Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Neuroinflammation ; 15(1): 52, 2018 Feb 21.
Article in English | MEDLINE | ID: mdl-29467000

ABSTRACT

BACKGROUND: We previously reported that ginsenoside Re (GRe) attenuated against methamphetamine (MA)-induced neurotoxicity via anti-inflammatory and antioxidant potentials. We also demonstrated that dynorphin possesses anti-inflammatory and antioxidant potentials against dopaminergic loss, and that balance between dynorphin and substance P is important for dopaminergic neuroprotection. Thus, we examined whether GRe positively affects interactive modulation between dynorphin and substance P against MA neurotoxicity in mice. METHODS: We examined changes in dynorphin peptide level, prodynorphin mRNA, and substance P mRNA, substance P-immunoreactivity, homeostasis in enzymatic antioxidant system, oxidative parameter, microglial activation, and pro-apoptotic parameter after a neurotoxic dose of MA to clarify the effects of GRe, prodynorphin knockout, pharmacological inhibition of κ-opioid receptor (i.e., nor-binaltorphimine), or neurokinin 1 (NK1) receptor (i.e., L-733,060) against MA insult in mice. RESULTS: GRe attenuated MA-induced decreases in dynorphin level, prodynorphin mRNA expression in the striatum of wild-type (WT) mice. Prodynorphin knockout potentiated MA-induced dopaminergic toxicity in mice. The imbalance of enzymatic antioxidant system, oxidative burdens, microgliosis, and pro-apoptotic changes led to the dopaminergic neurotoxicity. Neuroprotective effects of GRe were more pronounced in prodynorphin knockout than in WT mice. Nor-binaltorphimine, a κ-opioid receptor antagonist, counteracted against protective effects of GRe. In addition, we found that GRe significantly attenuated MA-induced increases in substance P-immunoreactivity and substance P mRNA expression in the substantia nigra. These increases were more evident in prodynorphin knockout than in WT mice. Although, we observed that substance P-immunoreactivity was co-localized in NeuN-immunreactive neurons, GFAP-immunoreactive astrocytes, and Iba-1-immunoreactive microglia. NK1 receptor antagonist L-733,060 or GRe selectively inhibited microgliosis induced by MA. Furthermore, L-733,060 did not show any additive effects against GRe-mediated protective activity (i.e., antioxidant, antimicroglial, and antiapoptotic effects), indicating that NK1 receptor is one of the molecular targets of GRe. CONCLUSIONS: Our results suggest that GRe protects MA-induced dopaminergic neurotoxicity via upregulatgion of dynorphin-mediated κ-opioid receptor and downregulation of substance P-mediated NK1 R.


Subject(s)
Dopaminergic Neurons/metabolism , Dynorphins/metabolism , Ginsenosides/pharmacology , Methamphetamine/toxicity , Receptors, Neurokinin-1/metabolism , Receptors, Opioid, kappa/metabolism , Substance P/metabolism , Animals , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurokinin-1 Receptor Antagonists/pharmacology , Piperidines/pharmacology , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
2.
J Neuroinflammation ; 9: 124, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22695044

ABSTRACT

BACKGROUND: The striato-nigral projecting pathway contains the highest concentrations of dynorphin in the brain. The functional role of this opioid peptide in the regulation of mesencephalic dopaminergic (DAergic) neurons is not clear. We reported previously that exogenous dynorphin exerts potent neuroprotective effects against inflammation-induced dopaminergic neurodegeneration in vitro. The present study was performed to investigate whether endogenous dynorphin has neuroprotective roles in vivo. METHODS: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and methamphetamine (MA), two commonly used neurotoxins in rodent models of Parkinson's disease, were administered to wild-type (Dyn⁺/⁺) and prodynorphin-deficient mice (Dyn⁻/⁻). We examined dopaminergic neurotoxicity by using an automated video tracking system, HPLC, immunocytochemistry, and reverse transcription and polymerase chain reaction (RT-PCR). RESULTS: Treatment with MPTP resulted in behavioral impairments in both strains. However, these impairments were more pronounced in Dyn-l- than in Dyn⁺/⁺. Dyn⁻/⁻ showed more severe MPTP-induced dopaminergic neuronal loss in the substantia nigra and striatum than Dyn⁺/⁺. Similarly, the levels of dopamine and its metabolites in the striatum were depleted to a greater extent in Dyn⁻/⁻ than in Dyn⁺/⁺. Additional mechanistic studies revealed that MPTP treatment caused a higher degree of microglial activation and M1 phenotype differentiation in Dyn⁻/⁻ than in Dyn⁺/⁺. Consistent with these observations, prodynorphin deficiency also exacerbated neurotoxic effects induced by MA, although this effect was less pronounced than that of MPTP. CONCLUSIONS: The in vivo results presented here extend our previous in vitro findings and further indicate that endogenous dynorphin plays a critical role in protecting dopaminergic neurons through its anti-inflammatory effects.


Subject(s)
Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Dynorphins/physiology , Motor Skills Disorders/prevention & control , Neurotoxins/toxicity , Substantia Nigra/metabolism , Animals , Corpus Striatum/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dynorphins/deficiency , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , MPTP Poisoning/prevention & control , Methamphetamine/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Motor Skills Disorders/metabolism , Motor Skills Disorders/pathology , Substantia Nigra/drug effects , Substantia Nigra/pathology
3.
Arch Pharm Res ; 44(7): 668-688, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34286473

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.


Subject(s)
Corpus Striatum/pathology , Dopaminergic Neurons/drug effects , Methamphetamine/toxicity , Parkinson Disease, Secondary/pathology , Animals , Apoptosis/drug effects , Corpus Striatum/cytology , Corpus Striatum/drug effects , Disease Models, Animal , Dopaminergic Neurons/cytology , Humans , Methamphetamine/administration & dosage , Mice , Mitochondrial Dynamics/drug effects , Oxidative Stress/drug effects , Rats
4.
Food Chem Toxicol ; 154: 112313, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34082047

ABSTRACT

We suggested that selenium-dependent glutathione peroxidase (GPx) plays a protective role against methamphetamine (MA)-induced dopaminergic toxicity. We focused on GPx-1, a major selenium-dependent enzyme and constructed a GPx-1 gene-encoded adenoviral vector (Ad-GPx-1) to delineate the role of GPx-1 in MA-induced dopaminergic neurotoxicity. Exposure to Ad-GPx-1 significantly induced GPx activity and GPx-1 protein levels in GPx-1-knockout (GPx-1-KO) mice. MA-induced dopaminergic impairments [i.e., hyperthermia; increased nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) DNA-binding activity; and decreased dopamine levels, TH activity, and behavioral activity] were more pronounced in GPx-1-KO mice than in WT mice. In contrast, exposure to Ad-GPx-1 significantly attenuated MA-induced dopaminergic loss in GPx-1-KO mice. The protective effect exerted by Ad-GPx-1 was comparable to that exerted by pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor against MA insult. Consistently, GPx-1 overexpression significantly attenuated MA dopaminergic toxicity in mice. PDTC did not significantly impact the protective effect of GPx-1 overexpression, suggesting that interaction between NF-κB and GPx-1 is critical for dopaminergic protection. Thus, NF-κB is a potential therapeutic target for GPx-1-mediated dopaminergic protective activity. This study for the first time demonstrated that Ad-GPx-1 rescued dopaminergic toxicity in vivo following MA insult. Furthermore, GPx-1-associated therapeutic interventions may be important against dopaminergic toxicity.


Subject(s)
Dependovirus/genetics , Genetic Vectors , Glutathione Peroxidase/genetics , Methamphetamine/toxicity , NF-kappa B/metabolism , Animals , Behavior, Animal/drug effects , Dopamine/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Glutathione Peroxidase GPX1
5.
J Neurochem ; 112(3): 773-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19922440

ABSTRACT

Trichloroethylene, a chlorinated solvent widely used as a degreasing agent, is a common environmental contaminant. Emerging evidence suggests that chronic exposure to trichloroethylene may contribute to the development of Parkinson's disease. The purpose of this study was to determine if selective loss of nigrostriatal dopaminergic neurons could be reproduced by systemic exposure of adult Fisher 344 rats to trichloroethylene. In our experiments, oral administration of trichloroethylene induced a significant loss of dopaminergic neurons in the substantia nigra pars compacta in a dose-dependent manner, whereas the number of both cholinergic and GABAergic neurons were not decreased in the striatum. There was a robust decline in striatal levels of 3, 4-dihydroxyphenylacetic acid without a significant depletion of striatal dopamine. Rats treated with trichloroethylene showed defects in rotarod behavior test. We also found a significantly reduced mitochondrial complex I activity with elevated oxidative stress markers and activated microglia in the nigral area. In addition, we observed intracellular alpha-synuclein accumulation in the dorsal motor nucleus of the vagus nerve, with some in nigral neurons, but little in neurons of cerebral cortex. Overall, our animal model exhibits some important features of Parkinsonism, and further supports that trichloroethylene may be an environmental risk factors for Parkinson's disease.


