Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
Biol Pharm Bull ; 46(11): 1576-1582, 2023.
Article in English | MEDLINE | ID: mdl-37914360

ABSTRACT

Chinese artichoke tuber (Stachys sieboldii Miq.) is used as an herbal medicine as well as edible food. This study examined the effect of the Chinese artichoke extracts on the nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway that induces the expression of antioxidant enzymes to explore its novel characteristics. Hot water extracts exhibited relatively high ARE activity. ARE activity was observed in two fractions when the hot water extracts were separated in the presence of trifluoroacetic acid using HPLC. Conversely, the highly active fraction disappeared when the hot water extracts were separated in the absence of trifluoroacetic acid. These results indicate that acidic degradation produces active ingredients. The structural analysis of the two active fractions identified harpagide, which is an iridoid glucoside, and harpagogenin. In vitro experiments revealed that harpagide was converted into harpagogenin under acidic conditions and that harpagogenin, but not harpagide, had potent ARE activity. Therefore, this study identified harpagogenin, which is an acid hydrolysate of harpagide, as an ARE activator and suggests that Nrf2-ARE pathway activation by Chinese artichoke contributes to the antioxidative effect.


Subject(s)
Stachys , Antioxidant Response Elements , Antioxidants/pharmacology , NF-E2-Related Factor 2 , Plant Extracts/pharmacology , Plant Extracts/chemistry , Stachys/chemistry , Trifluoroacetic Acid , Water
2.
Article in English | MEDLINE | ID: mdl-34894926

ABSTRACT

The present study investigated the therapeutic effects of the curcumin derivative 3-[(1E)-2-(1H-indol-6-yl)ethenyl]-5-[(1E)-2-[2-methoxy-4-(2-pyridylmethoxy)phenyl]ethenyl]-1H-pyrazole (GT863) in amyotrophic lateral sclerosis (ALS). The inhibitory effect of GT863 on superoxide dismutase 1 (SOD1) aggregation was evaluated in cell-free assays. GT863 interfered with the conformational changes of the SOD1 protein and later, oligomeric aggregation. Furthermore, its antioxidant, anti-inflammatory, and neuroprotective effects were evaluated in cell-free and cultured cell assays. GT863 inhibited H2O2- and glutamate-induced cytotoxicity and activated an antioxidant responsive element pathway. Additionally, in vivo effects of GT863 in the ALS mice model were evaluated by its oral administration to H46R mutant SOD1 transgenic mice. Rotarod test showed that GT863 administration significantly slowed the progression of motor dysfunction in the mice. In addition, GT863 substantially reduced highly-aggregated SOD1, further preserving large neurons in the spinal cord of GT863-treated mice. Collectively, these results indicated that GT863 could be a viable therapeutic agent with multiple vital actions for the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Curcumin , Mice , Animals , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Antioxidants/therapeutic use , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/therapeutic use , Mice, Transgenic , Superoxide Dismutase/genetics , Disease Models, Animal , Spinal Cord/metabolism
3.
Brain Res Bull ; 173: 45-52, 2021 08.
Article in English | MEDLINE | ID: mdl-33989723

ABSTRACT

Cerebral infarct is caused by cerebrovascular occlusion and results in brain damage. Although many rodent models of cerebral infarct exist, there is none based on zebrafish. In this study, we developed a novel ischemia-reperfusion model induced by hypoxic treatment using zebrafish. We first examined the changes in blood flow under hypoxic conditions. Hypoxic treatment interrupted the blood flow in 4 dpf (days post fertilization) zebrafish larvae. To quantify the trunk and cerebral blood flow, we selected the middle mesencephalic central artery (MMCtA) as a cerebral blood vessel and the dorsal aorta (DA) as a blood vessel of the trunk. Interestingly, the interruption of blood flow in MMCtA preceded that in DA. Considering these results, we hypothesized that reoxygenation immediately after hypoxia-induced cerebral ischemia leads to reperfusion. As a result, hypoxia-reoxygenation (H/R) treatment induced ischemia-reperfusion in cerebral vessels. Furthermore, brain cell death was increased 24 h after H/R treatment. Transgenic zebrafish (HuC:kaede), with neuronal cells expressing the kaede fluorescent protein, was used to investigate the effect of H/R on neuronal cells. The H/R treatment reduced the fluorescence intensity of kaede. Besides, glial fibrillary acidic protein immunoreactivity in H/R-treated larvae was significantly increased. In conclusion, H/R-treated zebrafish larvae may provide a novel ischemia-reperfusion model.


