Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
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.
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
4.
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
5.
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
6.
Biol Pharm Bull ; 37(6): 1050-5, 2014.
Article in English | MEDLINE | ID: mdl-24882416

ABSTRACT

Dried Nardostachys chinensis roots contain sesquiterpenoids that are widely used as herbal tranquilizers. We previously identified the highly sedative sesquiterpenoid valerena-4,7(11)-diene (VLD) from this plant. In the present study, we investigated stress reducing effects of VLD and the associated mechanisms of action. Application of 15-min restraint stresses induced excitatory behaviors in mice. Immobility times in the forced swim test and sleeping times in the pentobarbital sleep test were shortened in the stressed group by 47% and 43%, respectively, compared with the control group. Furthermore, restraint stress increased serum corticosterone levels by 75%, and cerebral serotonin (5-HT) and dopamine (DA) levels. Inhaled VLD (300 µg/cage) suppressed stress-induced excitatory behaviors and significantly reduced stress-induced blood corticosterone, cerebral 5-HT, and DA levels. These results suggest that VLD interacts with the hypothalamic-pituitary-adrenal axis and the sympathetic-adrenomedullary system. These interactions appear to involve GABAergic and D2 antagonist activities. Moreover, tests in anosmic and intravenously treated mice showed that the sedative effect of inhaled VLD was expressed via olfactory stimulation and pulmonary absorption. Although more studies are required to further elucidate the properties of this compound, our studies suggest that VLD may be an effective anti-stress aromatherapy for humans.


Subject(s)
Behavior, Animal/drug effects , Hypnotics and Sedatives/therapeutic use , Nardostachys/chemistry , Sesquiterpenes/therapeutic use , Stress, Psychological/drug therapy , Administration, Inhalation , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corticosterone/blood , Dopamine/metabolism , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/isolation & purification , Hypnotics and Sedatives/pharmacokinetics , Hypothalamo-Hypophyseal System/drug effects , Male , Mice, Inbred Strains , Pituitary-Adrenal System/drug effects , Plant Roots/chemistry , Restraint, Physical , Serotonin/metabolism , Sesquiterpenes/administration & dosage , Sesquiterpenes/isolation & purification , Sesquiterpenes/pharmacokinetics , Stress, Psychological/metabolism
7.
Free Radic Biol Med ; 53(4): 669-79, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22749808

ABSTRACT

The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway is a cellular defense system against oxidative stress. Activation of this pathway increases expression of antioxidant enzymes. Epidemiological studies have demonstrated that the consumption of fruits and vegetables is associated with reduced risk of contracting a variety of human diseases. The aim of this study is to find Nrf2-ARE activators in dietary fruits and vegetables. We first attempted to compare the potency of ARE activation in six fruit and six vegetables extracts. Green perilla (Perilla frutescens var. crispa f. viridis) extract exhibited high ARE activity. We isolated the active fraction from green perilla extract through bioactivity-guided fractionation. Based on nuclear magnetic resonance and mass spectrometric analysis, the active ingredient responsible for the ARE activity was identified as 2',3'-dihydroxy-4',6'-dimethoxychalcone (DDC). DDC induced the expression of antioxidant enzymes, such as γ-glutamylcysteine synthetase (γ-GCS), NAD(P)H: quinone oxidoreductase-1 (NQO1), and heme oxygenase-1. DDC inhibited the formation of intracellular reactive oxygen species and the cytotoxicity induced by 6-hydroxydopamine. Inhibition of the p38 mitogen-activated protein kinase pathway abolished ARE activation, the induction of γ-GCS and NQO1, and the cytoprotective effect brought about by DDC. Thus, this study demonstrated that DDC contained in green perilla enhanced cellular resistance to oxidative damage through activation of the Nrf2-ARE pathway.


Subject(s)
Antioxidant Response Elements , Chalcones/isolation & purification , NF-E2-Related Factor 2/genetics , Perilla frutescens/chemistry , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Transcriptional Activation/drug effects , Animals , Cell Survival/drug effects , Chalcones/pharmacology , Cytoprotection , Enzyme Induction/drug effects , Fruit/chemistry , Genes, Reporter , Glutathione/metabolism , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Molecular Structure , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , PC12 Cells , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reactive Oxygen Species/metabolism , Transcription, Genetic , Vegetables/chemistry , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Neuropharmacology ; 61(5-6): 975-80, 2011.
Article in English | MEDLINE | ID: mdl-21752338

ABSTRACT

Thrombin activates immunocompetent microglia and increases release of inflammatory cytokines under intracerebral hemorrhage (ICH) insults. Also, thrombin injection into the striatum evokes acute necrosis and delayed apoptosis of neurons. A nucleoprotein high-mobility group box 1 (HMGB1) that is released from necrotic cells has been suggested to behave like a cytokine and cause over-facilitation of immune functions. Here we examined the effect of glycyrrhizin, known as an inhibitor of HMGB1, on thrombin-induced injury in rat cortico-striatal slice cultures and in vivo rat ICH model. In slice cultures, thrombin-induced a drastic increase in propidium iodide fluorescence indicating necrotic cell death in the cortical region, and robust shrinkage of the striatal tissue. Glycyrrhizin (10-100 µM) attenuated thrombin-induced cortical injury in a concentration-dependent manner. The protective effect of glycyrrhizin was not mediated by glucocorticoid receptors or modulation of nitric oxide production, but was reversed by exogenous HMGB1 application. The injury induced by a high concentration of HMGB1 was suppressed by glycyrrhizin. In vivo, unilateral injection of type IV collagenase into rat striatum induced ICH associated with brain edema formation, contralateral paralysis and neuron death. Once daily intraperitoneal administration of glycyrrhizin attenuated ICH-induced edema in both the cortex and the basal ganglia, and improved behavioral performance of rats in forelimb placing. Moreover, glycyrrhizin partially but significantly ameliorated ICH-induced neuron loss inside hematoma. These findings suggest that an HMGB1 inhibitor glycyrrhizin is a potential candidate for a remedy for ICH.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/pathology , Glycyrrhizic Acid/pharmacology , HMGB1 Protein/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/therapeutic use , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Brain/metabolism , Brain Edema/chemically induced , Brain Edema/drug therapy , Brain Edema/metabolism , Brain Edema/pathology , Brain Injuries/chemically induced , Brain Injuries/complications , Brain Injuries/pathology , Cattle , Cell Death/drug effects , Cell Death/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Hemorrhage/chemically induced , Collagenases/physiology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Drug Evaluation, Preclinical , Glycyrrhizic Acid/metabolism , Glycyrrhizic Acid/therapeutic use , HMGB1 Protein/physiology , Hemostatics/pharmacology , Male , Molecular Targeted Therapy , Neostriatum/drug effects , Neostriatum/metabolism , Neurons/drug effects , Neurons/metabolism , Nitric Oxide/analysis , Nitric Oxide/biosynthesis , Rats , Rats, Sprague-Dawley , Rats, Wistar , Thrombin/pharmacology , Tissue Culture Techniques
9.
J Pharmacol Sci ; 115(3): 320-8, 2011.
Article in English | MEDLINE | ID: mdl-21358121

ABSTRACT

Oxidative stress plays pivotal roles in aging, neurodegenerative disease, and pathological conditions such as ischemia. We investigated the effect of sulforaphane and 6-(methysulfinyl) hexyl isothiocyanate (6-HITC), a naturally occurring isothiocyanate, on oxidative stress-induced cytotoxicity using primary neuronal cultures of rat striatum. Pretreatment with sulforaphane and 6-HITC significantly protected against H(2)O(2)- and paraquat-induced cytotoxicity in a concentration-dependent manner. Sulforaphane and 6-HITC induced the translocation of nuclear factor E2-related factor 2 (Nrf2) into the nucleus and increased the expression of γ-glutamylcysteine synthetase (γ-GCS), a rate-limiting enzyme in glutathione synthesis, and the intracellular glutathione content. Treatment with reduced glutathione (GSH) and N-acetyl-L-cysteine, a substance for glutathione synthesis, significantly prevented the cytotoxicity induced by H(2)O(2) and paraquat. Moreover, exposure to L-buthionine-sulfoximine, an irreversible inhibitor of γ-GCS, suppressed the protective effects of sulforaphane and 6-HITC. In contrast, sulforaphane and 6-HITC increased heme oxygenase-1 (HO-1) expression in neurons. However, zinc-protophorphyrin IX, a competitive inhibitor of HO-1, did not influence the protective effects of sulforaphane and 6-HITC. These results suggest that sulforaphane and 6-HITC prevent oxidative stress-induced cytotoxicity in rat striatal cultures by raising the intracellular glutathione content via an increase in γ-GCS expression induced by the activation of the Nrf2-antioxidant response element pathway.