Subject(s)
Dopamine/metabolism , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Solvents/toxicity , Substantia Nigra/metabolism , Trichloroethylene/toxicity , Animals , CD11b Antigen/metabolism , Caspase 3/metabolism , Choline O-Acetyltransferase/metabolism , Chromatography, High Pressure Liquid/methods , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Dose-Response Relationship, Drug , Electrochemistry/methods , Encephalitis/chemically induced , Gene Expression Regulation/drug effects , Male , Mitochondria/drug effects , Neurodegenerative Diseases/physiopathology , Oxidative Stress/drug effects , Rats , Rats, Inbred F344 , Rotarod Performance Test , Substantia Nigra/pathology , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
6.
Nature ; 424(6948): 556-61, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12891359

ABSTRACT

The neuropathological hallmarks of Alzheimer's disease and other tauopathies include senile plaques and/or neurofibrillary tangles. Although mouse models have been created by overexpressing specific proteins including beta-amyloid precursor protein, presenilin and tau, no model has been generated by gene knockout. Phosphorylation of tau and other proteins on serine or threonine residues preceding proline seems to precede tangle formation and neurodegeneration in Alzheimer's disease. Notably, these phospho(Ser/Thr)-Pro motifs exist in two distinct conformations, whose conversion in some proteins is catalysed by the Pin1 prolyl isomerase. Pin1 activity can directly restore the conformation and function of phosphorylated tau or it can do so indirectly by promoting its dephosphorylation, which suggests that Pin1 is involved in neurodegeneration; however, genetic evidence is lacking. Here we show that Pin1 expression is inversely correlated with predicted neuronal vulnerability and actual neurofibrillary degeneration in Alzheimer's disease. Pin1 knockout in mice causes progressive age-dependent neuropathy characterized by motor and behavioural deficits, tau hyperphosphorylation, tau filament formation and neuronal degeneration. Thus, Pin1 is pivotal in protecting against age-dependent neurodegeneration, providing insight into the pathogenesis and treatment of Alzheimer's disease and other tauopathies.


Subject(s)
Aging/physiology , Peptidylprolyl Isomerase/metabolism , Tauopathies/enzymology , Tauopathies/prevention & control , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Alzheimer Disease/prevention & control , Amino Acid Motifs , Animals , Behavior, Animal/physiology , Gene Deletion , Gene Expression , Humans , Mice , Mice, Knockout , Microscopy, Electron , Motor Activity/physiology , NIMA-Interacting Peptidylprolyl Isomerase , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/metabolism , Peptidylprolyl Isomerase/genetics , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Tauopathies/pathology , Tauopathies/physiopathology
7.
J Neurosci Res ; 87(8): 1913-21, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19224579

ABSTRACT

A role for inflammation has been hypothesized in the etiology and progression of Parkinson's disease (PD). In this study, we generated, characterized, and validated the first progressive PD-related mouse model (C57/B6) with intrastriatal injection of lipopolysaccharide (LPS). We showed progressive and specific dopaminergic neurodegeneration in the substantia nigra, which is accompanied by striatal dopamine depletion and progressive behavioral impairment, which was alleviated by the use of the PD drug L-Dopa. We focused on the role of nitric oxide (NO) in inflammation-promoted cell death and suggest that the expression of the inducible NO synthase plays a role in the progressive loss of dopaminergic neurons but not the initial loss induced by LPS. With this model, future research can be performed in gene knockout mice to study other potential mechanisms of inflammation-induced neurodegeneration. In addition, this model can be used to screen therapeutics for PD at a more clinically relevant time (i.e., after LPS injection but before manifestation of PD-related behavioral impairment), because most PD drugs are screened in animal models in which inhibitors are given predisease induction. Thus, this novel PD-related model should be further characterized and strongly considered as a tool for future drug studies.


Subject(s)
Corpus Striatum/drug effects , Encephalitis/metabolism , Nerve Degeneration/metabolism , Neurons/drug effects , Nitric Oxide/metabolism , Parkinsonian Disorders/metabolism , Animals , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Disease Progression , Dopamine/deficiency , Drug Evaluation, Preclinical/methods , Encephalitis/chemically induced , Encephalitis/physiopathology , Inflammation Mediators/toxicity , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Neurons/metabolism , Neurons/pathology , Nitric Oxide Synthase Type II/metabolism , Parkinsonian Disorders/physiopathology , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/physiopathology
8.
J Neurosci Res ; 87(16): 3679-86, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19598248

ABSTRACT

Microsomal epoxide hydrolase (mEH) and cytochrome P-450 (CYP) ensure the rapid detoxification of epoxides generated during the oxidative metabolism of xenobiotics. Although CYP has been demonstrated to modulate methamphetamine (METH)-induced behavioral effects, little is known about the role of the mEH gene on these effects. We examined the role of mEH gene expression in METH-induced conditioned place preference and behavioral sensitization by using mEH(-/-) and wild-type (WT) mice. Extracellular dopamine (DA) levels and DA uptake into synaptosomes were assessed by using an in vivo microdialysis and [(3)H]DA uptake assay. We applied double-label immunocytochemistry to characterize mEH-positive cellular types. METH-induced behavioral responses paralleled striatal c-Fos-like immunoreactivity. METH treatment resulted in increased extracellular DA levels in the nucleus accumbens but decreased synaptosomal DA uptake in the striatum. These behavioral and neurochemical changes were more pronounced in the mEH(-/-) mice than in WT mice. In WT mice, mEH-like immunoreactivity was expressed in astrocytes labeled by GFAP or S100B after METH treatment. The results suggest that epoxide intermediates mediate METH drug dependence and that astrocytic reactions of mEH protein are important in the endogenous modulation in response to METH drug dependence.


Subject(s)
Amphetamine-Related Disorders/enzymology , Conditioning, Operant/drug effects , Epoxide Hydrolases/metabolism , Amphetamine-Related Disorders/genetics , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Epoxide Hydrolases/genetics , Immunohistochemistry , Methamphetamine/pharmacology , Mice , Mice, Knockout , Microdialysis , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spatial Behavior/drug effects
9.
Ann Neurol ; 63(2): 184-92, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18157908

ABSTRACT

OBJECTIVE: To analyze a cluster of 30 industrial coworkers with Parkinson's disease and parkinsonism subjected to long-term (8-33 years) chronic exposure to trichloroethylene. METHODS: Neurological evaluations were conducted on the 30 coworkers, including a general physical and neurological examination and the Unified Parkinson's Disease Rating Scale. In addition, fine motor speed was quantified and an occupational history survey was administered. Next, animal studies were conducted to determine whether trichloroethylene exposure is neurotoxic to the nigrostriatal dopamine system that degenerates in Parkinson's disease. The experiments specifically analyzed complex 1 mitochondrial neurotoxicity because this is a mechanism of action of other known environmental dopaminergic neurotoxins. RESULTS: The three workers with workstations adjacent to the trichloroethylene source and subjected to chronic inhalation and dermal exposure from handling trichloroethylene-soaked metal parts had Parkinson's disease. Coworkers more distant from the trichloroethylene source, receiving chronic respiratory exposure, displayed many features of parkinsonism, including significant motor slowing. Neurotoxic actions of trichloroethylene were demonstrated in accompanying animal studies showing that oral administration of trichloroethylene for 6 weeks instigated selective complex 1 mitochondrial impairment in the midbrain with concomitant striatonigral fiber degeneration and loss of dopamine neurons. INTERPRETATION: Trichloroethylene, used extensively in industry and the military and a common environmental contaminant, joins other mitochondrial neurotoxins, MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and some pesticides, as a risk factor for parkinsonism.


Subject(s)
Brain/drug effects , Electron Transport Complex I/drug effects , Mitochondria/drug effects , Occupational Exposure/statistics & numerical data , Parkinson Disease, Secondary/chemically induced , Trichloroethylene/toxicity , Adult , Aged , Animals , Brain/metabolism , Brain/physiopathology , Cluster Analysis , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Dopamine/metabolism , Electron Transport Complex I/metabolism , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Humans , Male , Middle Aged , Mitochondria/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Parkinson Disease, Secondary/diagnosis , Parkinson Disease, Secondary/physiopathology , Rats , Rats, Inbred F344 , Severity of Illness Index , Solvents/poisoning , Solvents/toxicity , Substantia Nigra/drug effects , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Toxicity Tests, Acute , Trichloroethylene/poisoning
10.
Cell Signal ; 20(4): 645-58, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18248956

ABSTRACT

We have demonstrated that kainate (KA) induces a reduction in mitochondrial Mn-superoxide dismutase (Mn-SOD) expression in the rat hippocampus and that KA-induced oxidative damage is more prominent in senile-prone (SAM-P8) than senile-resistant (SAM-R1) mice. To extend this, we examined whether KA seizure sensitivity contributed to mitochondrial degeneration in these mouse strains. KA-induced seizure susceptibility in SAM-P8 mice paralleled prominent increases in lipid peroxidation and protein oxidation and was accompanied by significant impairment in glutathione homeostasis in the hippocampus. These findings were more pronounced in the mitochondrial fraction than in the hippocampal homogenate. Consistently, KA-induced decreases in Mn-SOD protein expression, mitochondrial transmembrane potential, and uncoupling protein (UCP)-2 expression were more prominent in SAM-P8 than SAM-R1 mice. Marked release of cytochrome c from mitochondria into the cytosol and a higher level of caspase-3 cleavage were observed in KA-treated SAM-P8 mice. Additionally, electron microscopic evaluation indicated that KA-induced increases in mitochondrial damage and lipofuscin-like substances were more pronounced in SAM-P8 than SAM-R1 animals. These results suggest that KA-mediated mitochondrial oxidative stress contributed to hippocampal degeneration in the senile-prone mouse.


Subject(s)
Aging, Premature/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Oxidative Stress , Aging, Premature/genetics , Aging, Premature/pathology , Animals , Caspase 3/metabolism , Cytochromes c/metabolism , Disease Models, Animal , Enzyme Activation , Glutathione/metabolism , Glutathione Disulfide/metabolism , Hippocampus/enzymology , Hippocampus/ultrastructure , Ion Channels/metabolism , Kainic Acid , Lipid Peroxidation/drug effects , Lipofuscin/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred Strains , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurons/enzymology , Neurons/ultrastructure , Oxidation-Reduction , Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Seizures/chemically induced , Seizures/genetics , Seizures/metabolism , Seizures/pathology , Superoxide Dismutase/metabolism , Time Factors , Uncoupling Protein 2
11.
Food Chem Toxicol ; 133: 110761, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31422080

ABSTRACT

We previously demonstrated that activation of protein kinase Cδ (PKCδ) is critical for methamphetamine (MA)-induced dopaminergic toxicity. It was recognized that microsomal epoxide hydrolase (mEH) also induces dopaminergic neurotoxicity. It was demonstrated that inhibition of PKC modulates the expression of mEH. We investigated whether MA-induced PKCδ activation requires mEH induction in mice. MA treatment (8 mg/kg, i.p., × 4; 2 h interval) significantly enhanced the level of phosphorylated PKCδ in the striatum of wild type (WT) mice. Subsequently, treatment with MA resulted in significant increases in the expression of cleaved PKCδ and mEH. Treatment with MA resulted in enhanced interaction between PKCδ and mEH. PKCδ knockout mice exhibited significant attenuation of the enhanced mEH expression induced by MA. MA-induced hyperthermia, oxidative stress, proapoptotic potentials, and dopaminergic impairments were attenuated by PKCδ knockout or mEH knockout in mice. However, treating mEH knockout in mice with PKCδ inhibitor, rottlerin did not show any additive beneficial effects, indicating that mEH is a critical mediator of neurotoxic potential of PKCδ. Our results suggest that MA-induced PKCδ activation requires mEH induction as a downstream signaling pathway and that the modulation of the PKCδ and mEH interaction is important for the pharmacological intervention against MA-induced dopaminergic neurotoxicity.


Subject(s)
Dopaminergic Neurons/metabolism , Epoxide Hydrolases/metabolism , Methamphetamine/adverse effects , Neurotoxicity Syndromes/metabolism , Protein Kinase C-delta/metabolism , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Dopaminergic Neurons/drug effects , Epoxide Hydrolases/genetics , Fever/genetics , Gene Knockout Techniques , Locomotion/genetics , Mice, Inbred C57BL , Mice, Knockout , Neurotoxicity Syndromes/genetics , Oxidative Stress/genetics , Protein Kinase C-delta/genetics
12.
Neurochem Int ; 124: 68-81, 2019 03.
Article in English | MEDLINE | ID: mdl-30597180

ABSTRACT

Previously we demonstrated that p53 mediates dopaminergic neurotoxicity via inducing mitochondrial burdens and proapoptotsis. However, little is known about the role of p53 in the excitotoxicity induced by psychostimulant, such as cocaine. Cocaine-induced kindling (convulsive) behaviors significantly increased p53 expression in the brain. Cocaine-induced p53 expression was more pronounced in hippocampus than in striatum or prefrontal cortex. Genetic depletion of p53 significantly attenuated cocaine-induced convulsive behaviors, followed by c-Fos immunoreactivity, and oxidative burdens in the hippocampus of mice. The antioxidant potentials mediated by genetic depletion of p53 were more pronounced in the mitochondrial-than cytosolic-fraction. Depletion of p53 significantly attenuated the changes in mitochondrial transmembrane potential, intramitochondrial Ca2+ level, and mitochondrial oxidative burdens induced by cocaine. Consistently, depletion of p53 significantly inhibited mitochondrial p53 translocation, and cleaved-PKCδ induced by cocaine. In addition, depletion of p53 protected from cytosolic cytochrome c release, and pro-apoptotic changes induced by cocaine. Importantly, the protective/anticonvulsant potentials by genetic depletion of p53 were comparable to those by pifithrin-µ (PFT), a p53 inhibitor. Our results suggest that depletion of p53 offers anticonvulsive and neuroprotective potentials mainly via attenuating mitochondrial oxidative burdens, mitochondrial dysfunction, and pro-apoptotic signalings against cocaine-induced convulsive neurotoxicity.


Subject(s)
Apoptosis/physiology , Cocaine/toxicity , Kindling, Neurologic/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Tumor Suppressor Protein p53/deficiency , Animals , Apoptosis/drug effects , Kindling, Neurologic/drug effects , Kindling, Neurologic/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/genetics , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Random Allocation , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
13.
J Neuroinflammation ; 5: 4, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18205920

ABSTRACT

BACKGROUND: Previous studies have suggested that peroxisome proliferator activated receptor-gamma (PPAR-gamma)-mediated neuroprotection involves inhibition of microglial activation and decreased expression and activity of inducible nitric oxide synthase (iNOS); however, the underlying molecular mechanisms have not yet been well established. In the present study we explored: (1) the effect of the PPAR-gamma agonist pioglitazone on lipopolysaccharide (LPS)-induced iNOS activity and nitric oxide (NO) generation by microglia; (2) the differential role of p38 mitogen-activated protein kinase (p38 MAPK), c-Jun NH(2)-terminal kinase (JNK), and phosphoinositide 3-kinase (PI3K) on LPS-induced NO generation; and (3) the regulation of p38 MAPK, JNK, and PI3K by pioglitazone. METHODS: Mesencephalic neuron-microglia mixed cultures, and microglia-enriched cultures were treated with pioglitazone and/or LPS. The protein levels of iNOS, p38 MAPK, JNK, PPAR-gamma, PI3K, and protein kinase B (Akt) were measured by western blot. Different specific inhibitors of iNOS, p38MAPK, JNK, PI3K, and Akt were used in our experiment, and NO generation was measured using a nitrite oxide assay kit. Tyrosine hydroxylase (TH)-positive neurons were counted in mesencephalic neuron-microglia mixed cultures. RESULTS: Our results showed that pioglitazone inhibits LPS-induced iNOS expression and NO generation, and inhibition of iNOS is sufficient to protect dopaminergic neurons against LPS insult. In addition, inhibition of p38 MAPK, but not JNK, prevented LPS-induced NO generation. Further, and of interest, pioglitazone inhibited LPS-induced phosphorylation of p38 MAPK. Wortmannin, a specific PI3K inhibitor, enhanced p38 MAPK phosphorylation upon LPS stimulation of microglia. Elevations of phosphorylated PPAR-gamma, PI3K, and Akt levels were observed with pioglitazone treatment, and inhibition of PI3K activity enhanced LPS-induced NO production. Furthermore, wortmannin prevented the inhibitory effect of pioglitazone on the LPS-induced NO increase. CONCLUSION: We demonstrate that pioglitazone protects dopaminergic neurons against LPS insult at least via inhibiting iNOS expression and NO generation, which is potentially mediated via inhibition of p38 MAPK activity. In addition, the PI3K pathway actively participates in the negative regulation of LPS-induced NO production. Our findings suggest that PPAR-gamma activation may involve differential regulation of p38 MAPK and of the PI3K/Akt pathway in the regulation of the inflammatory process.


Subject(s)
Encephalitis/drug therapy , Encephalitis/enzymology , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Thiazolidinediones/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Brain/enzymology , Cells, Cultured , Coculture Techniques , Dopamine/metabolism , Encephalitis/physiopathology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Hypoglycemic Agents/pharmacology , Inflammation Mediators , JNK Mitogen-Activated Protein Kinases/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides , Microglia/drug effects , Microglia/enzymology , Neurons/drug effects , Neurons/enzymology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/drug effects , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Pioglitazone , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/drug effects
14.
J Neurosci Res ; 86(12): 2792-801, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18500758

ABSTRACT

Parkinson's disease (PD) is the most prevalent neurodegenerative movement disorder. Epidemiological studies have suggested most cases of PD are linked to environmental risk factors. Microsomal epoxide hydrolase (mEH) is a conserved enzyme that catalyzes hydrolysis of a large number of epoxide intermediates such as drugs and epoxides of environmental toxins. We hypothesize that changes in mEH are involved in the pathogenesis of PD by modulating the vulnerability of dopaminergic neurons to environmental stress. Herein we reported that acute treatment with the neurotoxin MPTP (1-methyl-4-phemyl-1,2,3,6-tetrahydropyridine) markedly increased the mEH immunoreactivity in the nigrostriatal system of C57BL/6 mice. Next, mEH knockout (KO) mice were used, and we found that tyrosine hydroxylase (TH)-positive cell loss was significantly lower in the substantia nigra of mEH KO mice compared with wild-type (WT) mice after MPTP treatment. The mean dopamine turnover ratios were significantly increased in MPTP-treated mEH KO mice compared with WT. In addition, TH is the rate-limiting enzyme for dopamine biosynthesis, and its activity is mainly regulated by TH phosphorylation at Ser-31 (pSer31) and Ser-40 (pSer40). Double immunofluorescence showed that both pSer31 and pSer40 are completely colocalized in total TH-positive cells. However, immunoblotting confirmed that there was a significantly higher level of pSer31 in mEH-KO mice when compared with WT mice after MPTP, and no marked differences among TH and its phosphorylation levels occurred after saline injection. These data suggested that mEH deficiency facilitates TH phosphorylation in the nigrostriatal dopamine system, which may be associated with an increased resistance of dopaminergic neurons to environmental toxins.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Epoxide Hydrolases/deficiency , Epoxide Hydrolases/genetics , Gene Deletion , Microsomes/enzymology , Tyrosine 3-Monooxygenase/metabolism , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Astrocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microsomes/metabolism , Phosphorylation/drug effects
15.
Neurosci Lett ; 432(3): 198-201, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18207323

ABSTRACT

We created an inflammation-induced Parkinson's disease model, where microglia activation leads to oxidative stress, mitochondrial dysfunction, and dopaminergic neurodegeneration in the substantia nigra. Pioglitazone, an agonist of peroxisome proliferator activated receptor-gamma (PPAR-gamma), can prevent these deficits and protect dopaminergic neurons. To continue exploring the effects of pioglitazone in this model we focused on the expression of PPAR-gamma, uncoupling protein 2 (UCP2), and mitoNEET. We report that intrastriatal lipopolysaccharide (LPS) increases striatal PPAR-gamma, UCP2, and mitoNEET expression, and pioglitazone attenuates these LPS-induced changes.