Subject(s)
Brain Ischemia/physiopathology , Cell Death/physiology , Cerebrovascular Circulation/physiology , Neurons/physiology , Reperfusion Injury/physiopathology , Animals , Animals, Genetically Modified , Disease Models, Animal , Zebrafish
4.
Neurosci Lett ; 736: 135268, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32712353

ABSTRACT

Parkinson disease (PD) is a neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the substantia nigra, and oxidative stress is thought to contribute to this pathogenesis. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway, which induces the production of antioxidant enzymes, is thereby a potential target for therapeutics to reduce neurodegeneration in PD. Previously, we identified TPNA10168 from a chemical library as an activator of the Nrf2-ARE pathway, and the present study examined the effects of TPNA10168 on an in vivo PD model. Subcutaneous administration of TPNA10168 was associated with inhibited dopaminergic neuronal loss and behavioral impairment in 6-hydroxydopamine-induced PD model mice. Heme oxygenase-1 (HO-1) is an antioxidant enzyme expressed downstream of the Nrf2-ARE signaling pathway, and we observed that HO-1 protein levels were upregulated by TPNA10168 in the mouse brain. These results suggest that TPNA10168 inhibits dopaminergic neuronal death in PD model mice, and that upregulation of HO-1 might participate in this effect.


Subject(s)
Antioxidant Response Elements/drug effects , Cell Death/drug effects , Dopaminergic Neurons/drug effects , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/agonists , Parkinson Disease, Secondary/drug therapy , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Mice , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Up-Regulation/drug effects
5.
Biol Pharm Bull ; 43(1): 184-187, 2020.
Article in English | MEDLINE | ID: mdl-31902924

ABSTRACT

We have previously isolated 2',3'-dihydroxy-4',6'-dimethoxychalcone (DDC) from green perilla leaves as the activator of the nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway. This study aims to evaluate the effects of DDC against glutamate neurotoxicity using rat primary cortical cultures. Treatment of cultures with DDC for 24 h before glutamate exposure significantly inhibited glutamate neurotoxicity in a concentration-dependent manner. The involvement of hemeoxygenase-1 (HO-1) and reduced glutathione (GSH) in the protective effects of DDC on cortical cultures was also evaluated. While an HO-1 inhibitor did not have a significant effect on DDC-induced neuroprotection, a γ-glutamylcystein synthetase (γ-GCS) inhibitor significantly suppressed the protective effect of DDC. In an astrocyte culture, DDC induced a marked increase in the levels of intracellular reduced GSH. These results suggest that DDC mainly activates the Nrf2-ARE pathway of astrocytes, resulting in the increased extracellular release of reduced GSH, protecting neurons from glutamate neurotoxicity.


Subject(s)
Astrocytes/drug effects , Chalcones/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Astrocytes/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Fetus , Glutamic Acid , Glutathione/metabolism , Heme Oxygenase-1/metabolism , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Rats, Wistar
6.
Biochem Biophys Res Commun ; 519(4): 777-782, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31551151

ABSTRACT

Astrocytes have been reported to exhibit neuroprotective action via various chemokines. Reports of the chemokine CCL6 in central nervous system cells show expression in cultured microglia, but many unexplained effects on neurons and astrocytes remain. In this study, cultured cerebral cortical neurons, astrocytes, and a mixed culture system were constructed, and expression levels of CCL6 and its effects on glutamate neurotoxicity were examined. When neuron cultures and neuron-astrocyte mixed cultures were treated with glutamate, neuronal cell death was observed in both, but was induced by lower concentrations of glutamate in monocultured neurons. In addition, pretreatment of neuron cultures with conditioned media from neuron-astrocyte mixed cultures inhibited glutamate neurotoxicity. CCL6 expression was not observed in fluorescence activated cell sorting analyses of neuron and astrocyte cultures, but was observed in astrocytes from cocultures of neurons and astrocytes. Higher CCL6 concentrations were found in media from cocultures of neurons and astrocytes than in culture media from neuron cultures. Pretreatment of neuron cell cultures with CCL6 for 24 h also protected against glutamate neurotoxicity. This protective effect was suppressed by an antagonist of the chemokine receptor CCR1. Furthermore, glutamate neurotoxicity in mixed neuron and astrocyte cultures was enhanced by pretreatments with the CCR1 antagonist. Finally, cotreatments with the phosphatidylinositol-3 kinase (PI3K) inhibitor and CCL6 abolished the neuroprotective effects of CCL6. These data suggest that astrocytes protect neurons by activating CCR1 in neurons. Moreover, this neuroprotective action of astrocyte CCL6 is mediated by CCR1, and downstream by PI3K.


Subject(s)
Astrocytes/metabolism , Chemokines, CC/genetics , Neurons/metabolism , Neuroprotective Agents , Animals , Astrocytes/drug effects , Cells, Cultured , Chemokines, CC/metabolism , Dose-Response Relationship, Drug , Glutamic Acid/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar
7.
J Pharmacol Sci ; 141(1): 17-24, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31540843

ABSTRACT

Oxidative stress has been implicated in the pathogenesis of allergic contact dermatitis. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway, an in vivo antioxidant system, induces antioxidant enzymes. In our previous studies, we isolated 2',3'-dihydroxy-4',6'-dimethoxychalcone (DDC) from green perilla and identified it as a novel activator of the Nrf2-ARE pathway. We also discovered that it exerted cytoprotective effects against oxidative stress in PC12 cells. However, its effects on skin disease model animals in vivo remain unclear. In the present study, auricular thickness time-dependently increased with the repeated application of picryl chloride, and significant increases were observed from Day 2 in chronic contact hypersensitivity (cCHS) model mice. Histological changes, such as higher numbers of cells in the epidermis, were observed with increases in auricular thickness. The administration of DDC every two days from Day 6 suppressed the increases in auricular thickness and the number of scratching events in a dose-dependent manner. The expression levels of antioxidant enzymes increased in the mouse auricle 24 h after the administration of DDC. These results presume that DDC inhibits increases in auricular thickness in cCHS mice by up-regulating the expression of antioxidative enzymes through the activation of the Nrf2-ARE pathway.


Subject(s)
Chalcones/isolation & purification , Chalcones/pharmacology , Dermatitis, Contact/pathology , Ear Auricle/pathology , Perilla/chemistry , Animals , Antioxidant Response Elements , Chronic Disease , Dermatitis, Contact/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/metabolism , Inflammation , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , PC12 Cells , Rats
8.
Biol Pharm Bull ; 42(11): 1942-1946, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31462605

ABSTRACT

Amyloid ß protein (Aß) causes neurotoxicity and cognitive impairment in Alzheimer's disease (AD). Oxidative stress is closely related to the pathogenesis of AD. We have previously reported that 2',3'-dihydroxy-4',6'-dimethoxychalcone (DDC), a component of green perilla, enhances cellular resistance to oxidative damage through the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway. Here, we investigated the effects of DDC on cortical neuronal death induced by Aß. When Aß and DDC had been preincubated for 3 h, the aggregation of Aß was significantly suppressed. In this condition, we found that DDC provided a neuroprotective action on Aß-induced cytotoxicity. Treatment with DDC for 24 h increased the expression of heme oxygenase-1 (HO-1), and this was controlled by the activation of the Nrf2-ARE pathway. However, DDC did not affect Aß-induced neuronal death under any of these conditions. These results suggest that DDC prevents the aggregation of Aß and inhibits neuronal death induced by Aß, and although it activates the Nrf2-ARE pathway, this mechanism is less involved its neuroprotective effect.


