Network


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

Hotspot


Dive into the research topics where Takashi Okauchi is active.

Publication


Featured researches published by Takashi Okauchi.


The Journal of Neuroscience | 2007

Longitudinal, Quantitative Assessment of Amyloid, Neuroinflammation, and Anti-Amyloid Treatment in a Living Mouse Model of Alzheimer's Disease Enabled by Positron Emission Tomography

Jun Maeda; Bin Ji; Toshiaki Irie; Takami Tomiyama; Masahiro Maruyama; Takashi Okauchi; Matthias Staufenbiel; Nobuhisa Iwata; Maiko Ono; Takaomi C. Saido; Kazutoshi Suzuki; Hiroshi Mori; Makoto Higuchi; Tetsuya Suhara

We provide the first evidence for the capability of a high-resolution positron emission tomographic (PET) imaging system in quantitatively mapping amyloid accumulation in living amyloid precursor protein transgenic (Tg) mice. After the intravenous administration of N-[11C]methyl-2-(4′-methylaminophenyl)-6-hydroxybenzothiazole (or [11C]PIB for “Pittsburgh Compound-B”) with high-specific radioactivity, the Tg mice exhibited high-level retention of radioactivity in amyloid-rich regions. PET investigation for Tg mice over an extended range of ages, including longitudinal assessments, demonstrated age-dependent increase in radioligand binding consistent with progressive amyloid accumulation. Reduction in amyloid levels in the hippocampus of Tg mice was also successfully monitored by multiple PET scans along the time course of anti-amyloid treatment using an antibody against amyloid β peptide (Aβ). Moreover, PET scans with [18F]fluoroethyl-DAA1106, a radiotracer for activated glia, were conducted for these individuals parallel to amyloid imaging, revealing treatment-induced neuroinflammatory responses, the magnitude of which intimately correlated with the levels of pre-existing amyloid estimated by [11C]PIB. It is also noteworthy that the localization and abundance of [11C]PIB autoradiographic signals were closely associated with those of N-terminally truncated and modified Aβ, AβN3-pyroglutamate, in Alzheimers disease (AD) and Tg mouse brains, implying that the detectability of amyloid by [11C]PIB positron emission tomography is dependent on the accumulation of specific Aβ subtypes. Our results support the usefulness of the small animal-dedicated PET system in conjunction with high-specific radioactivity probes and appropriate Tg models not only for clarifying the mechanistic properties of amyloidogenesis in mouse models but also for preclinical tests of emerging diagnostic and therapeutic approaches to AD.


The Journal of Neuroscience | 2008

Imaging of Peripheral Benzodiazepine Receptor Expression as Biomarkers of Detrimental versus Beneficial Glial Responses in Mouse Models of Alzheimer's and Other CNS Pathologies

Bin Ji; Jun Maeda; Makoto Sawada; Maiko Ono; Takashi Okauchi; Motoki Inaji; Ming-Rong Zhang; Kazutoshi Suzuki; Kiyoshi Ando; Matthias Staufenbiel; John Q. Trojanowski; Virginia M.-Y. Lee; Makoto Higuchi; Tetsuya Suhara

We demonstrate the significance of peripheral benzodiazepine receptor (PBR) imaging in living mouse models of Alzheimers disease (AD) as biomarkers and functional signatures of glial activation. By radiochemically and immunohistochemically analyzing murine models of the two pathological hallmarks of AD, we found that AD-like Aβ deposition is concurrent with astrocyte-dominant PBR expression, in striking contrast with nonastroglial PBR upregulation in accumulations of AD-like phosphorylated tau. Because tau-induced massive neuronal loss was distinct from the marginal neurodegeneration associated with Aβ plaques in these models, cellular localization of PBR reflected deleterious and beneficial glial reactions to tau versus Aβ pathologies, respectively. This notion was subsequently examined in models of various non-AD neuropathologies, revealing the following reactive glial dynamics underlying differential PBR upregulation: (1) PBR(−) astrogliosis uncoupled with microgliosis or coupled with PBR(+) microgliosis associated with irreversible neuronal insults; and (2) PBR(+) astrogliosis coupled with PBR(− or ±) microgliosis associated with minimal or reversible neuronal toxicity. Intracranial transplantation of microglia also indicated that nontoxic microglia drives astroglial PBR expression. Moreover, levels of glial cell line-derived neurotrophic factor (GDNF) in astrocytes were correlated with astroglial PBR, except for increased GDNF in PBR(-) astrocytes in the model of AD-like tau pathology, thereby suggesting that PBR upregulation in astrocytes is an indicator of neurotrophic support. Together, PBR expressions in astrocytes and microglia reflect beneficial and deleterious glial reactions, respectively, in diverse neurodegenerative disorders including AD, pointing to new applications of PBR imaging for monitoring the impact of gliosis on the pathogenesis and treatment of AD.