Subject(s)
Glutathione/biosynthesis , Isothiocyanates/pharmacology , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Thiocyanates/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Cell Survival/drug effects , Cells, Cultured , Corpus Striatum/cytology , Drug Evaluation, Preclinical , Glutamate-Cysteine Ligase/biosynthesis , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/genetics , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Response Elements , Signal Transduction , Sulfoxides
10.
Peptides ; 31(6): 1131-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20307612

ABSTRACT

A hypothalamic neuropeptide orexin (hypocretin) is a critical regulator of physiological processes including sleep/wakefulness and feeding. Using organotypic slice culture of rat hypothalamus, we found that exposure to elevated extracellular concentration of K(+) (+10-30 mM) for 24-72h led to a substantial decrease in the number of neurons immunoreactive for orexin and a co-existing neuropeptide dynorphin-A. In contrast, the same treatment affected neither the number of melanin-concentrating hormone-immunoreactive neurons nor the number of total neurons. A substantial decrease of orexin-immunoreactive neurons was also induced by 72h treatment with 1-10 microM veratridine, a Na(+) channel activator. The effect of elevated K(+) was only partially reversible, and that of veratridine was virtually irreversible, although the decrease in orexin immunoreactivity was not associated with signs of cell damage assessed by propidium iodide uptake and Hoechst 33342 nuclear staining. In addition, the level of preproorexin mRNA did not decrease during treatment with elevated K(+) or veratridine. After treatment with elevated K(+) and veratridine, c-Fos immunoreactivity appeared in orexin-immunoreactive neurons but not in melanin-concentrating hormone-immunoreactive neurons, suggesting selective excitation of orexin neurons. However, the amount of orexin released extracellularly was paradoxically decreased by treatment with elevated K(+) and veratridine. Overall, these characteristics of orexin neurons may be taken into consideration to understand the behaviors of these neurons under physiological and pathophysiological conditions.


Subject(s)
Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Potassium/administration & dosage , Animals , Dynorphins/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/drug effects , Melanins/metabolism , Neuromuscular Depolarizing Agents/pharmacology , Neurons/drug effects , Neurons/metabolism , Orexins , Pituitary Hormones/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Veratridine/pharmacology
11.
Bioorg Med Chem ; 15(22): 7098-107, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17804246

ABSTRACT

We present efficient syntheses of serofendic acids A and B (SA-A and SA-B), novel neuroprotective substances isolated from fetal calf serum. Biological and pharmacological evaluation showed that SA-A and SA-B have potent protective action against glutamate-induced neurotoxicity, but do not interact directly with glutamate receptors. A pharmacokinetic study showed that they have good oral bioavailability in rats. The results indicate that SA-A and SA-B are potential lead compounds for candidate drugs to treat various neurological disorders.


Subject(s)
Diterpenes/chemical synthesis , Diterpenes/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Administration, Oral , Animals , Cattle , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Crystallography, X-Ray , Diterpenes/administration & dosage , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Fetal Blood/chemistry , Glutamic Acid/toxicity , Injections, Intravenous , Models, Molecular , Molecular Conformation , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution
12.
Neuroreport ; 18(8): 813-6, 2007 May 28.
Article in English | MEDLINE | ID: mdl-17471072

ABSTRACT

Mulberry leaf has been reported to possess medicinal properties, including hypoglycemic, hypotensive and diuretic effects. Little is known, however, about its medicinal properties for central nervous system disorders, including Alzheimer's disease. Accumulating evidence suggests that amyloid beta-peptide (1-42) plays an important role in the etiology of Alzheimer's disease. Here we show that mulberry leaf extract inhibits the amyloid beta-peptide (1-42) fibril formation by both the thioflavin T fluorescence assay and atomic force microscopy. Furthermore, mulberry leaf extract protected hippocampal neurons against amyloid beta-peptide (1-42)-induced cell death in a concentration-dependent manner. These results suggest that mulberry leaf extract provides a viable treatment for Alzheimer's disease through the inhibition of amyloid beta-peptide (1-42) fibril formation and attenuation of amyloid beta-peptide (1-42)-induced neurotoxicity.