Subject(s)
Amyloid beta-Peptides/metabolism , Chalcone/analogs & derivatives , Chalcone/pharmacology , NF-E2-Related Factor 2/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/metabolism , Heme Oxygenase-1/metabolism , Neurotoxicity Syndromes/metabolism , Oxidative Stress/drug effects , Perilla , Rats, Wistar , Reactive Oxygen Species/metabolism
9.
Biol Pharm Bull ; 42(11): 1936-1941, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31462615

ABSTRACT

Skin exposure to UV rays causes the production of reactive oxygen species (ROS), and it is a major risk factor for various skin disorders and diseases. In particular, exposure to UV-A is a major cause of photoaging. We have previously isolated 2',3'-dihydroxy-4',6'-dimethoxychalcone (DDC) from green perilla leaves as an activator of the nuclear factor erythroid 2-related factor-2 (Nrf2)-antioxidant response element (ARE) and demonstrated the protective effects of DDC both in vitro and in vivo in PC12 cells and Parkinson's disease models, respectively. In this study, we used HaCaT cells to examine the effects of DDC on ROS production and cell damage induced by UV-A. Our results indicated that UV-A irradiation in HaCaT cells increased ROS production in an energy-dependent manner. In addition, cell viability decreased in an energy-dependent manner 24 h after UV-A irradiation. However, treatment with DDC 24 h prior to UV-A irradiation significantly suppressed UV-A radiation-induced ROS production. In addition, DDC showed cytoprotective effects when used 24 h before and after UV-A irradiation. Treatment with DDC for 24 h also increased the expression levels of heme oxygenase-1 (HO-1) in a concentration-dependent manner. Pretreatment with the HO-1 inhibitor followed by DDC treatment before UV-A irradiation for 24 h reduced ROS production and the cytoprotective effect. These results suggest that DDC increases the expression levels of HO-1 and protects HaCaT cells through the suppression of UV radiation-induced ROS production.


Subject(s)
Chalcones/pharmacology , Ultraviolet Rays/adverse effects , Animals , Cell Line , Cell Survival/drug effects , Heme Oxygenase-1/metabolism , Humans , Keratinocytes , NF-E2-Related Factor 2 , Perilla , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Skin/metabolism
10.
Sci Rep ; 9(1): 11922, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31417133

ABSTRACT

Donepezil, a therapeutic drug for Alzheimer's disease, ameliorates cognitive dysfunction through selective inhibition of acetylcholinesterase. However, recent studies have also reported off-target effects of donepezil that likely contribute to its therapeutic effects. In this study, we investigated the (i) role of donepezil in amyloid precursor protein (APP) processing and (ii) involvement of sorting nexin protein 33 (SNX33), a member of the sorting nexin protein family, in this processing. Results showed that donepezil induces an increase in SNX33 expression in primary cortical neurons. The secretion of sAPPα in culture media increased, whereas the expression of full-length APP in the cell lysate remained unchanged. Exposure of cortical cultures to donepezil led to a decrease in amyloid ß (Aß) protein levels in a concentration- and time-dependent manner. This decrease was not affected by concomitant treatment with acetylcholine receptor antagonists. SNX33 knockdown by target-specific morpholino oligos inhibited the effects of donepezil. Donepezil treatment increased cell membrane surface expression of APP in SNX33 expression-dependent manner. These results suggested that donepezil decreases the level of Aß by increasing SNX33 expression and APP cleavage by α-secretase in cortical neurons.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cerebral Cortex/cytology , Donepezil/pharmacology , Endocytosis , Neurons/metabolism , Sorting Nexins/genetics , Up-Regulation , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cholinergic Antagonists/pharmacology , Donepezil/therapeutic use , Endocytosis/drug effects , Morpholinos/pharmacology , Neurons/drug effects , Protein Processing, Post-Translational/drug effects , Rats, Wistar , Receptors, Cholinergic/metabolism , Sorting Nexins/metabolism , Up-Regulation/drug effects
11.
Front Neural Circuits ; 13: 77, 2019.
Article in English | MEDLINE | ID: mdl-31998081

ABSTRACT

Neural circuits interconnect to organize large-scale networks that generate perception, cognition, memory, and behavior. Information in the nervous system is processed both through parallel, independent circuits and through intermixing circuits. Analyzing the interaction between circuits is particularly indispensable for elucidating how the brain functions. Monosynaptic circuit tracing with glycoprotein (G) gene-deleted rabies viral vectors (RVΔG) comprises a powerful approach for studying the structure and function of neural circuits. Pseudotyping of RVΔG with the foreign envelope EnvA permits expression of transgenes such as fluorescent proteins, genetically-encoded sensors, or optogenetic tools in cells expressing TVA, a cognate receptor for EnvA. Trans-complementation with rabies virus glycoproteins (RV-G) enables trans-synaptic labeling of input neurons directly connected to the starter neurons expressing both TVA and RV-G. However, it remains challenging to simultaneously map neuronal connections from multiple cell populations and their interactions between intermixing circuits solely with the EnvA/TVA-mediated RV tracing system in a single animal. To overcome this limitation, here, we multiplexed RVΔG circuit tracing by optimizing distinct viral envelopes (oEnvX) and their corresponding receptors (oTVX). Based on the EnvB/TVB and EnvE/DR46-TVB systems derived from the avian sarcoma leukosis virus (ASLV), we developed optimized TVB receptors with lower or higher affinity (oTVB-L or oTVB-H) and the chimeric envelope oEnvB, as well as an optimized TVE receptor with higher affinity (oTVE-H) and its chimeric envelope oEnvE. We demonstrated independence of RVΔG infection between the oEnvA/oTVA, oEnvB/oTVB, and oEnvE/oTVE systems and in vivo proof-of-concept for multiplex circuit tracing from two distinct classes of layer 5 neurons targeting either other cortical or subcortical areas. We also successfully labeled common input of the lateral geniculate nucleus to both cortico-cortical layer 5 neurons and inhibitory neurons of the mouse V1 with multiplex RVΔG tracing. These oEnvA/oTVA, oEnvB/oTVB, and oEnvE/oTVE systems allow for differential labeling of distinct circuits to uncover the mechanisms underlying parallel processing through independent circuits and integrated processing through interaction between circuits in the brain.


Subject(s)
Genetic Vectors/metabolism , Glycoproteins/metabolism , Nerve Net/metabolism , Neuroanatomical Tract-Tracing Techniques/methods , Rabies virus/metabolism , Visual Cortex/metabolism , Animals , Cricetinae , Gene Deletion , Genetic Vectors/administration & dosage , Genetic Vectors/analysis , Genetic Vectors/genetics , Glycoproteins/administration & dosage , Glycoproteins/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/chemistry , Nerve Net/drug effects , Rabies virus/chemistry , Rabies virus/genetics , Visual Cortex/chemistry , Visual Cortex/drug effects
12.
Yakugaku Zasshi ; 138(9): 1127-1133, 2018.
Article in Japanese | MEDLINE | ID: mdl-30175756

ABSTRACT

 I joined efforts to promote the pharmaceutical education system by participating in committees for developing a model core curriculum for pharmaceutical sciences (core curriculum), in pharmacist educator workshops, in the development of a pharmaceutical common achievement test, evaluation of pharmaceutical education programs, and the creation of a national examination for pharmacists. This review outlines the efforts to reform these pharmaceutical education systems. The core curriculum was prepared under the initiative of the Pharmaceutical Society of Japan. Pharmacist educator workshops were frequently held for the training of educators engaged in the six-year undergraduate course. The Subcommittee of Pharmaceutical Sciences of the Science Council of Japan also held workshops for the development of pharmaceutical education. Under these efforts, the education system consisted of the six- and four-year courses started in 2006. The Ministry of Health, Labour and Welfare revised the national examination for pharmacists. More than 10 years after formulating the core curriculum, the Ministry of Education, Culture, Sports, Science and Technology led the reform of the core curriculum in 2013. The basic idea of the revised core curriculum is outcome-based education. Minor revisions to the national examination for pharmacists were also made following this revision of the core curriculum. The Subcommittee of Pharmaceutical Sciences of the Science Council of Japan conducted reference standards for pharmaceutical sciences to promote education and research in the four-year course. Thus, we created education systems for developing pharmacists and researchers capable of contributing to medical care through the creation, production, and proper use of medicines.


Subject(s)
Education, Pharmacy/standards , Education, Pharmacy/trends , Academic Success , Certification , Clinical Competence , Curriculum , Education , Education, Pharmacy/methods , Educational Status , Japan , Societies, Pharmaceutical/organization & administration , Time Factors
13.
J Pharmacol Sci ; 138(1): 9-15, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30243517

ABSTRACT

10-oxo-trans-11-octadecenoic acid (KetoC) and 10-hydroxy-cis-12-octadecenoic acid (HYA) are long-chain fatty acids generated from linoleic acid by the gut lactic acid bacterium Lactobacillus plantarum. These fatty acids have been reported to have anti-inflammatory activity in the intestine. However, little is known about their effects in the brain. In this study, we aimed to investigate the effects of these fatty acids on lipopolysaccharide (LPS)-induced inflammatory processes in mouse microglial cells (BV-2 cells). KetoC and HYA inhibited LPS-induced nitric oxide (NO) production and suppressed the expression of inducible NO synthase in BV-2 cells. NO changes in these inhibitory effects were observed with AH7614, a G-protein coupled receptor 120 antagonist, or the peroxisome proliferator-activated receptors antagonists, GW6471 and GW9662. In addition, KetoC and HYA did not inhibit translocation of p65, a subunit of NF-κB, or IκB degradation. Similarly, no effect on p38 or JNK phosphorylation was observed. However, KetoC and HYA were found to inhibit ERK phosphorylation induced by LPS, suggesting that these fatty acids may exert their anti-inflammatory effects through the inhibition of ERK activation in microglial cells.


Subject(s)
Anti-Inflammatory Agents , Fatty Acids, Unsaturated/biosynthesis , Fatty Acids, Unsaturated/pharmacology , Gastrointestinal Microbiome , Lactobacillus plantarum/metabolism , Microglia/metabolism , Oleic Acids/biosynthesis , Oleic Acids/pharmacology , Animals , Cells, Cultured , Depression, Chemical , Extracellular Signal-Regulated MAP Kinases/metabolism , Linoleic Acid/metabolism , Lipopolysaccharides/adverse effects , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects
14.
Eur J Pharmacol ; 818: 470-479, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29154837

ABSTRACT

The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway, which induces the production of antioxidant enzymes, is a possible therapeutic target for treating diseases related to oxidative stress. Nrf2 activators often exhibit cytotoxicity due to nonspecific electrophilic reactions with thiol groups. We screened a chemical library to explore Nrf2 activators with a wide safety margin. In at least in vitro experiments, TPNA10168, identified from the library, showed a higher efficacy in Nrf2 activation and a lower cytotoxicity than sulforaphane, a well-known Nrf2 activator. The present study demonstrated the protective effect of TPNA10168 against 6-hydroxydopamine-induced cytotoxicity. In PC12 cells, NAD(P)H:quinone oxidoreductase 1 was upregulated by TPNA10168 and participated in the protective effect. In primary mesencephalic cultures, heme oxygenase-1, upregulated by TPNA10168 in astrocytes, provided protection of dopaminergic neurons via a guanylate cyclase/protein kinase G signaling pathway via carbon monoxide. These results suggest that the compound identified from the chemical library may be suitable as a neuroprotective agent with the ability to induce antioxidant enzymes.


Subject(s)
Antioxidants/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Response Elements/drug effects , Small Molecule Libraries/pharmacology , Animals , Cell Death/drug effects , Dopaminergic Neurons/cytology , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/genetics , Oxidopamine/toxicity , PC12 Cells , Rats , Up-Regulation/drug effects
15.
J Alzheimers Dis ; 59(1): 313-328, 2017.
Article in English | MEDLINE | ID: mdl-28598836

ABSTRACT

Aggregation of amyloid-ß (Aß) and tau plays a crucial role in the onset and progression of Alzheimer's disease (AD). Therefore, the inhibition of Aß and tau aggregation may represent a potential therapeutic target for AD. Herein, we designed and synthesized both Aß and tau dual aggregation inhibitors based on the structure of curcumin and developed the novel curcumin derivative PE859. In this study, we investigated the inhibitory activity of PE859 on Aß aggregationin vitro and the therapeutic effects of PE859 on cognitive dysfunction via dual inhibition of Aß and tau aggregation in vivo. PE859 inhibited Aß aggregation in vitro and protected cultured cells from Aß-induced cytotoxicity. Furthermore, PE859 ameliorated cognitive dysfunction and reduced the amount of aggregated Aß and tau in brains of senescence-accelerated mouse prone 8 (SAMP8). These results warrant consideration of PE859 as a candidate drug for AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Indoles/therapeutic use , Protein Aggregates/drug effects , Pyrazoles/therapeutic use , tau Proteins/metabolism , Aging/genetics , Amyloid beta-Peptides/ultrastructure , Animals , Brain/drug effects , Brain/metabolism , Brain/ultrastructure , Cell Line, Tumor , Cognition Disorders/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , L-Lactate Dehydrogenase/metabolism , Maze Learning/drug effects , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Motor Activity/drug effects , Neuroblastoma/pathology , Quartz Crystal Microbalance Techniques , Time Factors , tau Proteins/ultrastructure
16.
J Pharmacol Sci ; 134(1): 37-44, 2017 May.
Article in English | MEDLINE | ID: mdl-28499726

ABSTRACT

Donepezil is a potent and selective acetylcholinesterase inhibitor developed for the treatment of Alzheimer's disease. In the present study, we investigated the responses of astrocytes to bradykinin, an inflammatory mediator, and the effect of donepezil on these responses using cultured cortical astrocytes. Bradykinin induced a transient increase of intracellular calcium concentration ([Ca2+]i) in cultured astrocytes. Bradykinin-induced [Ca2+]i increase was inhibited by the exposure to thapsigargin, which depletes Ca2+ stores on endoplasmic reticulum, but not by the exclusion of extracellular Ca2+. Twenty four hours pretreatment of donepezil reduced the bradykinin-induced [Ca2+]i increase. This reduction was inhibited not only by mecamylamine, a nAChR antagonist, but also by PI3K and Akt inhibitors. In addition, donepezil inhibited bradykinin-induced increase of the intracellular reactive oxygen species level in astrocytes. These results suggest that donepezil inhibits the inflammatory response induced by bradykinin via nAChR and PI3K-Akt pathway in astrocytes.


Subject(s)
Astrocytes/metabolism , Bradykinin/pharmacology , Calcium/metabolism , Acetylcholinesterase/metabolism , Animals , Astrocytes/drug effects , Calcium/chemistry , Cells, Cultured , Cerebral Cortex/cytology , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Donepezil , Indans/therapeutic use , Inflammation , Mecamylamine/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Piperidines/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Thapsigargin/pharmacology
17.
Sci Rep ; 7: 42111, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176845

ABSTRACT

During development, dopaminergic neurons born in the substantia nigra extend their axons toward the striatum. However, the mechanisms by which the dopaminergic axons extend the striatum to innervate their targets remain unclear. We previously showed that paired-cultivation of mesencephalic cells containing dopaminergic neurons with striatal cells leads to the extension of dopaminergic neurites from the mesencephalic cell region to the striatal cell region. The present study shows that dopaminergic neurites extended along striatal neurons in the paired-cultures of mesencephalic cells with striatal cells. The extension of dopaminergic neurites was suppressed by the pharmacological inhibition of integrin α5ß1. Using lentiviral vectors, short hairpin RNA (shRNA)-mediated knockdown of integrin α5 in dopaminergic neurons suppressed the neurite outgrowth to the striatal cell region. In contrast, the knockdown of integrin α5 in non-dopaminergic mesencephalic and striatal cells had no effect. Furthermore, overexpression of integrin α5 in dopaminergic neurons differentiated from embryonic stem cells enhanced their neurite outgrowth on striatal cells. These results indicate that integrin α5ß1 expression on dopaminergic neurons plays an important role in the dopaminergic neurite outgrowth on striatal neurons.


Subject(s)
Dopaminergic Neurons/chemistry , Dopaminergic Neurons/physiology , Integrin alpha5beta1/analysis , Neuronal Outgrowth , Substantia Nigra/cytology , Ventral Striatum/cytology , Animals , Cells, Cultured , Embryonic Stem Cells/physiology , Rats
18.
Eur J Pharmacol ; 798: 26-34, 2017 Mar 05.
Article in English | MEDLINE | ID: mdl-28167258

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the substantia nigra (SN), and oxidative stress is thought to contribute to the pathogenesis. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway, which is a cellular defense system against oxidative stress, is a promising target for therapeutics aimed at reducing neuronal death in PD. Previously, we have isolated 2',3'-dihydroxy-4',6'-dimethoxychalcone (DDC) from green perilla leaves as an activator of the Nrf2-ARE pathway. The present study showed the protective effect of DDC on PD models in vivo and in vitro. In a 6-hydroxydopamine (6-OHDA)-induced hemiparkinson's disease mouse model, intracerebral administration of DDC suppressed the dopaminergic neuronal loss and behavioral dysfunction. DDC upregulated the expression of heme oxygenase-1 (HO-1), one of the ARE-driven antioxidant enzymes, in astrocytes and microglia of the SN. In primary mesencephalic cultures, treatment with DDC also increased the HO-1 expression in astrocytes and microglia. DDC showed a protective effect against 6-OHDA-induced dopaminergic neuronal death, and the effect was suppressed by an HO-1 inhibitor. These results suggest that DDC prevents dopaminergic neurons from oxidative stress by upregulation of glial expression of HO-1.