Subject(s)
Corpus Striatum/drug effects , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Lipopolysaccharides/pharmacology , Thiazolidinediones/pharmacology , Animals , Ion Channels/metabolism , Male , Mitochondrial Proteins/metabolism , PPAR gamma/metabolism , Pioglitazone , Rats , Rats, Sprague-Dawley , Uncoupling Protein 2
16.
Neurosci Lett ; 445(1): 1-6, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18760329

ABSTRACT

Anti-inflammatory drugs such as ibuprofen appear to prevent the development of Parkinson's disease (PD); however, long-term use has undesirable side-effects. A new strategy for anti-inflammatory drug therapy is using a dual inhibitor of COX and lipooxygenase (LOX). Here, we compared the dopaminergic neuroprotective property of phenidone (a dual COX and LOX inhibitor) with COX or LOX inhibitors including SC-560 (a COX-1 inhibitor), aspirin (a COX-1/2 inhibitor), meloxicam (a preferential COX-2 inhibitor), caffeic acid (a 5-LOX inhibitor), and esculetin (a 5, 12-LOX inhibitor) in our lipopolysaccharide (LPS)-induced PD animal model. Our results show that COX-2 and 5-LOX play a major role in LPS-induced dopaminergic neurotoxicity, as meloxicam and phenidone attenuated LPS-induced oxidative stress and meloxicam, phenidone, and caffeic acid attenuated dopaminergic neurodegeneration, while SC-560, aspirin, and esculetin did not. In addition, phenidone was superior in attenuating LPS-induced dopaminergic neurodegeneration and microglia activation, probably as a result of dual inhibition of COX-2 and LOX. Therefore, dual inhibition of COX and LOX with phenidone represents a promising new candidate for anti-inflammatory drug therapy, and may provide a novel therapeutic approach for inflammation-related neurodegenerative diseases including PD.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Dopamine/metabolism , Neurons/drug effects , Neurotoxicity Syndromes , Pyrazoles/pharmacology , Substantia Nigra/pathology , Analysis of Variance , Animals , CD11b Antigen/metabolism , Disease Models, Animal , Drug Interactions , Lipopolysaccharides/toxicity , Male , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/prevention & control , Rats , Rats, Sprague-Dawley
17.
Toxicology ; 410: 106-115, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30236991

ABSTRACT

Chlorpyrifos (CPF) may weaken the immune defenses of children, making them vulnerable to opportunistic bacterial infection. CPF combined with bacterial infection is a potential problem for children during their childhood development. However, there is a lack of studies on the joint effects of these two factors on children. Here, we assessed the effects of CPF combined with lipopolysaccharide (LPS) on the inflammation and development of the nervous system. In this study, the cell toxicity of CPF plus LPS in cultured astrocytes, and the pathogenic effects of CPF plus LPS in neonatal rat models were observed. The hydrogen (H2)-inhalation was used for treatment to explore its therapeutic potential. We found that CPF plus LPS activated the astrocyte, which increased the expressions of HMGB1, TLR4, and p-NF-κB p65, while H2-inhalation reduced the expressions (p < 0.05). We also found that CPF plus LPS induced long-lasting spatial memory deficits throughout brain maturation. However, H2-inhalation improved rat performance in these behavioral experiments (p < 0.05). In conclusion, the sub-toxic concentration of CPF did not cause a significant damage in short term, but induced a severe long-term damage to the brain when combined with LPS. H2-inhalation reduced the neuronal damage and behavioral abnormalities caused by CPF and LPS exposure.


Subject(s)
Chlorpyrifos/toxicity , Inflammation/chemically induced , Insecticides/toxicity , Lipopolysaccharides/toxicity , Memory Disorders/chemically induced , Memory Disorders/pathology , Spatial Memory/drug effects , Animals , Animals, Newborn , Astrocytes/drug effects , Female , Gene Expression/drug effects , HMGB1 Protein/biosynthesis , Hydrogen/pharmacology , Inflammation/pathology , Male , Maze Learning/drug effects , Pregnancy , Prenatal Exposure Delayed Effects , Primary Cell Culture , Rats , Toll-Like Receptor 4/drug effects
18.
Mol Neurobiol ; 55(7): 6201-6214, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29270919