Brain Research | 2007

Phase-dependent roles of reactive microglia and astrocytes in nervous system injury as delineated by imaging of peripheral benzodiazepine receptor.

Jun Maeda; Makoto Higuchi; Motoki Inaji; Bin Ji; Eisuke Haneda; Takashi Okauchi; Ming-Rong Zhang; Kazutoshi Suzuki; Tetsuya Suhara

Elevated levels of peripheral benzodiazepine receptor (PBR) in glia have been documented in diverse nervous system injuries, while the identity and spatiotemporal characteristics of the cells showing upregulation of PBR remain elusive. We examined the astrocytic and microglial expressions of PBR in rat brains during the duration of ethanol-induced neuronal insults in order to clarify the significance of PBR as a biomarker capable of detecting a distinctive subpopulation of these glial cells involved in the impairment and protection of neurons. The levels of PBR, as determined by autoradiographic analysis using a specific radioligand, [11C]DAA1106, began to significantly increase at 3 days after intrastriatal injection of ethanol, and peaked at 7 days. This was consistent with the results of double immunofluorescence staining and high-resolution emulsion autoradiography, which revealed upregulation of PBR in both microglia and astrocytes proliferating in nonoverlapping compartments of the injury site. Notably, increased expression of PBR in astrocytes was transiently observed in a manner parallel to the centripetal migration of these cells to the inflammatory lesion, which may be a response indispensable to the protection of intact tissue. Thereafter, astrocytic PBR was barely detectable, despite the presence of numerous glial fibrillary acidic protein-immunoreactive astrocytes forming glial scarring. By contrast, intense PBR signals were persistently present in microglia localized to the injury epicenter up to 90 days, notwithstanding a gradual reduction in the number of ionized calcium binding adapter molecule-1-positive amoeboid microglia between 7 and 90 days. The long-lasting PBR expression in microglia was finally supported by in vivo positron emission tomography imaging, and suggests that inflammatory tissue damage is potentially expandable unless it is tightly sealed by astrocytic scar. The present findings collectively support the utility of PBR in identifying a unique temporal pattern of astrocytic and microglial activation that conventional glial markers hardly pursue.


The International Journal of Neuropsychopharmacology | 1999

Extrastriatal dopamine D2 receptor density and affinity in the human brain measured by 3D PET.

Tetsuya Suhara; Yasuhiko Sudo; Takashi Okauchi; Jun Maeda; Koichi Kawabe; Kazutoshi Suzuki; Yoshiro Okubo; Yoshifumi Nakashima; Hiroshi Ito; Shuji Tanada; Christer Halldin; Lars Farde

The aim of the present study was to quantify the density and affinity of human extrastriatal dopamine D2 receptors using positron emission tomography (PET). [(11)C]FLB-457, a high-affinity dopamine D2 receptor antagonist with various specific radioactivities (SA) was used. Eight healthy male subjects, age 20-35 yr, participated twice or three times at different SAs (1-279 GBq/ µmol), and serial dynamic scans were performed in the 3D data acquisition mode. The peak of the specific binding was not well defined with high SA due to the flatness of the curves after 60 min but was observed within the PET measurement. In the experiment with low SA, the peak came earlier than that with high SA. Scatchard analysis was performed using the maximal specific binding value (transient equilibrium) and the radioactivity in the cerebellum as free ligand concentration. The highest density was observed in the thalamus (2.3+/-0.6 pmol/ml), followed by the temporal cortex (1.5+/-0.5 pmol/ml), hippocampus (1.4+/-0.5 pmol/ml), parietal cortex (0.9+/-0.4 pmol/ml), frontal cortex (0.8+/-0.2 pmol/ml) and occipital cortex (0.7+/-0.3 pmol/ml). There was no significant difference in K(d) values in these six regions. The present results demonstrate that dopamine D2 receptor densities in the extrastriatal regions were only 2-8% of that in the striatum. Although the density of extrastriatal dopamine D2 receptor was low, significant regional differences were observed in the present study, as reported in postmortem studies.