Subject(s)
Amyloid beta-Peptides/toxicity , Morus/chemistry , Neurofibrillary Tangles/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Peptide Fragments/toxicity , Plant Extracts/pharmacology , Animals , Benzothiazoles , Cell Count/methods , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Hippocampus/cytology , Mice , Mice, Inbred ICR , Microscopy, Atomic Force/methods , Neurons/drug effects , Thiazoles/metabolism
13.
J Neurosci ; 27(15): 4210-9, 2007 Apr 11.
Article in English | MEDLINE | ID: mdl-17428999

ABSTRACT

Regeneration in the mammalian CNS is severely limited. Unlike in the chick, current models hold that retinal neurons are never regenerated. Previously we demonstrated that, in the adult mammalian retina, Müller glia dedifferentiate and produce retinal cells, including photoreceptors, after acute neurotoxic injury in vivo. However, the number of newly generated retinal neurons is very limited. Here we demonstrate that Wnt (wingless-type MMTV integration site family)/beta-catenin signaling promotes proliferation of Müller glia-derived retinal progenitors and neural regeneration after damage or during degeneration. Wnt3a treatment increases proliferation of dedifferentiated Müller glia >20-fold in the photoreceptor-damaged retina. Supplementation with retinoic acid or valproic acid induces differentiation of these cells primarily into Crx (cone rod homeobox)-positive and rhodopsin-positive photoreceptors. Notably, injury induces nuclear accumulation of beta-catenin, cyclin D1 upregulation, and Wnt/beta-catenin reporter activity. Activation of Wnt signaling by glycogen synthase kinase-3beta inhibitors promotes retinal regeneration, and, conversely, inhibition of the signaling attenuates regeneration. This Wnt3a-mediated regeneration of retinal cells also occurs in rd mice, a model of retinal degeneration. These results provide evidence that Wnt/beta-catenin signaling contributes to CNS regeneration in the adult mammal.


Subject(s)
Nerve Regeneration/physiology , Retina/cytology , Retina/growth & development , Signal Transduction/physiology , Wnt Proteins/physiology , Animals , Macaca fascicularis , Male , Mice , Mice, Transgenic , Organ Culture Techniques , Rats , Wnt3 Protein , Wnt3A Protein
14.
Alzheimer Dis Assoc Disord ; 20(2 Suppl 1): S8-11, 2006.
Article in English | MEDLINE | ID: mdl-16772755

ABSTRACT

The protection of neurons from damage and death in neurodegenerative disorders, such as Alzheimer disease (AD), is a major challenge for neuroscientists in the 21st century. The amyloid beta-protein plays an important role in the degenerative process of the disease and increases the vulnerability of cultured cortical neurons to glutamate neurotoxicity. Glutamate may, therefore, play an important role in amyloid beta-protein-induced cytotoxicity in the cerebral cortex. Results show that cholinesterase inhibitors such as donepezil protect cortical neurons against glutamate neurotoxicity via alpha4beta2 and alpha7 nicotinic acetylcholine receptors at least partly by inhibiting the process of apoptosis. Donepezil also protects against ischemic insults such as those seen in vascular dementia; however, this does not seem to be mediated by nicotinic receptors. This review summarizes data that suggest donepezil possesses neuroprotective actions in addition to amelioration of cognitive deficits by inhibition of acetylcholinesterase.


Subject(s)
Alzheimer Disease/pathology , Cerebral Cortex/drug effects , Cholinesterase Inhibitors/pharmacology , Drug Evaluation, Preclinical , Neuroprotective Agents/pharmacology , Alzheimer Disease/physiopathology , Animals , Apoptosis/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Dementia, Vascular/pathology , Dementia, Vascular/physiopathology , Donepezil , Glutamic Acid/physiology , Humans , Indans/pharmacology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Piperidines/pharmacology , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/physiology , alpha7 Nicotinic Acetylcholine Receptor
15.
Neuroreport ; 16(11): 1157-61, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16012340

ABSTRACT

Selective degeneration of hypothalamic orexin neurons, a hallmark of pathology in narcolepsy patients, is in part reproduced in hypothalamic slice cultures by application of an endogenous excitotoxin quinolinic acid. Depolarized membrane potential may be responsible for the vulnerability of orexin neurons to excitotoxicity. We show that stimulation of gamma-aminobutyric acid type A receptors, which is known to hyperpolarize orexin neurons, by muscimol or isoguvacine potently inhibits quinolinic acid cytotoxicity on orexin neurons. In addition, the protective effect of gamma-aminobutyric acid and a gamma-aminobutyric acid uptake blocker nipecotic acid is abolished by a gamma-aminobutyric acid type A antagonist picrotoxin. Norepinephrine and serotonin do not provide a neuroprotective effect. Thus, GABAergic inhibitory control may be a decisive factor regulating survival of orexin neurons under excitotoxic insults.


Subject(s)
GABA Agonists/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/drug effects , Neuropeptides/metabolism , Quinolinic Acid/toxicity , Receptors, GABA-A/physiology , Animals , Animals, Newborn , Bicuculline/pharmacology , Cell Count/methods , Citalopram/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , GABA Antagonists/pharmacology , Glutamate Decarboxylase/metabolism , Hypothalamus/cytology , Immunohistochemistry/methods , In Vitro Techniques , Magnesium/metabolism , Neurons/metabolism , Nipecotic Acids/pharmacology , Norepinephrine/antagonists & inhibitors , Norepinephrine/pharmacology , Orexin Receptors , Orexins , Organophosphorus Compounds/pharmacology , Picrotoxin/pharmacology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Serotonin/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , gamma-Aminobutyric Acid/pharmacology
16.
Neurobiol Dis ; 15(1): 61-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14751771

ABSTRACT

Several lines of evidence indicate that narcolepsy, a sleep disorder, results from the loss of hypothalamic orexin (hypocretin)-containing neurons, but the mechanisms responsible for selective elimination of this neuronal population are unknown. Using organotypic rat hypothalamic slice cultures, we investigated vulnerability of orexin neurons to excitotoxic insults. Twenty-four hours of incubation with N-methyl-D-aspartate (NMDA) followed by a recovery period of 72 h resulted in a marked decrease in the number of orexin-immunoreactive neurons, whereas melanin-concentrating hormone (MCH)-immunoreactive neurons in the same cultures were relatively spared. In contrast, orexin neurons were more resistant to kainic acid cytotoxicity than MCH neurons. Examinations of the effects of several endogenous glutamate receptor agonists as well as a glutamate transporter blocker highlighted quinolinic acid as an endogenous excitotoxin that could cause selective loss of orexin neurons as compared to MCH neurons by activating NMDA receptors. In addition, quinolinic acid-induced decrease of orexin neurons was prevented by an inhibitor of poly(ADP-ribose) polymerases. These results provide the first evidence concerning cytotoxic consequences onto orexin neurons, and indicate that NMDA receptor-mediated injury may contribute to the selective loss of these neurons in the hypothalamus, a prominent neuropathological feature found in narcolepsy patients.


Subject(s)
Carrier Proteins/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins , Narcolepsy/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Neurotoxins/toxicity , Animals , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/toxicity , Hypothalamic Hormones/metabolism , Hypothalamus/pathology , Hypothalamus/physiopathology , In Vitro Techniques , Melanins/metabolism , N-Methylaspartate/pharmacology , N-Methylaspartate/toxicity , Narcolepsy/physiopathology , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/pathology , Neurotoxins/metabolism , Orexin Receptors , Orexins , Pituitary Hormones/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Quinolinic Acid/metabolism , Quinolinic Acid/toxicity , Rats , Rats, Wistar , Receptors, G-Protein-Coupled , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Neuropeptide
17.
Neuropharmacology ; 42(8): 1056-64, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12128007

ABSTRACT

The prevalence of Parkinson's disease is higher in males than in females. Although the reason for this gender difference is not clear, the level of female steroid hormones or their receptors may be involved in the pathogenesis. The estrogen receptor subtype expressed in the midbrain is limited to the novel beta subtype, whose role in the central nervous system has not been resolved. We demonstrated that ligand-activated estrogen receptor beta suppressed dopaminergic neuronal death in an in vitro Parkinson's disease model which uses 1-methyl-4-phenylpyridinium ions (MPP(+)). MPP(+) treatment caused the upregulation of c-Jun amino-terminal kinase (JNK) and dopaminergic neuronal death, the latter being blocked by curcumin, an inhibitor of the c-Jun/AP-1 cascade. 17alpha- and 17beta-estradiol both protected dopaminergic neurons from MPP(+)-induced neuronal death and this was blocked by a pure antagonist of the estrogen receptor, ICI 182,780, but not by an inhibitor of estrogen receptor dimerization, YP537. These data indicated that the neuroprotection provided by 17alpha-estradiol was via inhibitory transcriptional regulation at the activator protein-1 (AP-1) site mediated by estrogen receptor beta. Thus, 17alpha-estradiol is a suitable candidate for neuroprotective therapy of Parkinson's disease because it is associated with few undesirable feminizing effects.


Subject(s)
1-Methyl-4-phenylpyridinium/adverse effects , Apoptosis/drug effects , Dopamine/metabolism , Estradiol/pharmacology , Estradiol/therapeutic use , Neurons/drug effects , Parkinsonian Disorders/prevention & control , Animals , Apoptosis/physiology , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Embryo, Mammalian , Estrogen Receptor beta , Female , JNK Mitogen-Activated Protein Kinases , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/biosynthesis , Neurons/cytology , Neurons/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Pregnancy , Rats , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL