Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
2.
Mol Psychiatry ; 28(10): 4474-4484, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37648779

ABSTRACT

Mitochondrial dysfunction has been implicated in Parkinson's Disease (PD) progression; however, the mitochondrial factors underlying the development of PD symptoms remain unclear. One candidate is CR6-interacting factor1 (CRIF1), which controls translation and membrane insertion of 13 mitochondrial proteins involved in oxidative phosphorylation. Here, we found that CRIF1 mRNA and protein expression were significantly reduced in postmortem brains of elderly PD patients compared to normal controls. To evaluate the effect of Crif1 deficiency, we produced mice lacking the Crif1 gene in dopaminergic neurons (DAT-CRIF1-KO mice). From 5 weeks of age, DAT-CRIF1-KO mice began to show decreased dopamine production with progressive neuronal degeneration in the nigral area. At ~10 weeks of age, they developed PD-like behavioral deficits, including gait abnormalities, rigidity, and resting tremor. L-DOPA, a medication used to treat PD, ameliorated these defects at an early stage, although it was ineffective in older mice. Taken together, the observation that CRIF1 expression is reduced in human PD brains and deletion of CRIF1 in dopaminergic neurons leads to early-onset PD with stepwise PD progression support the conclusion that CRIF1-mediated mitochondrial function is important for the survival of dopaminergic neurons.


Subject(s)
Dopaminergic Neurons , Parkinson Disease , Humans , Mice , Animals , Aged , Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , Levodopa/pharmacology , Dopamine/metabolism , Brain/metabolism , Cell Cycle Proteins/genetics
3.
Mol Neurobiol ; 59(11): 6956-6970, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36057709

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the selective loss of dopamine(DA)rgic neurons in the substantia nigra of the midbrain, and primarily causes motor symptoms. While the pathological cause of PD remains uncertain, oxidative damage, neuroinflammation, and energy metabolic perturbation have been implicated. Pyruvate has been shown neuroprotective in animal models for many neurological disorders, presumably owing to its potent anti-oxidative, anti-inflammatory, and energy metabolic properties. We therefore investigated whether exogenous pyruvate could also protect nigral DA neurons from degeneration and reverse the associated motor deficits in an animal model of PD using the DA neuron-specific toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP (20 mg/kg) was injected four times every 2 h into the peritoneum of mice, which resulted in a massive loss of DA neurons as well as an increase in neuronal death and cytosolic labile zinc overload. There were rises in inflammatory and oxidative responses, a drop in the striatal DA level, and the emergence of PD-related motor deficits. In comparison, when sodium pyruvate was administered intraperitoneally at a daily dose of 250 mg/kg for 7 days starting 2 h after the final MPTP treatment, significant relief in the MPTP-induced neuropathology, neurodegeneration, DA depletion, and motor symptoms was observed. Equiosmolar dose of NaCl had no neuroprotective effect, and lower doses of sodium pyruvate did not have any statistically significant effects. These findings suggest that pyruvate has therapeutic potential for the treatment of PD and related neurodegenerative diseases.


Subject(s)
Neuroprotective Agents , Parkinson Disease , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Mice , Mice, Inbred C57BL , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/pathology , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Pyruvic Acid/pharmacology , Pyruvic Acid/therapeutic use , Sodium/metabolism , Sodium Chloride/therapeutic use , Substantia Nigra/pathology , Zinc/metabolism
4.
Mol Cells ; 45(3): 134-147, 2022 Mar 31.
Article in English | MEDLINE | ID: mdl-34887364

ABSTRACT

The anti-oxidant enzyme heme oxygenase-1 (HO-1) is known to exert anti-inflammatory effects. From a library of pyrazolo[3,4-d]pyrimidines, we identified a novel compound KKC080096 that upregulated HO-1 at the mRNA and protein levels in microglial BV-2 cells. KKC080096 exhibited anti-inflammatory effects via suppressing nitric oxide, interleukin-1ß (IL-1ß), and iNOS production in lipopolysaccharide (LPS)-challenged cells. It inhibited the phosphorylation of IKK and MAP kinases (p38, JNK, ERK), which trigger inflammatory signaling, and whose activities are inhibited by HO-1. Further, KKC080096 upregulated anti-inflammatory marker (Arg1, YM1, CD206, IL-10, transforming growth factor-ß [TGF-ß]) expression. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, KKC080096 lowered microglial activation, protected the nigral dopaminergic neurons, and nigral damage-associated motor deficits. Next, we elucidated the mechanisms by which KKC080096 upregulated HO-1. KKC080096 induced the phosphorylation of AMPK and its known upstream kinases LKB1 and CaMKKbeta, and pharmacological inhibition of AMPK activity reduced the effects of KKC080096 on HO-1 expression and LPS-induced NO generation, suggesting that KKC080096-induced HO-1 upregulation involves LKB1/AMPK and CaMKKbeta/AMPK pathway activation. Further, KKC080096 caused an increase in cellular Nrf2 level, bound to Keap1 (Nrf2 inhibitor protein) with high affinity, and blocked Keap1-Nrf2 interaction. This Nrf2 activation resulted in concurrent induction of HO-1 and other Nrf2-targeted antioxidant enzymes in BV-2 and in dopaminergic CATH.a cells. These results indicate that KKC080096 is a potential therapeutic for oxidative stress- and inflammation-related neurodegenerative disorders such as Parkinson's disease.


Subject(s)
Heme Oxygenase-1 , Neuroprotective Agents , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Heme Oxygenase-1/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Lipopolysaccharides/pharmacology , Mice , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Pyrimidines/pharmacology
5.
Exp Neurol ; 332: 113387, 2020 10.
Article in English | MEDLINE | ID: mdl-32580013

ABSTRACT

The transcription factor nuclear factor-erythroid 2-related factor-2 (Nrf2) is known to induce neuroprotective and anti-inflammatory effects and is considered to be an excellent molecular target for drugs related to neurodegenerative disease therapy. Nrf2 activators previously tested in clinical trials were electrophilic, causing adverse effects due to non-selective and covalent modification of cellular thiols. In order to circumvent this issue, we constructed and screened a chemical library consisting of 241 pyrazolo [3,4-d] pyrimidine derivatives and discovered a novel, non-electrophilic compound: 1-benzyl-6-(methylthio)-N-(1-phenylethyl)-1H-pyrazolo[3,4-d]pyrimidine-4-amine (KKC080106). KKC080106 was able to activate Nrf2 signaling as it increases the cellular levels of Nrf2, binds to the Nrf2 inhibitor protein Keap1, and causes the accumulation of nuclear Nrf2. We also observed an increase in the expression levels of Nrf2-dependent genes for antioxidative/neuroprotective enzymes in dopaminergic neuronal cells. In addition, in lipopolysaccharide-activated microglia, KKC080106 suppressed the generation of the proinflammatory markers, such as IL-1ß, TNF-α, cyclooxygenase-2, inducible nitric oxide synthase, and nitric oxide, and inhibited the phosphorylation of kinases known to be involved in inflammatory signaling, such as IκB kinase, p38, JNK, and ERK. As a drug, KKC080106 exhibited excellent stability against plasma enzymes and a good safety profile, evidenced by no mortality after the administration of 2000 mg/kg body weight, and minimal inhibition of the hERG channel activity. Pharmacokinetic analysis revealed that KKC080106 has good bioavailability and enters the brain after oral and intravenous administration, in both rats and mice. In MPTP-treated mice that received KKC080106 orally, the compound blocked microglial activation, protected the nigral dopaminergic neurons from degeneration, and prevented development of the dopamine deficiency-related motor deficits. These results suggest that KKC080106 has therapeutic potential for neurodegenerative disorders such as Parkinson's disease.


Subject(s)
Dopaminergic Neurons/drug effects , NF-E2-Related Factor 2/agonists , Neuroprotective Agents/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sambucus nigra/cytology , Sambucus nigra/drug effects , Animals , Antioxidants , Brain/metabolism , Cytokines/metabolism , Inflammation/genetics , Kelch-Like ECH-Associated Protein 1/drug effects , Kelch-Like ECH-Associated Protein 1/genetics , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neuroprotective Agents/pharmacokinetics , Nitric Oxide/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism , Signal Transduction/drug effects
6.
Curr Biol ; 30(2): 276-291.e9, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31928877

ABSTRACT

Current pharmacological treatments for Parkinson's disease (PD) are focused on symptomatic relief, but not on disease modification, based on the strong belief that PD is caused by irreversible dopaminergic neuronal death. Thus, the concept of the presence of dormant dopaminergic neurons and its possibility as the disease-modifying therapeutic target against PD have not been explored. Here we show that optogenetic activation of substantia nigra pars compacta (SNpc) neurons alleviates parkinsonism in acute PD animal models by recovering tyrosine hydroxylase (TH) from the TH-negative dormant dopaminergic neurons, some of which still express DOPA decarboxylase (DDC). The TH loss depends on reduced dopaminergic neuronal firing under aberrant tonic inhibition, which is attributed to excessive astrocytic GABA. Blocking the astrocytic GABA synthesis recapitulates the therapeutic effect of optogenetic activation. Consistently, SNpc of postmortem PD patients shows a significant population of TH-negative/DDC-positive dormant neurons surrounded by numerous GABA-positive astrocytes. We propose that disinhibiting dormant dopaminergic neurons by blocking excessive astrocytic GABA could be an effective therapeutic strategy against PD.


Subject(s)
Astrocytes/metabolism , Dopaminergic Neurons/physiology , Nerve Degeneration/physiopathology , Parkinson Disease/physiopathology , Tyrosine 3-Monooxygenase/metabolism , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Down-Regulation , Female , Humans , Immobility Response, Tonic/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Middle Aged , Parkinson Disease/therapy , Rats , Rats, Wistar , Tyrosine 3-Monooxygenase/antagonists & inhibitors , gamma-Aminobutyric Acid/biosynthesis
7.
Mol Cells ; 42(10): 702-710, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31656063

ABSTRACT

Neuroinflammation is an important contributor to the pathogenesis of neurodegenerative disorders including Parkinson's disease (PD). We previously reported that our novel synthetic compound KMS99220 has a good pharmacokinetic profile, enters the brain, exerts neuroprotective effect, and inhibits NFκB activation. To further assess the utility of KMS99220 as a potential therapeutic agent for PD, we tested whether KMS99220 exerts an anti-inflammatory effect in vivo and examined the molecular mechanism mediating this phenomenon. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, oral administration of KMS99220 attenuated microglial activation and decreased the levels of inducible nitric oxide synthase and interleukin 1 beta (IL-1b) in the nigrostriatal system. In lipopolysaccharide (LPS)-challenged BV-2 microglial cells, KMS99220 suppressed the production and expression of IL-1b. In the activated microglia, KMS99220 reduced the phosphorylation of IκB kinase, c-Jun N-terminal kinase, and p38 MAP kinase; this effect was mediated by heme oxygenase-1 (HO-1), as both gene silencing and pharmacological inhibition of HO-1 abolished the effect of KMS99220. KMS99220 induced nuclear translocation of the transcription factor Nrf2 and expression of the Nrf2 target genes including HO-1. Together with our earlier findings, our current results show that KMS99220 may be a potential therapeutic agent for neuroinflammation-related neurodegenerative diseases such as PD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chalcones/pharmacology , Heme Oxygenase-1/metabolism , I-kappa B Kinase/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , NF-E2-Related Factor 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Models, Biological , Signal Transduction/drug effects
8.
Exp Neurobiol ; 27(5): 408-418, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30429650

ABSTRACT

We have previously reported a novel synthetic compound KMS99220 that prevented degeneration of the nigral dopaminergic neurons and the associated motor deficits, suggesting a neuroprotective therapeutic utility for Parkinson's disease. Microglia are closely associated with neuroinflammation, which plays a key role in the pathogenesis of neurodegenerative diseases. In this study, we investigated the effects of KMS99220 on the signaling involving AMP-activated protein kinase (AMPK) and heme oxygenase-1 (HO-1), the enzymes thought to regulate inflammation. KMS99220 was shown to elevate the enzyme activity of purified AMPK, and phosphorylation of cellular AMPK in BV2 microglia. It increased the level of HO-1, and this was attenuated by AMPK inhibitors. KMS99220 lowered phosphorylation of IκB, nuclear translocation of NFκB, induction of inducible nitric oxide synthase, and generation of nitric oxide in BV2 cells that had been challenged with lipopolysaccharide. This anti-inflammatory response involved HO-1, because both its pharmacological inhibition and knockdown of its expression abolished the response. The AMPK inhibitors also reversed the anti-inflammatory effects of KMS99220. The induction of HO-1 by KMS99220 occurred within 1 h, and this appeared not to involve the transcription factor Nrf2, because Nrf2 knockdown did not affect the compound's HO-1 inducing- and anti-inflammatory effects in this time window. These findings indicated that KMS99220 leads to AMPK-induced HO-1 expression in microglia, which in turn plays an important role in early anti-inflammatory signaling. Together with its neuroprotective property, KMS99220 may serve as a feasible therapeutic agent against neuroinflammation and neurodegeneration.