Subject(s)
Antioxidants/metabolism , Dopaminergic Neurons/drug effects , NF-E2-Related Factor 2/metabolism , Parkinson Disease/pathology , Perilla/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Animals , Cell Death/drug effects , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Male , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , Response Elements/drug effects
19.
Article in English | MEDLINE | ID: mdl-27493787

ABSTRACT

BACKGROUND: Catheter ablation is a non-medication therapy for atrial fibrillation, and during the procedure, warfarin is withdrawn in the preoperative period to prevent the risk of bleeding. In case of emergency, vitamin K2 can be intravenously administered to antagonize the anticoagulant activity of warfarin. The aims of this study were to conduct population pharmacokinetic/pharmacodynamic modeling for retrospective clinical data and to investigate the effect of vitamin K2 on the anticoagulant activity of warfarin in the perioperative period of catheter ablation. METHODS: A total of 579 international normalized ratio (INR) values of prothrombin time from 100 patients were analyzed using the nonlinear mixed-effects modeling program NONMEM. A 1-compartment model was adapted to the pharmacokinetics of warfarin and vitamin K2, and the indirect response model was used to investigate the relationship between plasma concentration and the pharmacodynamic response of warfarin and vitamin K2. Since no plasma concentration data for warfarin and vitamin K2 were available, 3 literally available pharmacokinetic parameters were used to simultaneously estimate 1 pharmacokinetic parameter and 5 pharmacodynamic parameters. RESULTS: The population parameters obtained not only successfully explained the observed INR values, but also indicated an increase in sensitivity to warfarin in patients with reduced renal function. Simulations using these parameters indicated that vitamin K2 administration of more than 20 mg caused a slight dose-dependent decrease in INR on the day of catheter ablation and a delayed INR elevation after warfarin re-initiation. CONCLUSIONS: A pharmacokinetic/pharmacodynamic model was successfully built to explain the retrospective INR data during catheter ablation. Simulation studies suggest that vitamin K2 should be administered with care and that more than 20 mg is unnecessary in the preoperative period of catheter ablation.

20.
J Parkinsons Dis ; 6(3): 569-79, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27314756

ABSTRACT

BACKGROUND: The formation of intracellular aggregates containing α-synuclein (α-syn) is a main pathological feature of Parkinson disease. The propagation of α-syn aggregation via cell-to-cell transmission has been implicated in the progression of Parkinson disease. OBJECTIVE: Our aim is to clarify the molecular mechanisms underlying the formation of intracellular aggregation by extracellular α-syn. METHODS: We investigated the effects of exogenous α-syn aggregates on intracellular α-syn immunoreactivity in α-syn-overexpressing SH-SY5Y cells using two antibodies to distinct epitopes of α-syn. To obtain α-syn aggregates, α-syn solution was aged with continuous agitation. RESULTS: Immunoreactivity against the acidic C-terminal domain of the intracellular α-syn was reduced by exposure to agedα-syn, whereas that against the hydrophobic non-amyloid component region was not changed. The reduction in immunoreactivity was not suppressed by protease inhibitors but was mimicked by neutralization of the negative charges on the C-terminal of the intracellular α-syn induced by spermine or extracellular acidification. CONCLUSIONS: These results suggest that the reduction in immunoreactivity is attributed not to proteolytic cleavage but to a conformational change at the C-terminus of the intracellular α-syn. The conformational change at the C-terminus of the intracellular α-syn might be involved in an initial step of fibril formation by exogenous α-syn aggregates.


Subject(s)
alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Cell Line, Tumor , Humans , Immunohistochemistry , Protein Aggregates , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , alpha-Synuclein/immunology , alpha-Synuclein/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...