The Journal of Neuroscience | 2011

In vivo positron emission tomographic imaging of glial responses to amyloid-beta and tau pathologies in mouse models of Alzheimer's disease and related disorders.

Jun Maeda; Ming-Rong Zhang; Takashi Okauchi; Bin Ji; Maiko Ono; Satoko Hattori; Katsushi Kumata; Nobuhisa Iwata; Takaomi C. Saido; John Q. Trojanowski; Virginia M.-Y. Lee; Matthias Staufenbiel; Takami Tomiyama; Hiroshi Mori; Toshimitsu Fukumura; Tetsuya Suhara; Makoto Higuchi

Core pathologies of Alzheimers disease (AD) are aggregated amyloid-β peptides (Aβ) and tau, and the latter is also characteristic of diverse neurodegenerative tauopathies. These amyloid lesions provoke microglial activation, and recent neuroimaging technologies have enabled visualization of this response in living brains using radioligands for the peripheral benzodiazepine receptor also known as the 18 kDa translocator protein (TSPO). Here, we elucidated contributions of Aβ and tau deposits to in vivo TSPO signals in pursuit of mechanistic and diagnostic significance of TSPO imaging in AD and other tauopathies. A new antibody to human TSPO revealed induction of TSPO-positive microgliosis by tau fibrils in tauopathy brains. Emergence of TSPO signals before occurrence of brain atrophy and thioflavin-S-positive tau amyloidosis was also demonstrated in living mice transgenic for mutant tau by positron emission tomography (PET) with two classes of TSPO radioligands, [11C]AC-5216 and [18F]fluoroethoxy-DAA1106. Meanwhile, only modest TSPO elevation was observed in aged mice modeling Aβ plaque deposition, despite the notably enhanced in vivo binding of amyloid radiotracer, [11C]Pittsburgh Compound-B, to plaques. In these animals, [11C]AC-5216 yielded better TSPO contrasts than [18F]fluoroethoxy-DAA1106, supporting the possibility of capturing early neurotoxicity with high-performance TSPO probes. Furthermore, an additional line of mice modeling intraneuronal Aβ accumulation displayed elevated TSPO signals following noticeable neuronal loss, unlike TSPO upregulation heralding massive neuronal death in tauopathy model mice. Our data corroborate the utility of TSPO-PET imaging as a biomarker for tau-triggered toxicity, and as a complement to amyloid scans for diagnostic assessment of tauopathies with and without Aβ pathologies.


Brain Research | 2005

Correlation between quantitative imaging and behavior in unilaterally 6-OHDA-lesioned rats

Motoki Inaji; Takashi Okauchi; Kiyoshi Ando; Jun Maeda; Yuji Nagai; Takahito Yoshizaki; Hideyuki Okano; Tadashi Nariai; Kikuo Ohno; Shigeru Obayashi; Makoto Higuchi; Tetsuya Suhara

We evaluated correlation between neurochemical and functional alterations of the nigrostriatal dopaminergic system in rat brains lesioned with 6-hydroxydopamine (6-OHDA), that model hemi-Parkinsons disease (PD), by using three different quantitative in vivo and in vitro methods. Rats unilaterally lesioned with different doses of 6-OHDA underwent two types of in vivo experiments: (1) a rotational behavioral study with methamphetamine (MAP) or apomorphine (APO); and (2) a positron emission tomography (PET) study with [11C]PE2I (radioligand for dopamine transporters) or [11C]raclopride (radioligand for dopamine D2 receptors). An in vitro autoradiographic study with the same radioligands was also conducted. The number of rotations after the MAP or APO injection increased with increased doses of 6-OHDA. The in vitro and in vivo binding of [11C]PE2I dose-dependently decreased in response to the 6-OHDA injections, while that of [11C]raclopride dose-dependently increased. There was a significant negative hyperbolic correlation between the number of rotations after MAP injection and the binding of [11C]PE2I. In contrast, there was a significant positive linear correlation between the number of rotations after APO injections and the binding of [11C]raclopride. These results robustly reveal a molecular pharmacological basis of parkinsonian symptoms in animal models of PD, and indicate the utility and validity of in vivo PET measurements in assessing pre- and post-synaptic dopaminergic functions.


