Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neurotox Res ; 39(5): 1511-1523, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34417986

ABSTRACT

High mobility group box-1 (HMGB1) is a ubiquitous non-histone nuclear protein that plays a key role as a transcriptional activator, with its extracellular release provoking inflammation. Inflammatory responses are essential in methamphetamine (METH)-induced acute dopaminergic neurotoxicity. In the present study, we examined the effects of neutralizing anti-HMGB1 monoclonal antibody (mAb) on METH-induced dopaminergic neurotoxicity in mice. BALB/c mice received a single intravenous administration of anti-HMGB1 mAb prior to intraperitoneal injections of METH (4 mg/kg × 2, at 2-h intervals). METH injections induced hyperthermia, an increase in plasma HMGB1 concentration, degeneration of dopaminergic nerve terminals, accumulation of microglia, and extracellular release of neuronal HMGB1 in the striatum. These METH-induced changes were significantly inhibited by intravenous administration of anti-HMGB1 mAb. In contrast, blood-brain barrier disruption occurred by METH injections was not suppressed. Our findings demonstrated the neuroprotective effects of anti-HMGB1 mAb against METH-induced dopaminergic neurotoxicity, suggesting that HMGB1 could play an initially important role in METH toxicity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Dopamine Uptake Inhibitors/toxicity , Dopaminergic Neurons/drug effects , HMGB1 Protein/antagonists & inhibitors , Methamphetamine/toxicity , Neuroprotective Agents/pharmacology , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , HMGB1 Protein/blood , Male , Mice , Mice, Inbred BALB C
2.
Sci Rep ; 10(1): 20698, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33244123

ABSTRACT

Mirtazapine, a noradrenergic and specific serotonergic antidepressant (NaSSA), is known to activate serotonin (5-HT) 1A receptor. Our recent study demonstrated that stimulation of astrocytic 5-HT1A receptors promoted astrocyte proliferation and upregulated antioxidative property in astrocytes to protect dopaminergic neurons against oxidative stress. Here, we evaluated the neuroprotective effects of mirtazapine against dopaminergic neurodegeneration in models of Parkinson's disease (PD). Mirtazapine administration attenuated the loss of dopaminergic neurons in the substantia nigra and increased the expression of the antioxidative molecule metallothionein (MT) in the striatal astrocytes of 6-hydroxydopamine (6-OHDA)-injected parkinsonian mice via 5-HT1A receptors. Mirtazapine protected dopaminergic neurons against 6-OHDA-induced neurotoxicity in mesencephalic neuron and striatal astrocyte cocultures, but not in enriched neuronal cultures. Mirtazapine-treated neuron-conditioned medium (Mir-NCM) induced astrocyte proliferation and upregulated MT expression via 5-HT1A receptors on astrocytes. Furthermore, treatment with medium from Mir-NCM-treated astrocytes protected dopaminergic neurons against 6-OHDA neurotoxicity, and these effects were attenuated by treatment with a MT-1/2-specific antibody or 5-HT1A antagonist. Our study suggests that mirtazapine could be an effective disease-modifying drug for PD and highlights that astrocytic 5-HT1A receptors may be a novel target for the treatment of PD.


Subject(s)
Astrocytes/drug effects , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Mirtazapine/pharmacology , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Animals , Antioxidants/pharmacology , Astrocytes/metabolism , Cells, Cultured , Dopaminergic Neurons/metabolism , Female , Male , Metallothionein/metabolism , Mice , Mice, Inbred ICR , Oxidative Stress/drug effects , Oxidopamine/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Pregnancy , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
3.
Nat Prod Res ; 34(12): 1750-1755, 2020 Jun.
Article in English | MEDLINE | ID: mdl-30513209

ABSTRACT

An aliphatic ester of hydroxysalicylic acid (6), reported for the first time from a natural source in addition to five known compounds were isolated from the fermented Carica papaya L. preparation, a commercialized functional food. The known compounds were identified as 5-hydroxymethylfurfuraldehyde (1), trans-caffeic acid (2), butyl 4-hydroxybenzoate (butylparaben) (3), lycopene (4), benzyl isothiocyanate (5). Compounds 1 and 3 were reported for the first time from Papaya fruits through this study. The new compound showed a moderate antioxidant activity and a potent hair growth stimulating activity in vitro.


Subject(s)
Antioxidants/isolation & purification , Carica/chemistry , Hair/growth & development , Plant Preparations/chemistry , Salicylic Acid/chemistry , Antioxidants/pharmacology , Esters , Fruit/chemistry , Hair/drug effects , Plant Extracts/chemistry , Plant Extracts/pharmacology , Salicylic Acid/isolation & purification , Salicylic Acid/pharmacology
4.
Int J Mol Sci ; 20(3)2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30704073

ABSTRACT

In previous studies, we found regional differences in the induction of antioxidative molecules in astrocytes against oxidative stress, postulating that region-specific features of astrocytes lead region-specific vulnerability of neurons. We examined region-specific astrocytic features against dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA) as an oxidative stress using co-culture of mesencephalic neurons and mesencephalic or striatal astrocytes in the present study. The 6-OHDA-induced reduction of mesencephalic dopamine neurons was inhibited by co-culturing with astrocytes. The co-culture of midbrain neurons with striatal astrocytes was more resistant to 6-OHDA than that with mesencephalic astrocytes. Furthermore, glia conditioned medium from 6-OHDA-treated striatal astrocytes showed a greater protective effect on the 6-OHDA-induced neurotoxicity and oxidative stress than that from mesencephalic astrocytes. The cDNA microarray analysis showed that the number of altered genes in both mesencephalic and striatal astrocytes was fewer than that changed in either astrocyte. The 6-OHDA treatment, apparently up-regulated expressions of Nrf2 and some anti-oxidative or Nrf2-regulating phase II, III detoxifying molecules related to glutathione synthesis and export in the striatal astrocytes but not mesencephalic astrocytes. There is a profound regional difference of gene expression in astrocytes induced by 6-OHDA. These results suggest that protective features of astrocytes against oxidative stress are more prominent in striatal astrocytes, possibly by secreting humoral factors in striatal astrocytes.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Oxidopamine/pharmacology , Animals , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Mesencephalon/cytology , Mesencephalon/drug effects , Neuroprotection/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
5.
Nutr Neurosci ; 21(3): 176-184, 2018 Apr.
Article in English | MEDLINE | ID: mdl-27841081

ABSTRACT

OBJECTIVES: Nuclear factor erythroid 2-related factor (Nrf2) in astrocyte plays important roles in brain homeostasis. Fermented papaya preparation (FPP) has anti-oxidative, anti-inflammatory, immunoregulatory properties. The present study investigated the effects of FPP on activation of Nrf2 and release of Nrf2-regulated neuroprotective antioxidants and detoxifying molecules. METHODS: Primary cultured astrocytes from rat embryos were treated with FPP for 6 or 24 hours. The expression levels of nuclear Nrf2 and cytoplasmic Nrf2-regulated molecules were determined by western blot analysis and immunohistochemistry. Glutathione levels were measured in cells and medium. Dopaminergic neurons were exposed 6-hydroxydopamine (6-OHDA) with/without pre-treatment with FPP astrocytes. Mice were treated orally with FPP for 2 weeks. RESULTS: FPP increased nuclear translocation of Nrf2 in striatal astrocytes, induced up-regulation of NAD(P)H quinine oxidoreductase-1, glutathione-S transferase and hemeoxygenase-1, and increased glutathione level and the percentage of metallothionein-expressing astrocytes. Moreover, FPP suppressed 6-OHDA-induced dopaminergic neuronal loss in not only neuron-astrocyte mixed culture, but also neuron-rich cultures pre-treated with glial conditioned medium. Two-week oral treatment of mice with FPP resulted in Nrf2 activation and increase in glutathione level in striatum. DISCUSSION: The results indicated that FPP enhances the anti-oxidative capacity through activation of Nrf2 in astrocytes, suggesting it may provide neuroprotection in oxidative stress-related neurodegenerative diseases.


Subject(s)
Astrocytes/metabolism , Carica/chemistry , Dietary Supplements , Fruit/chemistry , NF-E2-Related Factor 2/agonists , Neuroprotective Agents/metabolism , Signal Transduction , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Astrocytes/cytology , Carica/growth & development , Cells, Cultured , Dietary Sugars/administration & dosage , Dietary Sugars/metabolism , Dietary Supplements/microbiology , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Fermentation , Fetus/cytology , Fruit/growth & development , Glucose/administration & dosage , Glucose/metabolism , Male , Mesencephalon/cytology , Mesencephalon/metabolism , Mice, Inbred ICR , NF-E2-Related Factor 2/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/administration & dosage , Oxidative Stress , Rats, Sprague-Dawley , Visual Cortex/cytology , Visual Cortex/metabolism
6.
J Clin Biochem Nutr ; 59(2): 93-99, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27698535

ABSTRACT

l-Theanine (γ-glutamylethylamide), a component of green tea, is considered to have regulatory and neuroprotective roles in the brain. The present study was designed to determine the effect of l-theanine on excess dopamine-induced neurotoxicity in both cell culture and animal experiments. The primary cultured mesencephalic neurons or co-cultures of mesencephalic neurons and striatal astrocytes were pretreated with l-theanine for 72 h, and then treated with excess dopamine for further 24 h. The cell viability of dopamine neurons and levels of glutathione were evaluated. Excess dopamine-induced neurotoxicity was significantly attenuated by 72 h preincubation with l-theanine in neuron-astrocyte co-cultures but not in neuron-rich cultures. Exposure to l-theanine increased the levels of glutathione in both astrocytes and glial conditioned medium. The glial conditioned medium from l-theanine-pretreated striatal astrocytes attenuated dopamine-induced neurotoxicity and quinoprotein formation in mesencephalic neurons. In addition, replacement of l-glutamate with l-theanine in an in vitro cell-free glutathione-synthesis system produced glutathione-like thiol compounds. Furthermore, l-theanine administration (4 mg/kg, p.o.) for 14 days significantly increased glutathione levels in the striatum of mice. The results suggest that l-theanine provides neuroprotection against oxidative stress-induced neuronal damage by humoral molecules released from astrocytes, probably including glutathione.

7.
Asian Pac J Cancer Prev ; 17(7): 3077-84, 2016.
Article in English | MEDLINE | ID: mdl-27509932

ABSTRACT

Various beneficial effects have been described for fermented papaya preparation (FPP: SAIDOPS501) based on its antioxidative and antiinflammatory functions. The present study was designed to determine the effects of FPP on carcinogenesis in vivo, and immunomodulatory function in vitro. Mice were injected with RL male 1 cells subcutaneously or 3methylcholantherene (MCA) intravenously to induce cancer and orally or intraperitoneally treated with FPP solution. Human peripheral blood mononuclear cells (PBMC) were obtained from healthy volunteers and patients with atopic dermatitis, treated with FPP, and subjected to measurement of cytokine production and changes in Foxp3expressing regulatory T cell (Treg) stimulated with phytohemagglutinin (PHA). Administration of FPP suppressed tumor size and the incidence of malignancy. In vitro, treatment of PBMC with FPP induced IL1?, TNFα and IFNγ production. Moreover, FPP suppressed proliferation of PHAstimulated Foxp3expressing Treg. These results suggest that FPP has chemotherapeutic properties.


Subject(s)
Antineoplastic Agents/pharmacology , Carica/chemistry , Immunologic Factors/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Animals , Female , Fermentation , Forkhead Transcription Factors/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
J Neurochem ; 136(1): 194-204, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26485447

ABSTRACT

Astrocytes but not neurons express cystine/glutamate exchange transporter (xCT), which takes up cystine, and consequently supplies the substrate for GSH synthesis in neurons. It is recognized that GSH synthesis in neurons is dependent on the expression of xCT in astrocytes. Previous studies reported that levetiracetam (LEV), an anti-epileptic drug, increased xCT expression in vivo. The purpose of this study was to examine neuroprotective effects of LEV in parkinsonian models and demonstrate xCT in astrocytes as a target of neuroprotection against dopaminergic neurodegeneration. We identified striatal astrocytes cultured with LEV showed significant increase in xCT expression and GSH levels. Preincubation of primary cultured mesencephalic dopamine neurons with conditioned media from LEV-treated astrocytes protected against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity. These protective effects were canceled by xCT inhibitor. Furthermore, reduction of nigrostriatal dopaminergic neurons in 6-OHDA-lesioned parkinsonian mice was significantly abrogated by repeated injections of LEV. Treatment with LEV significantly increased the expression of xCT in striatal astrocytes in the hemi-parkinsonian mice. In conclusion, LEV exerts neuroprotective effects against neurodegeneration via up-regulation of xCT and GSH in astrocytes. Thus, xCT in astrocytes could be a potential target in novel neuroprotective approaches to prevent degeneration of dopaminergic neurons. Glutathione (GSH) is the most potent intrinsic antioxidant. Since extracellular cysteine is readily oxidized to form cystine, cystine transport mechanisms are essential to provide cells with cysteine. Cystine uptake is mediated by cystine/glutamate exchange transporter (xCT), expressed primarily on astrocytes, but not on neurons. Astrocytes take up cystine via xCT and reduce it to cysteine to supply cysteine, the substrate for GSH synthesis in neurons. This study demonstrated that levetiracetam (LEV), an anti-epileptic drug, increased GSH in/from astrocytes via xCT up-regulation. GSH derived from astrocytes protects dopamine neurons against neurotoxicity induced by dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA). Thus, xCT in astrocytes could be a potential target in novel neuroprotective approaches to prevent degeneration of dopaminergic neurons.


Subject(s)
Amino Acid Transport System y+/biosynthesis , Astrocytes/metabolism , Drug Delivery Systems , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/metabolism , Piracetam/analogs & derivatives , Amino Acid Transport System y+/agonists , Animals , Astrocytes/drug effects , Cells, Cultured , Drug Delivery Systems/methods , Female , Levetiracetam , Male , Mice , Mice, Inbred ICR , Parkinsonian Disorders/prevention & control , Piracetam/administration & dosage , Pregnancy , Rats , Rats, Sprague-Dawley
9.
Neurochem Res ; 40(6): 1165-78, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25894684

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disease with motor and non-motor symptoms that precede the onset of motor symptoms. Rotenone is often used to induce PD-like pathology in the central nervous system (CNS) and enteric nervous system (ENS). However, there is little or no information on the temporal changes in other neural tissues and the spread of pathology throughout the entire body organs. Here, we recorded the serial immunohistochemical changes in neurons and glial cells of the striatum, substantia nigra (SN), olfactory bulb (OB), thoracic cord (ThC) and ascending colon (AC) induced by 1-, 3- and 6-week administration of rotenone (50 mg/kg/day) infused subcutaneously in C57BL mice using an osmotic pump. Rotenone exposure for 3 or 6 weeks caused neurodegeneration in the striatum, whereas neuronal damage was seen in the SN and OB only after 6 weeks. Moreover, rotenone induced neurodegeneration in the myenteric plexus of AC but not in ThC. Rotenone also activated glial cells before any apparent neurodegeneration in the CNS but not in the ENS. Our results demonstrated that subcutaneous administration of rotenone can cause progressive neurodegeneration in the OB and AC, in addition to the nigrostriatal pathway, and temporal differential glial activation, and that these changes do not spread retrogradely from OB or ENS to nigrostriatal pathway. The results suggested that the different vulnerability of neurons to the neurotoxic effects of rotenone administrated subcutaneously are due to glial activation in these neural tissues.


Subject(s)
Central Nervous System/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Peripheral Nervous System/pathology , Rotenone/toxicity , Uncoupling Agents/toxicity , Animals , Brain/pathology , Colon, Ascending/pathology , Dopaminergic Neurons/pathology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Motor Neurons/pathology , Neural Pathways/pathology , Neurons/pathology
10.
Acta Med Okayama ; 68(6): 317-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25519025

ABSTRACT

In vertebrates, almost all somatic cells extend a single immotile cilium, referred to as a primary cilium. Increasing evidence suggests that primary cilia serve as cellular antennae in many types of tissues by sensing chemical or mechanical stimuli in the milieu surrounding the cells. In rodents an antibody to adenylyl cyclase 3 (AC3) has been widely used to label the primary cilia of neurons in vivo by immunostaining, whereas the lack of markers for the primary cilia of astrocytes has made it difficult to observe astrocytic primary cilia in vivo. Here, we obtained a visualization of astrocytic primary cilia in the mouse brain. In the somatosensory cortex, a large portion of neurons and astrocytes at postnatal day 10 (P10), and of neurons at P56 had AC3-positive primary cilia, whereas only approx. one-half of the astrocytes in the P56 mice carried primary cilia weakly positive for AC3. In contrast, the majority of astrocytes had ADP-ribosylation factor-like protein 13B (Arl13b)-positive primary cilia in the somatosensory cortex and other brain regions of P56 mice. The lengths of astrocytic primary cilia positive for Arl13b varied among the brain regions. Our data indicate that Arl13b is a noteworthy marker of astrocytic primary cilia in the brain.


Subject(s)
ADP-Ribosylation Factors/metabolism , Astrocytes/metabolism , Brain/metabolism , Cilia/metabolism , Animals , Astrocytes/cytology , Biomarkers/metabolism , Brain/cytology , Cilia/ultrastructure , Female , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Models, Animal , Neurons/cytology , Neurons/metabolism , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism
11.
PLoS One ; 9(9): e106362, 2014.
Article in English | MEDLINE | ID: mdl-25188235

ABSTRACT

L-DOPA is therapeutically efficacious in patients with Parkinson's disease (PD), although dopamine (DA) neurons are severely degenerated. Since cortical astrocytes express neutral amino acid transporter (LAT) and DA transporter (DAT), the uptake and metabolism of L-DOPA and DA in striatal astrocytes may influence their availability in the dopaminergic system of PD. To assess possible L-DOPA- and DA-uptake and metabolic properties of striatal astrocytes, we examined the expression of L-DOPA, DA and DAT in striatal astrocytes of hemi-parkinsonian model rats after repeated L-DOPA administration, and measured the contents of L-DOPA, DA and their metabolite in primary cultured striatal astrocytes after L-DOPA/DA treatment. Repeated injections of L-DOPA induced apparent L-DOPA- and DA-immunoreactivities and marked expression of DAT in reactive astrocytes on the lesioned side of the striatum in hemi-parkinsonian rats. Exposure to DA for 4h significantly increased the levels of DA and its metabolite DOPAC in cultured striatal astrocytes. L-DOPA was also markedly increased in cultured striatal astrocytes after 4-h L-DOPA exposure, but DA was not detected 4 or 8h after L-DOPA treatment, despite the expression of aromatic amino acid decarboxylase in astrocytes. Furthermore, the intracellular level of L-DOPA in cultured striatal astrocytes decreased rapidly after removal of extracellular L-DOPA. The results suggest that DA uptaken into striatal astrocytes is rapidly metabolized and that striatal astrocytes act as a reservoir of L-DOPA that govern the uptake or release of L-DOPA depending on extracellular L-DOPA concentration, but are less capable of converting L-DOPA to DA.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Levodopa/metabolism , Animals , Blotting, Western , Cells, Cultured , Corpus Striatum/cytology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Immunohistochemistry , Male , Mesencephalon/cytology , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley
12.
Neurotox Res ; 26(3): 285-98, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24923464

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease with motor symptoms as well as non-motor symptoms that precede the onset of motor symptoms. Mitochondrial complex I inhibitor, rotenone, has been widely used to reproduce PD pathology in the central nervous system (CNS) and enteric nervous system (ENS). We reported previously that metallothioneins (MTs) released from astrocytes can protect dopaminergic neurons against oxidative stress. The present study examined the changes in MT expression by chronic systemic rotenone administration in the striatum and colonic myenteric plexus of C57BL mice. In addition, we investigated the effects of MT depletion on rotenone-induced neurodegeneration in CNS and ENS using MT-1 and MT-2 knockout (MT KO) mice, or using primary cultured neurons from MT KO mice. In normal C57BL mice, subcutaneous administration of rotenone for 6 weeks caused neurodegeneration, increased MT expression with astrocytes activation in the striatum and myenteric plexus. MT KO mice showed more severe myenteric neuronal damage by rotenone administration after 4 weeks than wild-type mice, accompanied by reduced astroglial activation. In primary cultured mesencephalic neurons from MT KO mice, rotenone exposure induced neurotoxicity in dopaminergic neurons, which was complemented by addition of recombinant protein. The present results suggest that MT seems to provide protection against neurodegeneration in ENS of rotenone-induced PD model mice.


Subject(s)
Corpus Striatum/drug effects , Metallothionein/metabolism , Myenteric Plexus/drug effects , Neuroprotective Agents/metabolism , Parkinsonian Disorders/metabolism , Rotenone/administration & dosage , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Cells, Cultured , Corpus Striatum/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Male , Metallothionein/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Myenteric Plexus/metabolism , Neurons/drug effects , Neurons/metabolism , Parkinsonian Disorders/chemically induced
13.
PLoS One ; 9(5): e97918, 2014.
Article in English | MEDLINE | ID: mdl-24830745

ABSTRACT

In the rodent brain, certain G protein-coupled receptors and adenylyl cyclase type 3 are known to localize to the neuronal primary cilium, a primitive sensory organelle protruding singly from almost all neurons. A recent chemical screening study demonstrated that many compounds targeting dopamine receptors regulate the assembly of Chlamydomonas reinhardtii flagella, structures which are analogous to vertebrate cilia. Here we investigated the effects of dopaminergic inputs loss on the architecture of neuronal primary cilia in the rodent striatum, a brain region that receives major dopaminergic projections from the midbrain. We first analyzed the lengths of neuronal cilia in the dorsolateral striatum of hemi-parkinsonian rats with unilateral lesions of the nigrostriatal dopamine pathway. In these rats, the striatal neuronal cilia were significantly longer on the lesioned side than on the non-lesioned side. In mice, the repeated injection of reserpine, a dopamine-depleting agent, elongated neuronal cilia in the striatum. The combined administration of agonists for dopamine receptor type 2 (D2) with reserpine attenuated the elongation of striatal neuronal cilia. Repeated treatment with an antagonist of D2, but not of dopamine receptor type 1 (D1), elongated the striatal neuronal cilia. In addition, D2-null mice displayed longer neuronal cilia in the striatum compared to wild-type controls. Reserpine treatment elongated the striatal neuronal cilia in D1-null mice but not in D2-null mice. Repeated treatment with a D2 agonist suppressed the elongation of striatal neuronal cilia on the lesioned side of hemi-parkinsonian rats. These results suggest that the elongation of striatal neuronal cilia following the lack of dopaminergic inputs is attributable to the absence of dopaminergic transmission via D2 receptors. Our results provide the first evidence that the length of neuronal cilia can be modified by the lack of a neurotransmitter's input.


Subject(s)
Cilia/pathology , Dopaminergic Neurons/pathology , Parkinson Disease, Secondary/pathology , Ventral Striatum/pathology , Animals , Astrocytes/pathology , Cell Shape , Dopamine Agonists/pharmacology , Dopamine D2 Receptor Antagonists/pharmacology , Dopaminergic Neurons/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Parkinson Disease, Secondary/metabolism , Rats, Sprague-Dawley , Reserpine/pharmacology , Substantia Nigra/pathology
14.
Neurobiol Dis ; 59: 244-56, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23959140

ABSTRACT

Astrocytes are abundant neuron-supporting glial cells that harbor a powerful arsenal of neuroprotective antioxidative molecules and neurotrophic factors. Here we examined whether enrichment with healthy striatal astrocytes can provide neuroprotection against progressive dopaminergic neurodegeneration. Serotonin 1A (5-HT1A) agonist 8-OH-DPAT induced astrocyte proliferation and increased metallothionein-1/-2 (MT-1/-2), antioxidative molecules, in cultured astrocytes and the striatum of mice. Primary cultured mesencephalic dopamine neurons were protected against oxidative stress by preincubation with conditioned media from 8-OH-DPAT-treated astrocytes. These protective effects were canceled by 5-HT1A antagonist or MT-1/-2-specific antibody. Furthermore, reduction of nigrostriatal dopaminergic neurons in 6-hydroxydopamine-lesioned parkinsonian model mice was significantly abrogated by repeated injections of 8-OH-DPAT. Treatment with 8-OH-DPAT markedly increased the expression of MT in striatal astrocytes in the hemi-parkinsonian mice. Our study provides a promising therapeutic strategy of neuroprotection against oxidative stress and progressive dopaminergic neurodegeneration by demonstrating the efficacy of targeting 5-HT1A receptors in astrocytes.


Subject(s)
Astrocytes/metabolism , Dopaminergic Neurons/physiology , Parkinson Disease/pathology , Receptor, Serotonin, 5-HT1A/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/administration & dosage , Adrenergic Agents/toxicity , Animals , Astrocytes/drug effects , Brain/cytology , Buspirone/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian , Female , Functional Laterality/drug effects , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred ICR , Oxidopamine/toxicity , Parkinson Disease/etiology , Parkinson Disease/prevention & control , Pregnancy , Rats , Rats, Sprague-Dawley , Serotonin Receptor Agonists/administration & dosage , Water/pharmacology
15.
Neurochem Res ; 37(9): 1944-51, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22674083

ABSTRACT

Prostaglandin H synthase exerts not only cyclooxygenase activity but also peroxidase activity. The latter activity of the enzyme is thought to couple with oxidation of dopamine to dopamine quinone. Therefore, it has been proposed that cyclooxygenase inhibitors could suppress dopamine quinone formation. In the present study, we examined effects of various cyclooxygenase inhibitors against excess methyl L-3,4-dihydroxyphenylalanine (L-DOPA)-induced quinoprotein (protein-bound quinone) formation and neurotoxicity using dopaminergic CATH.a cells. The treatment with aspirin inhibited excess methyl L-DOPA-induced quinoprotein formation and cell death. However, acetaminophen did not show protective effects, and indomethacin and meloxicam rather aggravated these methyl L-DOPA-induced changes. Aspirin and indomethacin did not affect the level of glutathione that exerts quenching dopamine quinone in dopaminergic cells. In contrast with inhibiting effects of higher dose in the previous reports, relatively lower dose of aspirin that affected methyl L-DOPA-induced quinoprotein formation and cell death failed to prevent cyclooxygenase-induced dopamine chrome generation in cell-free system. Furthermore, aspirin but not acetaminophen or meloxicam showed direct dopamine quinone-scavenging effects in dopamine-semiquinone generating systems. The present results suggest that cyclooxygenase shows little contribution to dopamine oxidation in dopaminergic cells and that protective effects of aspirin against methyl L-DOPA-induced dopamine quinone neurotoxicity are based on its cyclooxygenase-independent property.


Subject(s)
Aspirin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Dopamine Agents/toxicity , Levodopa/antagonists & inhibitors , Levodopa/toxicity , Neuroprotective Agents , Quinones/toxicity , Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Cell-Free System , Cells, Cultured , Dopaminergic Neurons/drug effects , Glutathione/metabolism , Indomethacin/pharmacology , Meloxicam , Methyldopa/toxicity , Mice , Neurons/drug effects , Quinones/antagonists & inhibitors , Sympatholytics/toxicity , Thiazines/pharmacology , Thiazoles/pharmacology
16.
Neurol Res ; 33(10): 1050-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22196758

ABSTRACT

OBJECTIVES: Baicalein, a flavonoid derived from the root of Scutelaria baicalensis Georgi, possesses anti-oxidative properties including reactive oxygen species scavenging and lipid peroxidation inhibiting activities. The present study was undertaken to investigate the neuroprotective effect of baicalein against dopamine (DA) neurotoxicity induced by exposure to a synthetic DA precursor, L-3,4-dihydroxyphenylalanine (L-DOPA), in cultured dopaminergic CATH.a cells. METHODS AND RESULTS: Exposure to L-DOPA for 24 hours reduced the number of viable cells and enhanced protein-bound quinone (quinoprotein) formation in the cell. Both effects were prevented by simultaneous treatment with baicalein. In addition, baicalein prevented the formation of DA semiquinone radicals from DA in an in vitro cell-free system. Long-term baicalein treatment for 96 hours also protected against excess L-DOPA-induced cell death, and also increased glutathione (GSH) levels in CATH.a cells. DISCUSSION: Our results indicate that baicalein has neuroprotective properties against excess L-DOPA-induced DA neurotoxicity through the suppression of DA quinone formation. Furthermore, the long-term treatment of baicalein upregulates intracellular GSH contents, which may also exert neuroprotective effects against oxidative stress-induced neuronal damage.


Subject(s)
Antiparkinson Agents/toxicity , Dopamine/analogs & derivatives , Flavanones/pharmacology , Levodopa/toxicity , Neuroprotective Agents/pharmacology , Neurotoxins/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antiparkinson Agents/pharmacokinetics , Cell Line , Dopamine/metabolism , Dopamine/toxicity , Flavanones/therapeutic use , Levodopa/pharmacokinetics , Mice , Neuroprotective Agents/therapeutic use
17.
Glia ; 59(3): 435-51, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21264950

ABSTRACT

Our previous studies demonstrated the involvement of quinone formation in dopaminergic neuron dysfunction in the L-DOPA-treated parkinsonian model and in methamphetamine (METH) neurotoxicity. We further reported that the cysteine-rich metal-binding metallothionein (MT) family of proteins protects dopaminergic neurons against dopamine (DA) quinone neurotoxicity by its quinone-quenching property. The aim of this study was to examine MT induction in astrocytes in response to excess DA and the potential neuroprotective effects of astrocyte-derived MTs against DA quinone toxicity. DA exposure significantly upregulated MT-1/-2 in cultured striatal astrocytes, but not in mesencephalic neurons. This DA-induced MT upregulation in astrocytes was blocked by treatment with a DA-transporter (DAT) inhibitor, but not by DA-receptor antagonists. Expression of nuclear factor erythroid 2-related factor (Nrf2) and its binding activity to antioxidant response element of MT-1 gene were significantly increased in the astrocytes after DA exposure. Nuclear translocation of Nrf2 was suppressed by the DAT inhibitor. Quinone formation and reduction of mesencephalic DA neurons after DA exposure were ameliorated by preincubation with conditioned media from DA-treated astrocytes. These protective effects were abrogated by MT-1/-2-specific antibody. Adding exogenous MT-1 to glial conditioned media also showed similar neuroprotective effects. Furthermore, MT-1/-2 expression was markedly elevated specifically in reactive astrocytes in the striatum of L-DOPA-treated hemi-parkinsonian mice or METH-injected mice. These results suggested that excess DA taken up by astrocytes via DAT upregulates MT-1/-2 expression specifically in astrocytes, and that MTs or related molecules secreted specifically by astrocytes protect dopaminergic neurons from damage through quinone quenching and/or scavenging of free radicals.


Subject(s)
Astrocytes/physiology , Dopamine/analogs & derivatives , Dopamine/physiology , Metallothionein/metabolism , Metallothionein/physiology , Neurons/metabolism , Neuroprotective Agents/toxicity , Animals , Astrocytes/metabolism , Cells, Cultured , Coculture Techniques , Dopamine/toxicity , Free Radical Scavengers/metabolism , Free Radical Scavengers/pharmacology , Free Radical Scavengers/toxicity , Neurons/drug effects , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley
18.
J Clin Biochem Nutr ; 43(3): 185-90, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19015753

ABSTRACT

We have previously shown that extremely high level of guanidino compounds such as methylguanidine (MG), known as a neurotoxin and also a nephrotoxin, generate reactive oxygen species (ROS) using an electron spin resonance (ESR) technique with spin trap 5,5-dimethyl-1-pyrroline-N-oxide (DMPO). In this in vitro study, the inhibitory effect of fermented papaya preparation (SAIDO-PS501:PS-501) on hydroxyl radical (.OH) generation from MG was examined using an ESR spectrometry, and it was found that PS-501 suppressed .OH generation from MG in a dose-dependent manner. The ID(50) value of PS-501 was 8 mg/ml. On the contrary, glucose itself did not suppress .OH generation from MG up to100 mg/ml, whereas PS-501 almost completely suppressed .OH generation from MG at a dose of 100 mg/ml. These results imply that PS-501 itself may have a beneficial effect of preventing ROS- and MG-related diseases.

SELECTION OF CITATIONS
SEARCH DETAIL
...