9.
Neurochem Int ; 112: 96-107, 2018 01.
Article in English | MEDLINE | ID: mdl-29158022

ABSTRACT

The transcription factor Nrf2 is known to induce gene expression of antioxidant enzymes and proteasome subunits. Because both oxidative stress and protein aggregation have damaging effects on neurons, activation of the Nrf2 signaling should be beneficial against neurodegeneration. In this study, we report a novel synthetic morpholine-containing chalcone KMS99220 that confers neuroprotection. It showed high binding affinity to the Nrf2 inhibitory protein Keap-1 and increased nuclear translocation of Nrf2 and gene expression of the antioxidant enzymes heme oxygenase-1, NAD(P)H:quinone oxidoreductase-1, and the catalytic and modifier subunits of glutamate-cysteine ligase in dopaminergic CATH.a cells. KMS99220 also increased expression of the proteasome subunits PSMB5, PSMB7, PSMB8 and PSMA1, and the respective chymotrypsin and trypsin-like proteasomal enzyme activities, and reduced α-synuclein aggregate in GFP-α-syn A53T-overexpressing cells. KMS99220 exhibited a favorable pharmacokinetic profile with excellent bioavailability and metabolic stability, did not interfere with activities of the cytochrome p450 isotypes, and showed no apparent in vivo toxicity when administered up to 2000 mg/kg. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, oral administration of KMS99220 prevented degeneration of the nigral dopaminergic neurons, induced the Nrf2 target genes, and effectively prevented the associated motor deficits. These results suggest KMS99220 as a potential candidate for therapy against Parkinson's disease.


Subject(s)
Dopaminergic Neurons/metabolism , Morpholines/pharmacology , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Substantia Nigra/metabolism , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , MPTP Poisoning/prevention & control , Male , Mice , Mice, Inbred C57BL , Morpholines/chemistry , Morpholines/therapeutic use , Neuroprotection/drug effects , Neuroprotection/physiology , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Substantia Nigra/drug effects
10.
Sci Rep ; 7(1): 11368, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900300

ABSTRACT

Kohlschutter-Tönz syndrome (KTS) is a rare genetic disorder with neurological dysfunctions including seizure and intellectual impairment. Mutations at the Rogdi locus have been linked to development of KTS, yet the underlying mechanisms remain elusive. Here we demonstrate that a Drosophila homolog of Rogdi acts as a novel sleep-promoting factor by supporting a specific subset of gamma-aminobutyric acid (GABA) transmission. Rogdi mutant flies displayed insomnia-like behaviors accompanied by sleep fragmentation and delay in sleep initiation. The sleep suppression phenotypes were rescued by sustaining GABAergic transmission primarily via metabotropic GABA receptors or by blocking wake-promoting dopaminergic pathways. Transgenic rescue further mapped GABAergic neurons as a cell-autonomous locus important for Rogdi-dependent sleep, implying metabotropic GABA transmission upstream of the dopaminergic inhibition of sleep. Consistently, an agonist specific to metabotropic but not ionotropic GABA receptors titrated the wake-promoting effects of dopaminergic neuron excitation. Taken together, these data provide the first genetic evidence that implicates Rogdi in sleep regulation via GABAergic control of dopaminergic signaling. Given the strong relevance of GABA to epilepsy, we propose that similar mechanisms might underlie the neural pathogenesis of Rogdi-associated KTS.


Subject(s)
Dopamine/metabolism , Drosophila/physiology , Nuclear Proteins/genetics , Signal Transduction , Sleep/genetics , Wakefulness/genetics , gamma-Aminobutyric Acid/metabolism , Alleles , Animals , Animals, Genetically Modified , Anticonvulsants/pharmacology , Brain/drug effects , Brain/metabolism , Circadian Rhythm/genetics , Female , GABAergic Neurons/metabolism , Loss of Function Mutation , Models, Biological , Mutation , Nuclear Proteins/metabolism , Receptors, GABA/metabolism , Signal Transduction/drug effects
11.
Free Radic Res ; 51(6): 633-645, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28770670

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterised by selective degeneration of the nigral dopaminergic neurons, and neuroinflammation and oxidative stress are believed to be involved in its pathogenesis. In the present study, we provide data that the synthetic steroid exemestane, which is currently being used to treat breast cancer, may be useful for PD therapy. In BV-2 microglial cells, exemestane activated the transcription factor Nrf2 and induced expression of the Nrf2-dependent genes that encode the antioxidant enzymes NAD(P)H: quinone oxidoreductase 1, haem oxygenase-1, and glutamylcysteine ligase. It also downregulated gene expression of inducible nitric oxide (NO) synthase, lowered the levels of NO and reactive oxygen species, interleukin-1ß and tumour necrosis factor-α in lipopolysaccharide-activated microglial cells. In CATH.a dopaminergic neuronal cells, exemestane also induced the same set of Nrf2-dependent antioxidant enzyme genes and provided neuroprotection against oxidative damage. In vivo, the drug protected the nigral dopaminergic neurons, decreased microglial activation, and prevented motor deficits in C57Bl/6 male mice that had been administered with the dopaminergic neurotoxin MPTP. Taken together, the results suggested a utility of repositioning exemestane towards disease-modifying therapy for PD.