ABSTRACT

This study was conducted to investigate the mechanism of action and extent of selective dopaminergic neurodegeneration caused by exposure to trichloroethylene (TCE) leading to the endogenous formation of the neurotoxin 1-trichloromethyl-1,2,3,4-tetrahydro-ß-carboline (TaClo) in rodents. Beginning at 3 months of age, male C57BL/6 mice received oral TCE dissolved in vehicle for 8 months. Dopaminergic neuronal loss was assessed by nigral tyrosine hydroxylase (TH) immunoreactivity. Selective dopaminergic neurodegeneration was determined based on histological analysis of non-dopaminergic neurons in the brain. Behavioral assays were evaluated using open field activity and rotarod tests. Mitochondrial complex I activity, oxidative stress markers, and microglial activation were also examined in the substantia nigra. The level of TaClo was detected using HPLC-electrospray ionization tandem mass spectrometry. Dopaminergic neurotoxicity of TaClo was determined in midbrain organotypic cultures from rat pups. Following 8 months of TCE treatment, there was a progressive and selective loss of 50% of the dopaminergic neurons in mouse substantia nigra (SN) and about 50% loss of dopamine and 72% loss of 3,4-dihydroxyphenylacetic acid in the striatum, respectively. In addition, motor deficits, mitochondrial impairment, oxidative stress, and inflammation were measured. TaClo content was quantified in the brain after TCE treatment. In organotypic cultures, TaClo rather than TCE induced dopaminergic neuronal loss, similar to MPP+. TCE exposure may stimulate the endogenous formation of TaClo, which is responsible for dopaminergic neurodegeneration. However, even prolonged administration of TCE was insufficient for producing a greater than 50% loss of nigral dopamine neurons, indicating that additional co-morbid factors would be needed for mimicking the profound loss of dopamine neurons seen in Parkinson's disease.


Subject(s)
Parkinson Disease/etiology , Risk Assessment , Trichloroethylene/toxicity , Administration, Oral , Animals , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dopamine/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Degeneration/pathology , Neurotoxins/toxicity , Oxidative Stress/drug effects , Parkinson Disease/pathology , Protein Folding/drug effects , Substantia Nigra/drug effects , Substantia Nigra/pathology , Trichloroethylene/administration & dosage , alpha-Synuclein/metabolism
19.
J Neuroimmunol ; 192(1-2): 89-98, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17976742

ABSTRACT

Increasing evidence links neuroinflammation to Parkinson's disease. Microglia are mediators of neuroinflammation. Overactivation of microglia contributes to the release of cyclooxygenase 2 and prostaglandin E(2) during neuronal insults. We have previously shown that pioglitazone, a peroxisome proliferator-activated receptor gamma agonist, inhibits microglia activation, reduces proinflammatory factors, and protects dopaminergic neurons. Here, we demonstrated that pioglitazone protects dopaminergic neurons by inhibiting abnormal microglia activation, interfering with phosphorylation of jun N-terminal kinase and nuclear factor kappa-B, and by suppressing cyclooxygenase 2 expression and the subsequent prostaglandin E(2) synthesis. Therefore, the anti-inflammatory properties of pioglitazone may be useful for ameliorating the progression of Parkinson's disease.


Subject(s)
Cyclooxygenase 2/metabolism , Dopamine/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Thiazolidinediones/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Encephalitis/chemically induced , Encephalitis/pathology , Encephalitis/prevention & control , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , Lipopolysaccharides , Mesencephalon/cytology , Microglia/drug effects , Pioglitazone , Pregnancy , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
20.
Neurochem Int ; 50(6): 791-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17386960

ABSTRACT

We showed that dextromethorphan (DM) provides neuroprotective/anticonvulsant effects and that DM and its major metabolite, dextrorphan, have a high-affinity for sigma(1) receptors, but a low affinity for sigma(2) receptors. In addition, we found that DM has a higher affinity than DX for sigma(1) sites, whereas DX has a higher affinity than DM for PCP sites. We extend our earlier findings by showing that DM attenuated trimethyltin (TMT)-induced neurotoxicity (convulsions, hippocampal degeneration and spatial memory impairment) in rats. This attenuation was reversed by the sigma(1) receptor antagonist BD 1047, but not by the sigma(2) receptor antagonist ifenprodil. DM attenuates TMT-induced reduction in the sigma(1) receptor-like immunoreactivity of the rat hippocampus, this attenuation was blocked by the treatment with BD 1047, but not by ifenprodil. These results suggest that DM prevents TMT-induced neurotoxicity, at least in part, via sigma(1) receptor stimulation.


Subject(s)
Dextromethorphan/pharmacology , Neurotoxicity Syndromes/prevention & control , Receptors, sigma/drug effects , Trimethyltin Compounds/antagonists & inhibitors , Trimethyltin Compounds/toxicity , Adrenergic alpha-Antagonists/pharmacology , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Ethylenediamines/pharmacology , Immunohistochemistry , Learning Disabilities/chemically induced , Learning Disabilities/prevention & control , Learning Disabilities/psychology , Maze Learning/drug effects , Memory/drug effects , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Piperidines/pharmacology , Radioligand Assay , Rats , Rats, Inbred F344 , Receptors, Phencyclidine/drug effects , Seizures/chemically induced , Seizures/psychology , Sigma-1 Receptor
SELECTION OF CITATIONS
SEARCH DETAIL