsuya Mizuno; Shin Miyakawa; Masatoshi Fujiwara; Yoshikazu Nakamura; Takuma Kato; Hisako Muramatsu; Takashi Muramatsu; Akio Suzumura

CD4+CD25+ regulatory T (Treg) cells are crucial mediators of autoimmune tolerance. The factors that regulate Treg cells, however, are largely unknown. Here, we show that deficiency in midkine (MK), a heparin-binding growth factor involved in oncogenesis, inflammation, and tissue repair, attenuated experimental autoimmune encephalomyelitis (EAE) because of an expansion of the Treg cell population in peripheral lymph nodes and decreased numbers of autoreactive T-helper type 1 (TH1) and TH17 cells. MK decreased the Treg cell population ex vivo in a dose-dependent manner by suppression of STAT5 phosphorylation that is essential for Foxp3 expression. Moreover, administration of anti-MK RNA aptamers significantly expanded the Treg cell population and alleviated EAE symptoms. These observations indicate that MK serves as a critical suppressor of Treg cell expansion, and inhibition of MK using RNA aptamers may provide an effective therapeutic strategy against autoimmune diseases, including multiple sclerosis.


Journal of Immunology | 2011

IL-9 Promotes Th17 Cell Migration into the Central Nervous System via CC Chemokine Ligand-20 Produced by Astrocytes

Yan Zhou; Yoshifumi Sonobe; Tomohiko Akahori; Shijie Jin; Jun Kawanokuchi; Mariko Noda; Yoichiro Iwakura; Tetsuya Mizuno; Akio Suzumura

Newly discovered IL-9–producing helper T cells (Th9) reportedly exert both aggravating and suppressive roles on experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. However, it is still unclear whether Th9 is a distinct Th cell subset and how IL-9 functions in the CNS. In this study, we show that IL-9 is produced by naive CD4+ T cells that were stimulated with anti-CD3 and anti-CD28 Abs under the conditions of Th2-, inducible regulatory T cell-, Th17-, and Th9-polarizing conditions and that IL-9 production is significantly suppressed in the absence of IL-4, suggesting that IL-4 is critical for the induction of IL-9 by each producing cell. The IL-9 receptor complex, IL-9R and IL-2Rγ, is constitutively expressed on astrocytes. IL-9 induces astrocytes to produce CCL-20 but not other chemokines, including CCL-2, CCL-3, and CXCL-2 by astrocytes. The conditioned medium of IL-9–stimulated astrocytes induces Th17 cell migration in vitro, which is cancelled by adding anti–CCL-20 neutralizing Abs. Treating with anti–IL-9 neutralizing Abs attenuates experimental autoimmune encephalomyelitis, decreases the number of infiltrating Th17 cells, and reduces CCL-20 expression in astrocytes. These results suggest that IL-9 is produced by several Th cell subsets in the presence of IL-4 and induces CCL-20 production by astrocytes to induce the migration of Th17 cells into the CNS.


Brain Research | 2008

Excitatory amino acid transporter expression by astrocytes is neuroprotective against microglial excitotoxicity

Jianfeng Liang; Hideyuki Takeuchi; Yukiko Doi; Jun Kawanokuchi; Yoshifumi Sonobe; Shijie Jin; Izumi Yawata; Hua Li; Satoko Yasuoka; Tetsuya Mizuno; Akio Suzumura

Glutamate-induced excitotoxicity is considered as a major cause of neurodegenerative disease. Excitatory amino acid transporters (EAATs) on glial cells are responsible for the homeostasis of extracellular glutamate in the central nervous system which may contribute to the prevention of excitotoxic neurodegeneration. However, the differential EAAT expression in astrocytes and microglia is not fully understood. In this study, we compared the expression of EAATs in astrocytes and microglia, and we assessed the neuroprotective and neurotoxic function of astrocytes and microglia by a co-culture system. RT-PCR analyses detected that astrocytes expressed each EAAT (EAAT1-5) whereas microglia did not express EAAT4. Western blot analyses demonstrated that astrocytes express a much larger amount of membrane-localized EAATs than microglia. Astrocytes prevented excito-neurotoxicity by the reduction of exogenous glutamate whereas microglia did not. Conversely, activated microglia released an excess of glutamate that induced excitotoxic neuronal death. Astrocytes rescued neurons from microglial glutamate-induced death in a ratio-dependent manner. Inhibition of EAATs abolished glutamate uptake and the neuroprotective effect of astrocytes, but it did not alter any microglial neurotoxic or neuroprotective effects. These results revealed that astrocytic EAATs can counteract microglial glutamate-induced neuronal death whereas microglial EAATs are inconsequential to neurotoxicity and neuroprotection.


Brain Research | 2005

Production of IL-27 and other IL-12 family cytokines by microglia and their subpopulations.

Yoshifumi Sonobe; Izumi Yawata; Jun Kawanokuchi; Hideyuki Takeuchi; Tetsuya Mizuno; Akio Suzumura

Production of IL-27 and other IL-12 family cytokines by murine microglia were examined using RT-PCR, real-time RT-PCR and Western blot analysis. We show for the first time that murine microglia produce IL-27 in response to lipopolysaccharide (LPS) and/or interferon-gamma. Primary microglia, but not their cell lines, also induce IL-12 and IL-23 upon above stimulation. Therefore, microglia may play a critical role initiating Th1 responses via producing IL-12 family cytokines in the brain.

Collaboration


Dive into the Yoshifumi Sonobe'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