The Journal of Neuroscience | 2009

Neuroimaging and Physiological Evidence for Involvement of Glutamatergic Transmission in Regulation of the Striatal Dopaminergic System

Masaki Tokunaga; Nicholas Seneca; Ryong-Moon Shin; Jun Maeda; Shigeru Obayashi; Takashi Okauchi; Yuji Nagai; Ming-Rong Zhang; Ryuji Nakao; Hiroshi Ito; Robert B. Innis; Christer Halldin; Kazutoshi Suzuki; Makoto Higuchi; Tetsuya Suhara

Aberrant neurotransmissions via glutamate and dopamine receptors have been the focus of biomedical research on the molecular basis of psychiatric disorders, but the mode of their interaction is yet to be uncovered. In this study, we demonstrated the pharmacological reversal of methamphetamine-stimulated dopaminergic overflow by suppression of group I metabotropic glutamate (mGlu) receptor in living primates and rodents. In vivo positron emission tomography (PET) was conducted on cynomolgus monkeys and rats using a full agonistic tracer for dopamine D2/3 receptor, [11C]MNPA [(R)-2-11CH3O-N-n-propylnorapomorphine], and fluctuation of kinetic data resulting from anesthesia was avoided by scanning awake subjects. Excessive release of dopamine induced by methamphetamine and abolishment of this alteration by treatment with an antagonist of group I mGlu receptors, 2-methyl-6-(phenylethynyl)pyridine (MPEP), were measured in both species as decreased binding potential because of increased dopamine and its recovery to baseline levels, respectively. Counteraction of MPEP to the methamphetamine-induced dopamine spillover was also supported neurochemically by microdialysis of unanesthetized rat striatum. Moreover, patch-clamp electrophysiological assays using acute brain slices prepared from rats indicated that direct targets of MPEP mechanistically involved in the effects of methamphetamine are present locally within the striatum. Because MPEP alone did not markedly alter the baseline dopaminergic neurotransmission according to our PET and electrophysiological data, the present findings collectively extend the insights on dopamine–glutamate cross talk from extrastriatal localization of responsible mGlu receptors to intrastriatal synergy and support therapeutic interventions in case of disordered striatal dopaminergic status using group I mGlu receptor antagonists assessable by in vivo imaging techniques.


Journal of Neurotrauma | 2010

Glial cell-mediated deterioration and repair of the nervous system after traumatic brain injury in a rat model as assessed by positron emission tomography.

Iwae Yu; Motoki Inaji; Jun Maeda; Takashi Okauchi; Tadashi Nariai; Kikuo Ohno; Makoto Higuchi; Tetsuya Suhara

Traumatic brain injury (TBI) is one of the most acute degenerative pathologies in the central nervous system, and in vivo indices enabling an assessment of TBI on a mechanistic basis have yet to be established. The aim of this work was to pursue neuroinflammatory changes and their link to functional disruptions of traumatically-damaged neurons in a rat model of TBI by longitudinal positron emission tomographic (PET) assays. TBI was induced in the unilateral frontal cortex of craniotomied rats according to a lateral fluid percussion brain injury protocol. The use of [(18)F]fluoroethyl-DAA1106 as a PET tracer for translocator protein (TSPO) permitted demonstration of the inflammatory response to the injury, peaking at 1 week after impact. This alteration was parallel to metabolic deficits assessed by PET with [(18)F]fluorodeoxyglucose, but the difference in TSPO levels between impacted and non-impacted frontal cortices was more than threefold of the interlateral metabolic difference, indicating superiority of TSPO imaging for sensitive detection of post-traumatic pathologies. Comparative PET, autoradiographic. and immunohistochemical investigations illustrated the primary contribution of hypertrophic microglia and macrophages to acute TSPO signals in the vicinity of the impact. Astrocytes also formed a TSPO-positive glial scar encompassing necrotic inflammation, and were clustered with PET-detectable TSPO signals in the bilateral external and internal capsules at late stages, putatively reacting with diffuse axonal injury. These observations support the applicability of TSPO-PET as an imaging-based preclinical and clinical biomarker assay in TBI, and indicate its potential capability to clarify aggressive and protective roles of glial responses to injury when combined with emerging anti-inflammatory and immunomodulatory treatments.


Neuroscience Letters | 2003

Different roles of group I and group II metabotropic glutamate receptors on phencyclidine-induced dopamine release in the rat prefrontal cortex

Jun Maeda; Tetsuya Suhara; Takashi Okauchi; Jun'ichi Semba

The dopamine system in the limbic-prefrontal cortex has been assumed to play an important role in the cognitive dysfunction of schizophrenia and phencyclidine (PCP)-induced psychosis. In the present study, the role of metabotropic glutamate (mGlu) receptor subtypes on PCP-induced cortical dopamine release was investigated using the microdialysis technique. Infusion of 50 and 100 microM of non-selective mGlu receptor agonist trans-(1S,3R)-1-amino-1,3-cyclopentanedicarboxylic acid inhibited PCP-induced dopamine release, while the basal dopamine level was not significantly affected. A similar inhibition of PCP-induced dopamine release was observed with 100 and 500 microM of selective group I mGlu receptor agonist, (+)-3-hydroxy-phenylglycine. On the other hand, infusion of 10 microM of selective group II mGlu receptor agonist, 2-(2, 3-dicarboxycyclopropyl)-glycine, enhanced the PCP-induced dopamine increase. These results suggest that group I and II mGlu receptors exert opposite modulations on the PCP-induced dopamine release.


Journal of Pharmacology and Experimental Therapeutics | 2013

Evaluation of Oatp and Mrp2 Activities in Hepatobiliary Excretion Using Newly Developed Positron Emission Tomography Tracer [11C]Dehydropravastatin in Rats

Tomotaka Shingaki; Tadayuki Takashima; Ryosuke Ijuin; Xuan Zhang; Tomohiro Onoue; Yumiko Katayama; Takashi Okauchi; Emi Hayashinaka; Yilong Cui; Yasuhiro Wada; Masaaki Suzuki; Kazuya Maeda; Hiroyuki Kusuhara; Yuichi Sugiyama; Yasuyoshi Watanabe

We developed a pravastatin derivative, sodium (3R,5R)-3,5-dihydroxy-7-((1S,2S,6S,8S)-6-hydroxy-2-methyl-8-((1-[11C]-(E)-2-methyl-but-2-enoyl)oxy)-1,2,6,7,8,8a-hexahydronaphthalen-1-yl)heptanoate ([11C]DPV), as a positron emission tomography (PET) probe for noninvasive measurement of hepatobiliary transport, and conducted pharmacokinetic analysis in rats as a feasibility study for future clinical study. Transport activities of DPV in freshly isolated rat hepatocytes and rodent multidrug resistance–associated protein 2 (rMrp2; human, MRP2)-expressing membrane vesicles were similar to those of pravastatin. Rifampicin diminished the uptake of DPV and pravastatin by the hepatocytes, with similar inhibition potency. [11C]DPV underwent biotransformation to produce at least two metabolites in rat, but metabolism of [11C]DPV occurred negligibly in human hepatocytes during a 90-minute incubation. After intravenous injection, [11C]DPV was mainly distributed to the liver and kidneys, where the tissue uptake clearances (CLuptake,liver and CLuptake,kidney) were blood-flow–limited (73.6 ± 4.8 and 24.6 ± 0.6 ml/min per kilogram, respectively). Systemic elimination of [11C]DPV was delayed in rifampicin-treated rat and an Mrp2-deficient mutant rat, Eisai hyperbilirubinemic mutant rat (EHBR). Rifampicin treatment decreased both CLuptake,liver and CLuptake,kidney of [11C]DPV by 30% (P < 0.05), whereas these parameters were unchanged in EHBR. Meanwhile, the canalicular efflux clearance (CLint,bile) of [11C]DPV, which was 12.2 ± 1.5 ml/min per kilogram in the control rat, decreased by 60% and 89% in rifampicin-treated rat and EHBR (P < 0.05), respectively. These results indicate that [11C]DPV is taken up into the liver by organic anion-transporting polypeptides (rodent, Oatps; human, OATP) and excreted into bile by Mrp2 in rat, and that rifampicin may inhibit Mrp2 as well as Oatps, and consequently increase systemic exposure of [11C]DPV. PET using [11C]DPV is feasible for studies prior to the future clinical investigation of OATP and MRP2 functionality, especially for personalized medicine.

Collaboration


Dive into the Takashi Okauchi's collaboration.

Top Co-Authors

Avatar

Tetsuya Suhara

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Jun Maeda

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Kazutoshi Suzuki

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Ming-Rong Zhang

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Shigeru Obayashi

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Makoto Higuchi

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Terushi Haradahira

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Motoki Inaji

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Yuji Nagai

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Takayo Kida

National Institute of Radiological Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge