Network


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

Hotspot


Dive into the research topics where Hidemitsu Nakajima is active.

Publication


Featured researches published by Hidemitsu Nakajima.


Journal of Biological Chemistry | 2007

The Active Site Cysteine of the Proapoptotic Protein Glyceraldehyde-3-phosphate Dehydrogenase Is Essential in Oxidative Stress-induced Aggregation and Cell Death

Hidemitsu Nakajima; Wataru Amano; Akikazu Fujita; Ayano Fukuhara; Yasu-Taka Azuma; Fumiaki Hata; Takashi Inui; Tadayoshi Takeuchi

Recent studies have revealed that the redox-sensitive glycolytic enzyme, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), is involved in neuronal cell death that is triggered by oxidative stress. GAPDH is locally deposited in disulfide-bonded aggregates at lesion sites in certain neurodegenerative diseases. In this study, we investigated the molecular mechanism that underlies oxidative stress-induced aggregation of GAPDH and the relationship between structural abnormalities in GAPDH and cell death. Under nonreducing in vitro conditions, oxidants induced oligomerization and insoluble aggregation of GAPDH via the formation of intermolecular disulfide bonds. Because GAPDH has four cysteine residues, including the active site Cys149, we prepared the cysteine-substituted mutants C149S, C153S, C244A, C281S, and C149S/C281S to identify which is responsible for disulfide-bonded aggregation. Whereas the aggregation levels of C281S were reduced compared with the wild-type enzyme, neither C149S nor C149S/C281S aggregated, suggesting that the active site cysteine plays an essential role. Oxidants also caused conformational changes in GAPDH concomitant with an increase in β-sheet content; these abnormal conformations specifically led to amyloid-like fibril formation via disulfide bonds, including Cys149. Additionally, continuous exposure of GAPDH-overexpressing HeLa cells to oxidants produced disulfide bonds in GAPDH leading to both detergent-insoluble and thioflavin-S-positive aggregates, which were associated with oxidative stress-induced cell death. Thus, oxidative stresses induce amyloid-like aggregation of GAPDH via aberrant disulfide bonds of the active site cysteine, and the formation of such abnormal aggregates promotes cell death.


Journal of Biological Chemistry | 2009

Glyceraldehyde-3-phosphate Dehydrogenase Aggregate Formation Participates in Oxidative Stress-induced Cell Death

Hidemitsu Nakajima; Wataru Amano; Takeya Kubo; Ayano Fukuhara; Hideshi Ihara; Yasu Taka Azuma; Hisao Tajima; Takashi Inui; Akira Sawa; Tadayoshi Takeuchi

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH)2 is a classic glycolytic enzyme that also mediates cell death by its nuclear translocation under oxidative stress. Meanwhile, we previously presented that oxidative stress induced disulfide-bonded GAPDH aggregation in vitro. Here, we propose that GAPDH aggregate formation might participate in oxidative stress-induced cell death both in vitro and in vivo. We show that human GAPDH amyloid-like aggregate formation depends on the active site cysteine-152 (Cys-152) in vitro. In SH-SY5Y neuroblastoma, treatment with dopamine decreases the cell viability concentration-dependently (IC50 = 202 μm). Low concentrations of dopamine (50–100 μm) mainly cause nuclear translocation of GAPDH, whereas the levels of GAPDH aggregates correlate with high concentrations of dopamine (200–300 μm)-induced cell death. Doxycycline-inducible overexpression of wild-type GAPDH in SH-SY5Y, but not the Cys-152-substituted mutant (C152A-GAPDH), accelerates cell death accompanying both endogenous and exogenous GAPDH aggregate formation in response to high concentrations of dopamine. Deprenyl, a blocker of GAPDH nuclear translocation, fails to inhibit the aggregation both in vitro and in cells but reduced cell death in SH-SY5Y treated with only a low concentration of dopamine (100 μm). These results suggest that GAPDH participates in oxidative stress-induced cell death via an alternative mechanism in which aggregation but not nuclear translocation of GAPDH plays a role. Moreover, we observe endogenous GAPDH aggregate formation in nigra-striatum dopaminergic neurons after methamphetamine treatment in mice. In transgenic mice overexpressing wild-type GAPDH, increased dopaminergic neuron loss and GAPDH aggregate formation are observed. These data suggest a critical role of GAPDH aggregates in oxidative stress-induced brain damage.


Journal of Biological Chemistry | 2004

Critical Role of the Automodification of Poly(ADP-ribose) Polymerase-1 in Nuclear Factor-κB-dependent Gene Expression in Primary Cultured Mouse Glial Cells

Hidemitsu Nakajima; Hiroshi Nagaso; Nobukazu Kakui; Midori Ishikawa; Toyokazu Hiranuma; Shigeru Hoshiko

Synthesis of ADP-ribose polymers catalyzed by poly-(ADP-ribose) polymerase-1 (PARP-1) has been implicated in transcriptional regulation. Recent studies with PARP-1 null mice and PARP-1 inhibitors have also demonstrated that PARP-1 has an essential role in nuclear factor-κB (NF-κB)-dependent gene expression induced by various inflammatory stimuli. In this study, we used primary cultured mouse glial cells to investigate the role of poly(ADP-ribosyl)ation by PARP-1 in NF-κB-dependent gene expression. PARP-1 inhibitors and the antisense RNA for PARP-1 mRNA suppressed lipopolysaccharide (LPS)-induced expression of tumor necrosis factor-α and inducible nitric-oxide synthase, suggesting that PARP-1 activity has a critical role in synthesis. Western blotting with anti-poly(ADP-ribose) antibody revealed that PARP-1 itself was mainly poly(ADP-ribosyl)ated in glial cells, i.e. automodified PARP-1 (AM-PARP). The amounts of AM-PARP were not affected by LPS treatment, but were decreased by PARP-1 inhibitors. Electrophoretic mobility shift assay revealed that PARP-1 inhibitors and the antisense RNA for PARP-1 mRNA reduced the LPS-induced DNA binding of NF-κB. Non-modified PARP-1 also reduced the DNA binding of NF-κB via its physical association with NF-κB, whereas AM-PARP had no effect. On the other hand, enhancement of the automodification of PARP-1 by the addition of NAD+, its substrate, promoted the DNA binding of NF-κB. Furthermore, in in vitro transcription assay, the addition of AM-PARP or NAD+ to nuclear extracts promoted NF-κB p50-dependent transcription. These results indicate that automodification of PARP-1 positively up-regulates formation of the NF-κB·DNA complex and enhances transcriptional activation. Therefore, AM-PARP may be critical for the NF-κB-dependent gene expression of some inflammatory mediators in glial cells.


Inflammatory Bowel Diseases | 2010

Interleukin-19 protects mice from innate-mediated colonic inflammation

Yasu-Taka Azuma; Yukiko Matsuo; Mitsuru Kuwamura; George D. Yancopoulos; David M. Valenzuela; Andrew J. Murphy; Hidemitsu Nakajima; Margaret Karow; Tadayoshi Takeuchi

Background: Inflammatory bowel disease (IBD) results from the chronic dysregulation of the mucosal immune system and the aberrant activation of both the innate and the adaptive immune responses. We used two complementary models of colonic inflammation to examine the roles of interleukin (IL)‐19 in colonic inflammation and thus its possible role in IBD. Methods: Using gene‐targeting, we generated IL‐19‐deficient mice. To study the activation of the innate immune response during colonic inflammation we characterized an innate immune‐mediated model of colitis induced by dextran sulfate sodium (DSS). DSS can induce not only acute colitis but also chronic colitis. In addition to the acute DSS‐induced colitis model, we used a chronic DSS‐induced colitis model that is associated with the activation of both Th1 and Th2 cytokines as well as innate immune response in the colon. Results: We show that IL‐19‐deficient mice are more susceptible to experimental acute colitis induced by DSS, and this increased susceptibility is correlated with the accumulation of macrophages and the increased production of IFN‐&ggr;, IL‐1&bgr;, IL‐6, IL‐12, TNF‐&agr;, and KC. Additionally, cytokine production in IL‐19‐deficient macrophages was enhanced on stimulation of lipopolysaccharide (LPS) through reduced phosphorylation of STAT1 and STAT3. Moreover, our results clearly demonstrate that IL‐19 is required for B‐cell infiltration during chronic DSS‐induced colitis, which may be mediated by IL‐13 and IL‐6. Conclusions: The finding that IL‐19 drives pathogenic innate immune responses in the colon suggests that the selective targeting of IL‐19 may be an effective therapeutic approach in the treatment of human IBD. Inflamm Bowel Dis 2009


Journal of Cellular Physiology | 2008

PACAP provides colonic protection against dextran sodium sulfate induced colitis

Yasu-Taka Azuma; Kiyomi Hagi; Norihito Shintani; Mitsuru Kuwamura; Hidemitsu Nakajima; Hitoshi Hashimoto; Akemichi Baba; Tadayoshi Takeuchi

Pituitary adenylate cyclase‐activating polypeptide (PACAP) plays a crucial role in immunity and inflammation. Our aim was to obtain insight in the role of PACAP in experimental colitis in mice and thus its possible role in inflammatory bowel disease. PACAP‐deficient (PACAP−/−) mice and wild‐type control mice were challenged by colitis‐inducing agent, dextran sulfate sodium (DSS). We monitored clinical symptoms, intestinal morphology, and difference of cytokine production in the proximal and distal colon. After DSS administration, mortality was more severe in PACAP−/− mice versus wild‐type control mice. The histological score and the disease activity index of PACAP−/− mice were significantly higher than those of wild‐type control mice. In proximal colon, production of IL‐1β and IL‐6 in PACAP−/− mice were significantly upregulated on day 8 after DSS administration, compared to wild‐type control mice. In distal colon, furthermore, production of IFNγ, IL‐1β, IL‐6, IL‐12, and KC were significantly higher in PACAP−/− mice than in wild‐type control mice on day 4. Our findings indicate that PACAP regulates the production of pro‐inflammatory cytokine in the experimental colitis. J. Cell. Physiol. 216: 111–119, 2008.


Journal of Pharmacology and Experimental Therapeutics | 2004

A Newly Synthesized Poly(ADP-Ribose) Polymerase Inhibitor, DR2313 [2-Methyl-3,5,7,8-tetrahydrothiopyrano[4,3-d]-pyrimidine-4-one]: Pharmacological Profiles, Neuroprotective Effects, and Therapeutic Time Window in Cerebral Ischemia in Rats

Hidemitsu Nakajima; Nobukazu Kakui; Kunihiro Ohkuma; Midori Ishikawa; Toshifumi Hasegawa

We investigated the pharmacological profiles of DR2313 [2-methyl-3,5,7,8-tetrahydrothiopyrano[4,3-d]pyrimidine-4-one], a newly synthesized poly(ADP-ribose) polymerase (PARP) inhibitor, and its neuroprotective effects on ischemic injuries in vitro and in vivo. DR2313 competitively inhibited poly(ADP-ribosyl)ation in nuclear extracts of rat brain in vitro (Ki = 0.23 μM). Among several NAD+-utilizing enzymes, DR2313 was specific for PARP but not selective between PARP-1 and PARP-2. DR2313 also showed excellent profiles in water solubility and rat brain penetrability. In in vitro models of cerebral ischemia, exposure to hydrogen peroxide or glutamate induced cell death with overactivation of PARP, and treatment with DR2313 reduced excessive formation of poly(ADP-ribose) and cell death. In both permanent and transient focal ischemia models in rats, pretreatment with DR2313 (10 mg/kg i.v. bolus and 10 mg/kg/h i.v. infusion for 6 h) significantly reduced the cortical infarct volume. To determine the therapeutic time window of neuroprotection by DR2313, the effect of post-treatment was examined in transient focal ischemia model and compared with that of a free radical scavenger, MCI-186 (3-methyl-1-phenyl-2-pyrazolone-5-one). Pretreatment with MCI-186 (3 mg/kg i.v. bolus and 3 mg/kg/h i.v. infusion for 6 h) significantly reduced the infarct volume, whereas the post-treatment failed to show any effects. In contrast, post-treatment with DR2313 (same regimen) delaying for 2 h after ischemia still prevented the progression of infarction. These results indicate that DR2313 exerts neuroprotective effects via its potent PARP inhibition, even when the treatment is initiated after ischemia. Thus, a PARP inhibitor like DR2313 may be more useful in treating acute stroke than a free radical scavenger.


International Immunopharmacology | 2010

PPARα contributes to colonic protection in mice with DSS-induced colitis

Yasu-Taka Azuma; Kazuhiro Nishiyama; Yukiko Matsuo; Mitsuru Kuwamura; Ai Morioka; Hidemitsu Nakajima; Tadayoshi Takeuchi

Inflammatory bowel disease (IBD) is characterized by repeated chronic inflammation of the gastrointestinal tract. We have used the complementary model of colonic inflammation to examine the roles of peroxisome proliferator-activated receptor α (PPARα) in colonic inflammation and thus its possible role in IBD. We characterized an innate immune-mediated model of colitis induced by dextran sulfate sodium (DSS). Mice with DSS-induced colitis were injected with Wy-14643 (2 mg/kg) as a PPARα agonist every day from day 0 to day 5. We show that mice given Wy-14643 were less susceptible to experimental acute colitis induced by DSS, and this decreased susceptibility was correlated with decreased production of IFNγ, IL-1β, IL-6, and TNF-α. Our findings suggest that PPARα has a role in controlling colonic inflammation and mucosal tissue homeostasis.


Journal of Biotechnology | 2012

A rapid, targeted, neuron-selective, in vivo knockdown following a single intracerebroventricular injection of a novel chemically modified siRNA in the adult rat brain

Hidemitsu Nakajima; Takeya Kubo; Yuko Semi; Masanori Itakura; Mitsuru Kuwamura; Takeshi Izawa; Yasu-Taka Azuma; Tadayoshi Takeuchi

There has been a dramatic expansion of the literature on RNA interference and with it, increasing interest in the potential clinical utility of targeted inhibition of gene expression and associated protein knockdown. However, a critical factor limiting the experimental and therapeutic application of RNA interference is the ability to deliver small interfering RNAs (siRNAs), particularly in the central nervous system, without complications such as toxicity and inflammation. Here we show that a single intracerebroventricular injection of Accell siRNA, a new type of naked siRNA that has been modified chemically to allow for delivery in the absence of transfection reagents, even into differentiated cells such mature neurons, leads to neuron-specific protein knockdown in the adult rat brain. Following in vivo delivery, targeted Accell siRNAs were incorporated successfully into various types of mature neurons, but not glia, for 1 week in diverse brain regions (cortex, striatum, hippocampus, midbrain, and cerebellum) with an efficacy of delivery of approximately 97%. Immunohistochemical and Western blotting analyses revealed widespread, targeted inhibition of the expression of two well-known reference proteins, cyclophilin-B (38-68% knockdown) and glyceraldehyde 3-phosphate dehydrogenase (23-34% knockdown). These findings suggest that this novel procedure is likely to be useful in experimental investigations of neuropathophysiological mechanisms.


Journal of Biological Chemistry | 2015

Glyceraldehyde-3-phosphate Dehydrogenase Aggregates Accelerate Amyloid-β Amyloidogenesis in Alzheimer Disease.

Masanori Itakura; Hidemitsu Nakajima; Takeya Kubo; Yuko Semi; Satoshi Kume; Shusaku Higashida; Akihiro Kaneshige; Mitsuru Kuwamura; Naoki Harada; Akinori Kita; Yasu-Taka Azuma; Ryoichi Yamaji; Takashi Inui; Tadayoshi Takeuchi

Background: There is currently no strong evidence for a linkage between glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and Alzheimer disease (AD). Results: GAPDH aggregates enhanced amyloid-β peptide (Aβ) amyloidogenesis and augmented Aβ40-induced neurotoxicity, both in vitro and in vivo, concomitant with mitochondrial dysfunction. Conclusion: GAPDH aggregates accelerate Aβ amyloidogenesis. Significance: Aβ amyloidogenesis associated with GAPDH aggregation might underlie AD pathogenesis. Alzheimer disease (AD) is a progressive neurodegenerative disorder characterized by loss of neurons and formation of pathological extracellular deposits induced by amyloid-β peptide (Aβ). Numerous studies have established Aβ amyloidogenesis as a hallmark of AD pathogenesis, particularly with respect to mitochondrial dysfunction. We have previously shown that glycolytic glyceraldehyde-3-phosphate dehydrogenase (GAPDH) forms amyloid-like aggregates upon exposure to oxidative stress and that these aggregates contribute to neuronal cell death. Here, we report that GAPDH aggregates accelerate Aβ amyloidogenesis and subsequent neuronal cell death both in vitro and in vivo. Co-incubation of Aβ40 with small amounts of GAPDH aggregates significantly enhanced Aβ40 amyloidogenesis, as assessed by in vitro thioflavin-T assays. Similarly, structural analyses using Congo red staining, circular dichroism, and atomic force microscopy revealed that GAPDH aggregates induced Aβ40 amyloidogenesis. In PC12 cells, GAPDH aggregates augmented Aβ40-induced cell death, concomitant with disruption of mitochondrial membrane potential. Furthermore, mice injected intracerebroventricularly with Aβ40 co-incubated with GAPDH aggregates exhibited Aβ40-induced pyramidal cell death and gliosis in the hippocampal CA3 region. These observations were accompanied by nuclear translocation of apoptosis-inducing factor and cytosolic release of cytochrome c from mitochondria. Finally, in the 3×Tg-AD mouse model of AD, GAPDH/Aβ co-aggregation and mitochondrial dysfunction were consistently detected in an age-dependent manner, and Aβ aggregate formation was attenuated by GAPDH siRNA treatment. Thus, this study suggests that GAPDH aggregates accelerate Aβ amyloidogenesis, subsequently leading to mitochondrial dysfunction and neuronal cell death in the pathogenesis of AD.


Biochemical Journal | 2012

Lipocalin-type prostaglandin D synthase protects against oxidative stress-induced neuronal cell death

Ayano Fukuhara; Mao Yamada; Ko Fujimori; Yuya Miyamoto; Toshihide Kusumoto; Hidemitsu Nakajima; Takashi Inui

L-PGDS [lipocalin-type PGD (prostaglandin D) synthase] is a dual-functional protein, acting as a PGD2-producing enzyme and a lipid transporter. L-PGDS is a member of the lipocalin superfamily and can bind a wide variety of lipophilic molecules. In the present study we demonstrate the protective effect of L-PGDS on H2O2-induced apoptosis in neuroblastoma cell line SH-SY5Y. L-PGDS expression was increased in H2O2-treated neuronal cells, and the L-PGDS level was highly associated with H2O2-induced apoptosis, indicating that L-PGDS protected the neuronal cells against H2O2-mediated cell death. A cell viability assay revealed that L-PGDS protected against H2O2-induced cell death in a concentration-dependent manner. Furthermore, the titration of free thiols in H2O2-treated L-PGDS revealed that H2O2 reacted with the thiol of Cys65 of L-PGDS. The MALDI-TOF (matrix-assisted laser-desorption ionization-time-of-flight)-MS spectrum of H2O2-treated L-PGDS showed a 32 Da increase in the mass relative to that of the untreated protein, showing that the thiol was oxidized to sulfinic acid. The binding affinities of oxidized L-PGDS for lipophilic molecules were comparable with those of untreated L-PGDS. Taken together, these results demonstrate that L-PGDS protected against neuronal cell death by scavenging reactive oxygen species without losing its ligand-binding function. The novel function of L-PGDS could be useful for the suppression of oxidative stress-mediated neurodegenerative diseases.

Collaboration


Dive into the Hidemitsu Nakajima's collaboration.

Top Co-Authors

Avatar

Tadayoshi Takeuchi

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Yasu-Taka Azuma

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Kazuhiro Nishiyama

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Takeya Kubo

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Masanori Itakura

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Mitsuru Kuwamura

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Takashi Inui

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Yasuyuki Fujimoto

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Ai Morioka

Osaka Prefecture University

View shared research outputs
Top Co-Authors

Avatar

Natsuho Yoshida

Osaka Prefecture University

View shared research outputs
Researchain Logo
Decentralizing Knowledge