Subject(s)
Androstadienes/pharmacology , Antiparkinson Agents/pharmacology , Dopaminergic Neurons/drug effects , Drug Repositioning , NF-E2-Related Factor 2/genetics , Parkinsonian Disorders/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antineoplastic Agents/pharmacology , Cell Line , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Gene Expression Regulation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hindlimb Suspension , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides , Male , Membrane Proteins/agonists , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Rotarod Performance Test , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Neurobiol Aging ; 50: 119-133, 2017 02.
Article in English | MEDLINE | ID: mdl-27960106

ABSTRACT

Fungicide exposure causes degeneration of dopaminergic neurons and contributes to Parkinson's disease (PD). Benomyl inhibits enzymes responsible for detoxifying the reactive dopamine metabolite 3,4-dihydroxyphenylacetaldehyde. Aldose reductase (AR) is known as tetrahydrobiopterin (BH4) reductase that generates BH4, a cofactor for tyrosine hydroxylase (TH) involved in dopamine synthesis. AR also acts as an aldehyde reductase involved in detoxifying 3,4-dihydroxyphenylacetaldehyde. In PD patients, the level of AR is significantly lower in the cerebellum. To determine if AR deficiency contributes to PD, AR wild-type (AR+/+) and knockout (AR-/-) mice were administrated with 1-methyl-4-phenyl -1,2,3,6- tetrahydropyridine (MPTP). The MPTP-treated AR-/- mice showed more severe behavioral deficits and brain damage than that of AR+/+ mice. Contrary to expectation, under normal or MPTP-treated condition, AR-/- mice showed a significant elevation of BH4 and dopamine in the midbrain, suggesting that either AR does not contribute to BH4 production, or other BH4 synthetic pathways are induced. The AR-/- brain showed upregulation of peroxynitrite, inducible nitric oxide synthase and downregulation of antioxidant enzymes, Cu/Zn superoxide dismutase (SOD) and peroxiredoxin 2 (Prx2), which indicate an increase in oxidative stress. In line with the animal data, pretreating the SH-SY5Y cells with AR inhibitors (Fidarestat or Epalrestat) before MPP+ treatment, increased severe cell death and mitochondrial fragmentation with downregulation of SOD were observed when compared to the MPP+ treatment alone. Cycloxygenase 2 (COX2), which can lead to the oxidation of dopamine, was upregulated in AR-/- brains. Autophagic proteins, beclin-1 and LC3B were also downregulated. The loss of dopaminergic neurons was associated with activation of p-ERK1/2. These findings suggest that AR plays an important role in protecting dopaminergic neuron against neurotoxic metabolites in PD.


Subject(s)
Aldehyde Reductase/deficiency , Autophagy , Dopaminergic Neurons/pathology , Oxidative Stress/physiology , Parkinson Disease/etiology , Parkinson Disease/pathology , Animals , Cells, Cultured , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Transgenic , Parkinson Disease/physiopathology
13.
Free Radic Res ; 50(11): 1188-1199, 2016.
Article in English | MEDLINE | ID: mdl-27598306

ABSTRACT

The degenerative process of the nigral dopamine(DA)rgic neurons in Parkinson's disease (PD) involves both oxidative stress and neuroinflammation. In the present study, we aimed at developing a novel antioxidant and anti-inflammatory agent for PD therapy. Toward this end, we screened a novel focused library of isothiocyanate derivatives that we have generated for an anti-inflammatory property. We obtained a novel compound ITC-57 and found that ITC-57 effectively induced gene expression of the antioxidant enzymes NAD(P)H quinone oxidoreductase-1, the catalytic and modulatory subunits of glutamylcysteine ligase, and HO-1 in DAergic neuronal CATH.a cells and protected CATH.a cells from oxidative damages. The compound also induced the same antioxidant enzymes in microglial BV-2 cells and suppressed the production of the proinflammatory molecules nitric oxide, interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) in lipopolysaccharide-activated BV-2 cells. In the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-elicited mouse model of PD, ITC-57 protected the DAergic neurons from degeneration, induced HO-1, lowered TNF-α, and suppressed microglial activation in the nigra. Furthermore, ITC-57 prevented the PD-associated motor deficits from occurring. Taken together, ITC-57 would be useful toward development of a disease-modifying therapy for PD.


Subject(s)
Isothiocyanates/pharmacology , Parkinson Disease/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cell Death , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Oxidative Stress
14.
Stereotact Funct Neurosurg ; 94(1): 41-53, 2016.
Article in English | MEDLINE | ID: mdl-26962855

ABSTRACT

BACKGROUND: The inhibition of neuronal activity by electrical deep brain stimulation is one of the mechanisms explaining the amelioration of levodopa-induced dyskinesia. However, electrical deep brain stimulation cannot specifically activate or inactivate selected types of neurons. OBJECTIVES: We applied optogenetics as an alternative treatment to deep brain stimulation for levodopa-induced dyskinesia, and also to confirm that the mechanism of levodopa-induced dyskinesia amelioration by subthalamic nucleus deep brain stimulation is mediated through neuronal inhibition. METHODS: 6-hydroxydopamine-induced hemiparkinsonian rats received injections of hSynapsin1-NpHR-YFP adeno-associated virus (AAV) or hSynapsin1-YFP AAV. Two weeks after viral injections, all rats were treated with daily injections of levodopa. Then, the optic fiber was implanted into the ipsilateral subthalamic nucleus. We performed various behavioral tests to evaluate the changes in levodopa-induced dyskinesias after optogenetic expression and illumination in the subthalamic nucleus. RESULTS: The behavioral tests revealed that optical inhibition of the subthalamic nucleus significantly ameliorated levodopa-induced dyskinesia by reducing the duration of the dyskinesias as well as the severity of axial dyskinesia. CONCLUSIONS: These findings will provide a useful foundation for the future development of optogenetic modulation systems that could be considered as an approach to dyskinesia therapy.


Subject(s)
Dyskinesia, Drug-Induced/therapy , Optogenetics , Parkinson Disease/therapy , Subthalamic Nucleus/physiopathology , Animals , Disease Models, Animal , Dyskinesia, Drug-Induced/physiopathology , Levodopa , Parkinson Disease/physiopathology , Rats
15.
Eur J Pharmacol ; 771: 152-61, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26687634

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by degeneration of dopamine(DA)ergic neurons. Neuroinflammation caused by microglial activation is believed to be involved in the pathogenesis of neurodegenerative diseases including PD. In the present study, we tested the effects of a novel compound 2-acetyl-7-hydroxy-6-methoxy-1-methyl-1,2,3,4,-tetarhydroisoquinoline (AMTIQ) on neuroinflammatory response and DAergic neurodegeneration. In lipopolysaccharide-activated BV-2 microglial cells, AMTIQ lowered nitric oxide and tetrahydrobiopterin levels and downregulated gene expression of inducible nitric oxide synthase and GTP cyclohydrolase I. AMTIQ also repressed gene expression of the proinflammatory cytokines IL-1ß and TNF-α, and attenuated nuclear translocation of NF-κB. AMTIQ was stable against liver microsomal enzymes from human and mouse and did not interfere with activities of the cytochrome p450 enzymes 1A2, 2D6, 2C9, 2C19 and 3A4. Pharmacokinetic studies revealed the brain to plasma ratio of AMTIQ to be 45%, suggesting it can penetrate the blood brain barrier. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mouse PD model, AMTIQ led to decreased microglial activation, increased survival of DAergic neurons and their fibers, and improved behavioral scores on rotarod and vertical grid tests. Taken together, these results suggest that AMTIQ might serve as a candidate preventive-therapeutic agent for neurodegenerative diseases such as PD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antiparkinson Agents/pharmacology , Dopaminergic Neurons/drug effects , Isoquinolines/pharmacology , Neuroprotective Agents/pharmacology , Substantia Nigra/cytology , Substantia Nigra/drug effects , Animals , Anti-Inflammatory Agents/pharmacokinetics , Antiparkinson Agents/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Line , Cytokines/biosynthesis , Cytokines/genetics , Humans , In Vitro Techniques , Isoquinolines/pharmacokinetics , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Microglia/drug effects , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Neuroprotective Agents/pharmacokinetics , Nitric Oxide/metabolism
16.
Neurotox Res ; 28(4): 332-45, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26233727

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder accompanied by a selective loss of the dopamine(DA)ergic neurons residing in the substantia nigra. There is ample evidence that neuroinflammation and oxidative stress are involved in the pathogenesis of PD. In the present study, we aimed at protecting the DAergic neurons by suppressing these cellular events and generated a novel synthetic isothiocyanate ITC-3. The compound led to elevation of nuclear and total levels of the transcription factor Nrf2 and interacted with its binding protein Keap1 with high affinity, suggesting Nrf2 activation. ITC-3 was able to suppress production of the proinflammatory mediators in lipopolysaccharide-activated BV-2 microglial cells. It also increased mRNA and protein levels of the Nrf2-dependent antioxidant enzymes NAD(P)H quinone oxidoreductase, heme oxygenase-1, and glutamylcysteine ligase in both BV-2 and DAergic neuronal CATH.a cells. The compound protected the DAergic cells against oxidative stress. In vivo, ITC-3 attenuated the loss of tyrosine hydroxylase-immunopositive nigrostriatal DAergic neurons, suppressed microglial activation, and abolished PD-associated motor deficits in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-elicited animal model of PD. Taken together, ITC-3 may be useful toward development of neuroprotective therapy for PD.


Subject(s)
Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Isothiocyanates/pharmacology , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Parkinson Disease/metabolism , Animals , Antioxidants/metabolism , Cells, Cultured , Disease Models, Animal , Heme Oxygenase-1/metabolism , Inflammation Mediators/metabolism , Isothiocyanates/administration & dosage , Isothiocyanates/chemical synthesis , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemical synthesis , Nuclear Proteins/metabolism , Parkinson Disease/prevention & control , Signal Transduction/drug effects
17.
J Mol Neurosci ; 56(2): 263-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25702135

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with a selective loss of the neurons containing dopamine (DA) in the substantia nigra pars compacta. Lines of evidence suggest that oxidative stress is a major factor contributing to the vulnerability of DA cells and that the enzyme NAD(P)H quinone oxidoreductase (NQO1) provides protection in these cells. In the present study, we report the synthesis of a novel compound KMS04014 and show that it induces NQO1 gene expression and protects DAergic neuronal cells in both cell culture and animal models of PD. In vitro, KMS04014 increased both mRNA and protein levels of NQO1 and induced nuclear translocation of Nrf2 in the DAergic neuronal cell line CATH.a. It also protected the cells against oxidative stress generated by tetrahydrobiopterin, 1-methyl-4-phenylpyridinium (MPP(+)), and H2O2. In vivo, KMS04014 attenuated the loss of tyrosine hydroxylase-immunopositive DAergic neurons in the substantia nigra and reduced degeneration of the nigral neurons and striatal fibers in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice, an animal model of PD. Taken together, KMS04014 may be utilized toward development of neuroprotective therapy for PD.


Subject(s)
Antioxidants/therapeutic use , Coumaric Acids/pharmacology , MPTP Poisoning/drug therapy , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroprotective Agents/therapeutic use , Animals , Antioxidants/chemical synthesis , Antioxidants/pharmacology , Cell Line, Tumor , Coumaric Acids/chemical synthesis , Coumaric Acids/therapeutic use , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Male , Mice , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism
18.
Br J Pharmacol ; 172(4): 1087-100, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25297649

ABSTRACT

BACKGROUND AND PURPOSE: Neuroinflammation through microglial activation is involved in the pathogenesis of neurodegenerative diseases including Parkinson's disease (PD), a major neurodegenerative disorder characterized by dopaminergic neuronal death in the substantia nigra. We examined our novel synthetic compound VSC2 for its anti-inflammatory properties towards development of a PD therapy. EXPERIMENTAL APPROACH: We tested the effects of VSC2 on production of various NF-κB-dependent proinflammatory molecules and Nrf2-dependent antioxidant enzymes in BV-2 microglia and in vivo. KEY RESULTS: The vinyl sulfone compound, VSC2, most effectively suppressed the production of NO in LPS-activated microglia. It also down-regulated expression of inducible NOS (iNOS), COX-2, IL-1ß and TNF-α and inhibited nuclear translocalization and transcriptional activity of NF-κB. VSC2 increased total and nuclear Nrf2 levels, induced Nrf2 transcriptional activity and was bound to Keap1 with high affinity. Expression of the Nrf2-regulated antioxidant enzyme genes NAD(P)H quinone oxidoreducase-1 (NQO-1), haem oxygenase-1 (HO-1) and glutamylcysteine ligase (GCL) were up-regulated by VSC2. In the MPTP mouse model of PD, oral administration of VSC2 decreased the number of activated microglia in the substantia nigra, lowered the levels of iNOS, COX-2 and IL-1ß, and protected the dopaminergic neurons. VSC2 also elevated the levels of NQO1, HO-1, GCL and Nrf2 in the nigrostriatal area. CONCLUSIONS AND IMPLICATIONS: VSC2 has both anti-inflammatory and antioxidant properties and prevented neuroinflammation in microglia and in an animal model of PD. This suggests VSC2 as a potential candidate for PD therapy.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Microglia/drug effects , Parkinson Disease/drug therapy , Sulfones , Vinyl Compounds , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Adaptor Proteins, Signal Transducing/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cell Line , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kelch-Like ECH-Associated Protein 1 , Male , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Parkinson Disease/pathology , Sulfones/pharmacology , Sulfones/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Vinyl Compounds/pharmacology , Vinyl Compounds/therapeutic use
19.
J Korean Neurosurg Soc ; 56(5): 383-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25535514

ABSTRACT

OBJECTIVE: Neural tissue transplantation has been a promising strategy for the treatment of Parkinson's disease (PD). However, transplantation has the disadvantages of low-cell survival and/or development of dyskinesia. Transplantation of cell aggregates has the potential to overcome these problems, because the cells can extend their axons into the host brain and establish synaptic connections with host neurons. In this present study, aggregates of human brain-derived neural stem cells (HB-NSC) were transplanted into a PD animal model and compared to previous report on transplantation of single-cell suspensions. METHODS: Rats received an injection of 6-OHDA into the right medial forebrain bundle to generate the PD model and followed by injections of PBS only, or HB-NSC aggregates in PBS into the ipsilateral striatum. Behavioral tests, multitracer (2-deoxy-2-[(18)F]-fluoro-D-glucose ([(18)F]-FDG) and [(18)F]-N-(3-fluoropropyl)-2-carbomethoxy-3-(4-iodophenyl)nortropane ([(18)F]-FP-CIT) microPET scans, as well as immunohistochemical (IHC) and immunofluorescent (IF) staining were conducted to evaluate the results. RESULTS: The stepping test showed significant improvement of contralateral forelimb control in the HB-NSC group from 6-10 weeks compared to the control group (p<0.05). [(18)F]-FP-CIT microPET at 10 weeks posttransplantation demonstrated a significant increase in uptake in the HB-NSC group compared to pretransplantation (p<0.05). In IHC and IF staining, tyrosine hydroxylase and human ß2 microglobulin (a human cell marker) positive cells were visualized at the transplant site. CONCLUSION: These results suggest that the HB-NSC aggregates can survive in the striatum and exert therapeutic effects in a PD model by secreting dopamine.

20.
Biochem Biophys Res Commun ; 453(3): 563-8, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25285627

ABSTRACT

We have previously demonstrated that matrix metalloprotease-3 (MMP-3) can act inside the cell to trigger apoptosis in response to various cell stresses in dopaminergic neuronal cells. However, the mechanism by which MMP-3 activity leads to caspase-3 activation in apoptotic signaling was not known. In the present study, we found that MMP-3 acts upstream of caspase-9. Overexpression of wild type MMP-3, but not mutant MMP-3, generated the enzymatically active 35kD caspase-9. The caspase-9 activation was absent in MMP-3 knockout cells, but was present when these cells were transfected with wild type MMP-3 cDNA. It was elevated in cells that were under a MMP-3-inducing ER stress condition, and this was attenuated by pharmacologic inhibition and gene knockdown of MMP-3. Incubation of recombinant catalytic domain of MMP-3 (cMMP-3) with procaspase-9 was not sufficient to cause caspase-9 activation, and an additional cytosolic factor was required. cMMP-3 was found to bind to the cytosolic protein Apaf-1, as determined by changes in surface plasmon resonance, and to cleave Apaf-1. Pharmacological inhibition, knockout, and knockdown of MMP-3 attenuated the cleavage. Taken together, the present study demonstrates that MMP-3 leads to caspase-9 activation and suggests that this occurs indirectly via a cytosolic protein, possibly involving Apaf-1.


Subject(s)
Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/metabolism , Matrix Metalloproteinase 3/metabolism , Animals , Apoptosis , Endoplasmic Reticulum/metabolism , Enzyme Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Proteolysis , Signal Transduction , Stress, Physiological , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL