Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
1.
Br J Pharmacol ; 176(23): 4574-4591, 2019 12.
Article in English | MEDLINE | ID: mdl-31389613

ABSTRACT

BACKGROUND AND PURPOSE: Accumulating evidence indicates that mitochondrial dynamics play an important role in the progressive deterioration of dopaminergic neurons. Andrographolide has been found to exert neuroprotective effects in several models of neurological diseases. However, the mechanism of how andrographolide protects neurons in Parkinson's disease (PD) remains not fully understood. EXPERIMENTAL APPROACH: Behavioural experiments were performed to examine the effect of andrographolide in 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-PD mice. Mitochondrial mass and morphology were visualized using transmission electron microscopy (TEM). SH-SY5Y cells and primary mouse neurons were exposed to rotenone to mimic PD in vitro. Western blotting, co-immunoprecipitation and immunofluorescence were performed. The target protein of andrographolide was identified by biotin-andrographolide pulldown assay as well as drug affinity responsive target stability (DARTS), cellular thermal shift (CETSA), and surface plasmon resonance (SPR) assays. KEY RESULTS: Andrographolide administration improved behavioural deficits and attenuated loss of dopaminergic neurons in MPTP-exposed mice and reduced cell death induced by rotenone in vitro. An increased mitochondrial mass, and decreased surface area were found in the striatum from MPTP-PD mice, as well as in rotenone-treated primary neurons and SH-SY5Y cells, while andrographolide treatment preserved mitochondrial mass and morphology. Dynamin-related protein 1 (DRP1) was identified as a target protein of andrographolide. Andrographolide bound to DRP1 and inhibited its GTPase activity, thereby preventing excessive mitochondria fission and neuronal damage in PD. CONCLUSIONS AND IMPLICATIONS: Our findings suggest that andrographolide may protect neurons against rotenone- or MPTP-induced damage in vitro and in vivo through inhibiting mitochondrial fission.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Diterpenes/pharmacology , Dynamins/antagonists & inhibitors , Mitochondrial Dynamics/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/administration & dosage , Dynamins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/administration & dosage , Parkinson Disease/metabolism , Surface Plasmon Resonance
2.
PLoS One ; 10(12): e0144848, 2015.
Article in English | MEDLINE | ID: mdl-26658949

ABSTRACT

Neuronal death in Parkinson's disease (PD) is often preceded by axodendritic tree retraction and loss of neuronal functionality. The presence of non-functional but live neurons opens therapeutic possibilities to recover functionality before clinical symptoms develop. Considering that iron accumulation and oxidative damage are conditions commonly found in PD, we tested the possible neuritogenic effects of iron chelators and antioxidant agents. We used three commercial chelators: DFO, deferiprone and 2.2'-dypyridyl, and three 8-hydroxyquinoline-based iron chelators: M30, 7MH and 7DH, and we evaluated their effects in vitro using a mesencephalic cell culture treated with the Parkinsonian toxin MPP+ and in vivo using the MPTP mouse model. All chelators tested promoted the emergence of new tyrosine hydroxylase (TH)-positive processes, increased axodendritic tree length and protected cells against lipoperoxidation. Chelator treatment resulted in the generation of processes containing the presynaptic marker synaptophysin. The antioxidants N-acetylcysteine and dymetylthiourea also enhanced axodendritic tree recovery in vitro, an indication that reducing oxidative tone fosters neuritogenesis in MPP+-damaged neurons. Oral administration to mice of the M30 chelator for 14 days after MPTP treatment resulted in increased TH- and GIRK2-positive nigra cells and nigrostriatal fibers. Our results support a role for oral iron chelators as good candidates for the early treatment of PD, at stages of the disease where there is axodendritic tree retraction without neuronal death.


Subject(s)
Antioxidants/pharmacology , Iron Chelating Agents/pharmacology , MPTP Poisoning/drug therapy , Nerve Fibers/drug effects , Neurites/drug effects , Neuroprotective Agents/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 2,2'-Dipyridyl/pharmacology , Animals , Deferiprone , Deferoxamine/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/biosynthesis , Hydroxyquinolines/pharmacology , Lipid Peroxidation/drug effects , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Mesencephalon/drug effects , Mesencephalon/metabolism , Mesencephalon/pathology , Mice , Mice, Inbred C57BL , Nerve Fibers/metabolism , Nerve Fibers/pathology , Neurites/metabolism , Neurites/pathology , Primary Cell Culture , Pyridones/pharmacology , Rats , Rats, Sprague-Dawley , Synaptophysin/agonists , Synaptophysin/biosynthesis , Tyrosine 3-Monooxygenase/biosynthesis
3.
J Neurochem ; 130(2): 280-90, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24341912

ABSTRACT

Complex pathophysiology of Parkinson's disease involves multiple CNS cell types. Degeneration in spinal cord neurons alongside brain has been shown to be involved in Parkinson's disease and evidenced in experimental parkinsonism. However, the mechanisms of these degenerative pathways are not well understood. To unravel these mechanisms SH-SY5Y neuroblastoma cells were differentiated into dopaminergic and cholinergic phenotypes, respectively, and used as cell culture model following exposure to two parkinsonian neurotoxicants MPP(+) and rotenone. SNJ-1945, a cell-permeable calpain inhibitor was tested for its neuroprotective efficacy. MPP(+) and rotenone dose-dependently elevated the levels of intracellular free Ca(2+) and induced a concomitant rise in the levels of active calpain. SNJ-1945 pre-treatment significantly protected cell viability and preserved cellular morphology following MPP(+) and rotenone exposure. The neurotoxicants elevated the levels of reactive oxygen species more profoundly in SH-SY5Y cells differentiated into dopaminergic phenotype, and this effect could be attenuated with SNJ-1945 pre-treatment. In contrast, significant levels of inflammatory mediators cyclooxygenase-2 (Cox-2 and cleaved p10 fragment of caspase-1) were up-regulated in the cholinergic phenotype, which could be dose-dependently attenuated by the calpain inhibitor. Overall, SNJ-1945 was efficacious against MPP(+) or rotenone-induced reactive oxygen species generation, inflammatory mediators, and proteolysis. A post-treatment regimen of SNJ-1945 was also examined in cells and partial protection was attained with calpain inhibitor administration 1-3 h after exposure to MPP(+) or rotenone. Taken together, these results indicate that calpain inhibition is a valid target for protection against parkinsonian neurotoxicants, and SNJ-1945 is an efficacious calpain inhibitor in this context. SH-SY5Y cells, differentiated as dopaminergic (TH positive) and cholinergic (ChAT positive), were used as in vitro models for Parkinson's disease. MPP+ and rotenone induced up-regulation of calpain, expression, and activity as a common mechanism of neurodegeneration. SNJ-1945, a novel calpain inhibitor, protected both the cell phenotypes against MPP+ and rotenone.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Calpain/antagonists & inhibitors , Carbamates/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Dopamine Agents/toxicity , Neuroprotective Agents , Rotenone/antagonists & inhibitors , Rotenone/toxicity , Uncoupling Agents/antagonists & inhibitors , Uncoupling Agents/toxicity , Blotting, Western , Calcium/metabolism , Calpain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation/drug effects , Fluorescent Antibody Technique , Humans , Inflammation Mediators/metabolism , Reactive Oxygen Species/metabolism
4.
Neuromolecular Med ; 15(2): 238-51, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23292542

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder that is characterized by the degeneration of dopaminergic neurons in the substantia nigra and dopamine depletion in the striatum. Although the motor symptoms are still regarded as the main problem, non-motor symptoms in PD also markedly impair the quality of life. Several non-motor symptoms, such as sleep disturbances and depression, are suggested to be implicated in the alteration in circadian clock function. In this study, we investigated circadian disruption and the mechanism in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. MPTP-treated mice exhibited altered 24-h rhythms in body temperature and locomotor activity. In addition, MPTP treatment also affected the circadian clock system at the genetic level. The exposure of human neuroblastoma cells (SH-SY5Y) to 1-metyl-4-phenylpyridinium (MPP(+)) increased or decreased the mRNA levels of several clock genes in a dose-dependent manner. MPP(+)-induced changes in clock genes expression were reversed by Compound C, an inhibitor of AMP-activated protein kinase (AMPK). Most importantly, addition of ATP to the drinking water of MPTP-treated mice attenuated neurodegeneration in dopaminergic neurons, suppressed AMPK activation and prevented circadian disruption. The present findings suggest that the activation of AMPK caused circadian dysfunction, and ATP may be a novel therapeutic strategy based on the molecular clock in PD.


Subject(s)
Chronobiology Disorders/chemically induced , MPTP Poisoning/physiopathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , AMP-Activated Protein Kinases/antagonists & inhibitors , ARNTL Transcription Factors/biosynthesis , ARNTL Transcription Factors/genetics , Adenosine Triphosphate/therapeutic use , Animals , Body Temperature/drug effects , Body Temperature/physiology , Catalytic Domain/drug effects , Cell Line, Tumor , Chronobiology Disorders/genetics , Cryptochromes/biosynthesis , Cryptochromes/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , MPTP Poisoning/drug therapy , MPTP Poisoning/genetics , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Neostriatum/drug effects , Neostriatum/physiology , Neuroblastoma/pathology , Nuclear Receptor Subfamily 1, Group D, Member 1/biosynthesis , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology
5.
Free Radic Biol Med ; 55: 36-45, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23178948

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by a gradual loss of dopaminergic (DA) neurons in the substantia nigra (SN) of the brain. Ribosomal protein S3 (rpS3) has multiple functions related to protein synthesis, antioxidative activity, and UV endonuclease III activity. We have previously shown that PEP-1-rpS3 inhibits skin inflammation and provides neuroprotection against experimental cerebral ischemic damage. In this study, we examined whether PEP-1-rpS3 can protect DA neurons against oxidative stress in SH-SY5Y neuroblastoma cells and in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. PEP-1-rpS3 was efficiently delivered to SH-SY5Y cells and the SN of the brain as confirmed by Western blot and immunohistochemical analysis. PEP-1-rpS3 significantly inhibited reactive oxygen species generation and DNA fragmentation induced by 1-methyl-4-phenylpyridinium, consequently leading to the survival of SH-SY5Y cells. The neuroprotection was related to the antiapoptotic activity of PEP-1-rpS3 that affected the levels of proapoptotic and antiapoptotic mediators. In addition, immunohistochemical data collected using a tyrosine hydroxylase antibody and cresyl violet staining demonstrated that PEP-1-rpS3 markedly protected DA cells in the SN against MPTP-induced oxidative stress. Therefore, our results suggest that PEP-1-rpS3 may be a potential therapy for PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Dopaminergic Neurons/metabolism , Neuroprotective Agents/metabolism , Parkinson Disease/metabolism , Parkinson Disease/prevention & control , Recombinant Fusion Proteins/metabolism , Ribosomal Proteins/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Parkinson Disease/pathology , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/chemistry , Tumor Cells, Cultured
6.
Brain ; 135(Pt 11): 3336-47, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23169921

ABSTRACT

Parkinson's disease is a neurodegenerative disorder that can, at least partly, be mimicked by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. S100B is a calcium-binding protein expressed in, and secreted by, astrocytes. There is increasing evidence that S100B acts as a cytokine or damage-associated molecular pattern protein not only in inflammatory but also in neurodegenerative diseases. In this study, we show that S100B protein levels were higher in post-mortem substantia nigra of patients with Parkinson's disease compared with control tissue, and cerebrospinal fluid S100B levels were higher in a large cohort of patients with Parkinson's disease compared with controls. Correspondingly, mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine showed upregulated S100B messenger RNA and protein levels. In turn, ablation of S100B resulted in neuroprotection, reduced microgliosis and reduced expression of both the receptor for advanced glycation endproducts and tumour necrosis factor-α. Our results demonstrate a role of S100B in the pathophysiology of Parkinson's disease. Targeting S100B may emerge as a potential treatment strategy in this disorder.


Subject(s)
Gliosis/pathology , Nerve Growth Factors/metabolism , Neuroprotective Agents/metabolism , Parkinson Disease/metabolism , Receptors, Immunologic/metabolism , S100 Proteins/metabolism , Substantia Nigra/metabolism , Tumor Necrosis Factor-alpha/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Aged , Animals , Case-Control Studies , Cell Line , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/genetics , Parkinson Disease/blood , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/genetics , Parkinson Disease/pathology , Receptor for Advanced Glycation End Products , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , Substantia Nigra/pathology , Up-Regulation
7.
Proc Natl Acad Sci U S A ; 109(42): 17010-5, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027934

ABSTRACT

We previously reported the discovery of P7C3, an aminopropyl carbazole having proneurogenic and neuroprotective properties in newborn neural precursor cells of the dentate gyrus. Here, we provide evidence that P7C3 also protects mature neurons in brain regions outside of the hippocampus. P7C3 blocks 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated cell death of dopaminergic neurons in the substantia nigra of adult mice, a model of Parkinson disease (PD). Dose-response studies show that the P7C3 analog P7C3A20 blocks cell death with even greater potency and efficacy, which parallels the relative potency and efficacy of these agents in blocking apoptosis of newborn neural precursor cells of the dentate gyrus. P7C3 and P7C3A20 display similar relative effects in blocking 1-methyl-4-phenylpyridinium (MPP(+))-mediated death of dopaminergic neurons in Caenorhabditis elegans, as well as in preserving C. elegans mobility following MPP(+) exposure. Dimebon, an antihistaminergic drug that is weakly proneurogenic and neuroprotective in the dentate gyrus, confers no protection in either the mouse or the worm models of PD. We further demonstrate that the hippocampal proneurogenic efficacy of eight additional analogs of P7C3 correlates with their protective efficacy in MPTP-mediated neurotoxicity. In vivo screening of P7C3 analogs for proneurogenic efficacy in the hippocampus may thus provide a reliable means of predicting neuroprotective efficacy. We propose that the chemical scaffold represented by P7C3 and P7C3A20 provides a basis for optimizing and advancing pharmacologic agents for the treatment of patients with PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/antagonists & inhibitors , Carbazoles/pharmacology , Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Substantia Nigra/cytology , Animals , Apoptosis/drug effects , Caenorhabditis elegans , Carbazoles/chemical synthesis , Carbazoles/chemistry , Carbazoles/pharmacokinetics , Dose-Response Relationship, Drug , Hippocampus/cytology , Hippocampus/drug effects , Indoles/pharmacokinetics , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Molecular Structure , Substantia Nigra/drug effects
8.
J Enzyme Inhib Med Chem ; 27(6): 810-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-21992679

ABSTRACT

Monoamine oxidase (MAO) enzymes catalyze the oxidative deamination of amines and neurotransmitters and inhibitors of MAO are useful as neuroprotectants. This work evaluates the human MAO-catalyzed oxidation of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a dopaminergic neurotoxin, to the directly-acting neurotoxic metabolites, 1-methyl-4-phenyl-2,3-dihydropyridinium (MPDP(+)) and 1-methyl-4-phenylpyridinium (MPP(+)) measured by High-Performance Liquid Chromatography (HPLC), and this approach is subsequently used as a new method for screening of MAO inhibitors and protective agents. Oxidation of MPTP by human MAO-B was more efficient than by MAO-A. R-Deprenyl, a known neuroprotectant, norharman (ß-carboline), 5-nitroindazole and menadione (vitamin K3) inhibited MAO-B and reduced the formation of toxic pyridinium cations. Clorgyline and the ß-carbolines, harman and norharman, inhibited the oxidation of MPTP by MAO-A. Cigarette smoke, as well as the naturally occurring ß-carbolines (norharman and harman) isolated from smoke and coffee inhibited the oxidation of MPTP by MAO-B and/or MAO-A, suggesting protective effects against MPTP. The results show the suitability of the approach used to search for new MAO inhibitors with eventual neuroprotective activity.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/chemistry , 1-Methyl-4-phenylpyridinium/chemistry , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase/chemistry , Neuroprotective Agents/chemistry , Pyridinium Compounds/chemistry , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Carbolines , Chromatography, High Pressure Liquid , Clorgyline/chemistry , Enzyme Assays , Harmine/analogs & derivatives , Harmine/chemistry , Humans , Indazoles/chemistry , Isoenzymes/antagonists & inhibitors , Oxidation-Reduction , Recombinant Proteins/antagonists & inhibitors , Selegiline/chemistry , Smoke , Nicotiana/chemistry , Vitamin K 3/chemistry
9.
J Immunol ; 187(12): 6508-17, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22079984

ABSTRACT

This study examined whether the cannabinoid receptor type 1 (CB(1)) receptor contributes to the survival of nigrostriatal dopaminergic (DA) neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. MPTP induced significant loss of nigrostriatal DA neurons and microglial activation in the substantia nigra (SN), visualized with tyrosine hydroxylase or macrophage Ag complex-1 immunohistochemistry. Real-time PCR, ELISA, Western blotting, and immunohistochemistry disclosed upregulation of proinflammatory cytokines, activation of microglial NADPH oxidase, and subsequent reactive oxygen species production and oxidative damage of DNA and proteins in MPTP-treated SN, resulting in degeneration of DA neurons. Conversely, treatment with nonselective cannabinoid receptor agonists (WIN55,212-2 and HU210) led to increased survival of DA neurons in the SN, their fibers and dopamine levels in the striatum, and improved motor function. This neuroprotection by cannabinoids was accompanied by suppression of NADPH oxidase reactive oxygen species production and reduced expression of proinflammatory cytokines from activated microglia. Interestingly, cannabinoids protected DA neurons against 1-methyl-4-phenyl-pyridinium neurotoxicity in cocultures of mesencephalic neurons and microglia, but not in neuron-enriched mesencephalic cultures devoid of microglia. The observed neuroprotection and inhibition of microglial activation were reversed upon treatment with CB(1) receptor selective antagonists AM251 and/or SR14,716A, confirming the involvement of the CB(1) receptor. The present in vivo and in vitro findings clearly indicate that the CB(1) receptor possesses anti-inflammatory properties and inhibits microglia-mediated oxidative stress. Our results collectively suggest that the cannabinoid system is beneficial for the treatment of Parkinson's disease and other disorders associated with neuroinflammation and microglia-derived oxidative damage.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Corpus Striatum/immunology , Growth Inhibitors/physiology , Microglia/drug effects , Microglia/immunology , Neurotoxins/adverse effects , Receptor, Cannabinoid, CB1/physiology , Substantia Nigra/immunology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Benzoxazines/pharmacology , Cells, Cultured , Coculture Techniques , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine Agents/administration & dosage , Dopamine Agents/adverse effects , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neurotoxins/administration & dosage , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , Parkinsonian Disorders/prevention & control , Receptor, Cannabinoid, CB1/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
10.
J Immunol ; 187(2): 960-9, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21685323

ABSTRACT

This study examined whether ethyl pyruvate (EP) promotes the survival of nigrostriatal dopaminergic (DA) neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. MPTP induced degeneration of nigrostriatal DA neurons and glial activation as visualized by tyrosine hydroxylase, macrophage Ag complex-1, and/or glial fibrillary acidic protein immunoreactivity. Western blotting and immunohistochemistry showed activation of microglial NADPH oxidase and astroglial myeloperoxidase (MPO) and subsequent reactive oxygen species/reactive nitrogen species production and oxidative DNA damage in the MPTP-treated substantia nigra. Treatment with EP prevented degeneration of nigrostriatal DA neurons, increased striatal dopamine levels, and improved motor function. This neuroprotection afforded by EP was associated with the suppression of astroglial MPO expression, NADPH oxidase-, and/or inducible NO synthase-derived reactive oxygen species/reactive nitrogen species production by activated microglia. Interestingly, EP was found to protect DA neurons from 1-methyl-4-phenyl-pyridinium neurotoxicity in cocultures of mesencephalic neurons and microglia but not in neuron-enriched mesencephalic cultures devoid of microglia. The present findings show that EP may inhibit glial-mediated oxidative stress, suggesting that EP may have therapeutic value in the treatment of aspects of Parkinson's disease related to glia-derived oxidative damage.


Subject(s)
Dopamine/physiology , Neuroglia/immunology , Parkinson Disease/drug therapy , Parkinson Disease/immunology , Pyruvates/therapeutic use , Substantia Nigra/immunology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Cells, Cultured , Coculture Techniques , Corpus Striatum/drug effects , Corpus Striatum/immunology , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/adverse effects , Dopamine Antagonists/administration & dosage , Dopamine Antagonists/therapeutic use , Inflammation Mediators/administration & dosage , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Neuroglia/drug effects , Neuroglia/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidative Stress/immunology , Parkinson Disease/pathology , Pyruvates/administration & dosage , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/pathology
11.
Food Chem Toxicol ; 49(8): 1773-81, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21554916

ABSTRACT

Monoamine oxidase (MAO) enzymes located in human mitochondria oxidize neurotransmitters and bioactivate the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) by oxidation to directly-acting neurotoxic pyridinium cations (MPDP⁺/MPP⁺) that produce Parkinsonism. Antioxidants and MAO inhibitors are useful as neuroprotectants. Naturally-occurring substances, antioxidants and redox agents were assessed as inhibitors of the oxidation (bioactivation) of MPTP by human mitochondria and MAO enzymes. Methylene blue, 5-nitroindazole, norharman (ß-carboline), 9-methylnorharman (9-methyl-ß-carboline) and menadione (vitamin-K analogue) highly inhibited the oxidation of MPTP to the neurotoxic species, MPDP⁺/MPP⁺, in human mitochondria (IC50 of 0.18, 3.1, 9.9, 7.3, and 12.6 µM, respectively). Inhibition by methylene blue was similar to R-deprenyl (IC50 of 0.15 µM), a known neuroprotectant. The naturally-occurring ß-carbolines, harmine, harmaline and tetrahydro-ß-carboline, and the antioxidants, melatonin, resveratrol, quercetin and catechin showed little or no inhibition. Oxidation of MPTP in mitochondria was performed by human MAO-B and the above active compounds were also inhibitors of this isozyme. Norharman and 5-nitroindazole were competitive inhibitors of MAO-B whereas methylene blue inhibited MPTP oxidation (IC50 of 50 nM) under a mixed type and predominantly uncompetitive mechanism. Methylene blue, 5-nitroindazole, norharman, 9-methylnorharman and menadione inhibit MAO-B in mitochondria and afford protective effects, as suggested by a reduced conversion of MPTP to neurotoxic species.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Antioxidants/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Neurotoxins/antagonists & inhibitors , Carbolines/pharmacology , Chromatography, High Pressure Liquid , Harmine/analogs & derivatives , Harmine/pharmacology , Humans , Indazoles/pharmacology , Mass Spectrometry , Methylene Blue/pharmacology , Mitochondria/metabolism , Monoamine Oxidase/adverse effects , Monoamine Oxidase/metabolism , Neuroprotective Agents/pharmacology , Oxidation-Reduction , Selegiline/pharmacology
12.
Amino Acids ; 40(5): 1305-13, 2011 May.
Article in English | MEDLINE | ID: mdl-21448659

ABSTRACT

There is a substantial body of literature, which has demonstrated that creatine has neuroprotective effects both in vitro and in vivo. Creatine can protect against excitotoxicity as well as against ß-amyloid toxicity in vitro. We carried out studies examining the efficacy of creatine as a neuroprotective agent in vivo. We demonstrated that creatine can protect against excitotoxic lesions produced by N-methyl-D: -aspartate. We also showed that creatine is neuroprotective against lesions produced by the toxins malonate and 3-nitropropionic acid (3-NP) which are reversible and irreversible inhibitors of succinate dehydrogenase, respectively. Creatine produced dose-dependent neuroprotective effects against MPTP toxicity reducing the loss of dopamine within the striatum and the loss of dopaminergic neurons in the substantia nigra. We carried out a number of studies of the neuroprotective effects of creatine in transgenic mouse models of neurodegenerative diseases. We demonstrated that creatine produced an extension of survival, improved motor performance, and a reduction in loss of motor neurons in a transgenic mouse model of amyotrophic lateral sclerosis (ALS). Creatine produced an extension of survival, as well as improved motor function, and a reduction in striatal atrophy in the R6/2 and the N-171-82Q transgenic mouse models of Huntington's disease (HD), even when its administration was delayed until the onset of disease symptoms. We recently examined the neuroprotective effects of a combination of coenzyme Q10 (CoQ10) with creatine against both MPTP and 3-NP toxicity. We found that the combination of CoQ and creatine together produced additive neuroprotective effects in a chronic MPTP model, and it blocked the development of alpha-synuclein aggregates. In the 3-NP model of HD, CoQ and creatine produced additive neuroprotective effects against the size of the striatal lesions. In the R6/2 transgenic mouse model of HD, the combination of CoQ and creatine produced additive effects on improving survival. Creatine may stabilize mitochondrial creatine kinase, and prevent activation of the mitochondrial permeability transition. Creatine, however, was still neuroprotective in mice, which were deficient in mitochondrial creatine kinase. Administration of creatine increases the brain levels of creatine and phosphocreatine. Due to its neuroprotective effects, creatine is now in clinical trials for the treatment of Parkinson's disease (PD) and HD. A phase 2 futility trial in PD showed approximately a 50% improvement in Unified Parkinson's Disease Rating Scale at one year, and the compound was judged to be non futile. Creatine is now in a phase III clinical trial being carried out by the NET PD consortium. Creatine reduced plasma levels of 8-hydroxy-2-deoxyguanosine in HD patients phase II trial and was well-tolerated. Creatine is now being studied in a phase III clinical trial in HD, the CREST trial. Creatine, therefore, shows great promise in the treatment of a variety of neurodegenerative diseases.


Subject(s)
Creatine/pharmacology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Creatine/administration & dosage , Humans , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/administration & dosage , Nitro Compounds/antagonists & inhibitors , Propionates/antagonists & inhibitors
13.
Int J Neurosci ; 121(4): 209-17, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21265705

ABSTRACT

AIM: This study was designed to investigate the anti-inflammatory effects of bee venom (BV) in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of Parkinson's disease (PD). METHOD: MPTP was administered by intraperitoneal (IP) injection at 2-hr intervals over an 8-hr period. Mice were then subjected to BV subcutaneous injection and sacrificed on days 1 and 3 following the final MPTP injection. The loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) was assessed by tyrosine hydroxylase (TH) immunohistochemistry. Microglial activation was measured by immunohistochemistry for macrophage antigen complex-1 (MAC-1) and inducible nitric oxide synthase (iNOS). The staining intensities of MAC-1 and iNOS were quantified with respect to optical density. RESULT: In animals treated with MPTP, the survival percentages of TH+ cells in the SNpc were 32% on day 1 and 46% on day 3 compared with normal mice. In BV-treated mice, the survival percentages of TH+ cells improved to 70% on day 1 and 78% on day 3 compared with normal mice. BV treatment also resulted in reduced expression of the inflammation markers MAC-1 and iNOS in the SNpc. CONCLUSION: These data suggest that BV injection may have a neuroprotective effect that attenuates the activation of the microglial response, which has implications for the treatment of PD.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bee Venoms/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Neurons/pathology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Bee Venoms/administration & dosage , Cell Death/drug effects , Cell Death/physiology , Disease Models, Animal , Dopamine Antagonists/administration & dosage , Dopamine Antagonists/therapeutic use , Inflammation Mediators/adverse effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/enzymology , Neuroprotective Agents/therapeutic use , Random Allocation , Substantia Nigra/drug effects , Substantia Nigra/enzymology , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/biosynthesis , Tyrosine 3-Monooxygenase/physiology
14.
Toxicology ; 271(1-2): 5-12, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20064577

ABSTRACT

Dopaminergic neurons are lost mainly through apoptosis in Parkinson's disease. Insulin like growth factor-1 (IGF-1) inhibits apoptosis in a wide variety of tissues. Here we have shown that IGF-1 protects PC12 cells from toxic effects of 1-methyl-4-phenylpyridiniumion (MPP(+)). Treatment of PC12 cells with recombinant human IGF-1 significantly decreased apoptosis caused by MPP(+) as measured by acridine orange/ethidium bromide staining. IGF-1 treatment induced sustained phosphorylation of glycogen synthase kinase-3beta (GSK-3beta) as shown by western blot analysis. The anti-apoptotic effect of IGF-1 was abrogated by LY294002, which indirectly inhibits phosphorylation of GSK-3beta. Lithium chloride (LiCl), a known inhibitor of GSK-3beta, also blocked MPP(+)-induced apoptosis. Finally, although IGF-1 enhanced phosphorylation of extracellular signal-regulated kinases ERK1 and 2 (ERK1/2), PD98059, a specific inhibitor of ERK1/2, did not alter the survival effect of IGF-1. Thus, our findings indicate that IGF-1 protects PC12 cells exposed to MPP(+) from apoptosis via the GSK-3beta signaling pathway.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Apoptosis/drug effects , Glycogen Synthase Kinase 3/metabolism , Insulin-Like Growth Factor I/pharmacology , Neurotoxins/antagonists & inhibitors , Parkinson Disease/pathology , Animals , Apoptosis/physiology , Blotting, Western , Cell Survival , Chromones/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Lithium Chloride/pharmacology , Morpholines/pharmacology , PC12 Cells , Parkinson Disease/enzymology , Parkinson Disease/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Recombinant Proteins/pharmacology
15.
J Neurosci Res ; 88(7): 1522-36, 2010 May 15.
Article in English | MEDLINE | ID: mdl-19998477

ABSTRACT

An excessive expression of poly(ADP-ribose)polymerase (PARP) has been demonstrated to play a key role in the pathogenesis of Parkinson's disease (PD). Here we investigated the therapeutic effect of the PARP inhibitor benzamide against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity in mice. In our HPLC and Western blot analysis, pretreatment with benzamide showed a neuroprotective effect against MPTP neurotoxicity in mice. Posttreatment with benzamide also attenuated MPTP neurotoxicity in mice. Furthermore, our immunohistochemical study showed that posttreatment with benzamide significantly prevented neuronal damage by suppressing overexpression of neuronal, microglial, and astroglial PARP after MPTP treatment. These findings have important implications for the therapeutic time window and choice of PARP inhibitors in PD patients. Our present findings provide further evidence that PARP inhibitor may offer a novel therapeutic strategy for PD.


Subject(s)
Benzamides/pharmacology , Nerve Degeneration/drug therapy , Nerve Degeneration/enzymology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Benzamides/therapeutic use , Brain/drug effects , Brain/enzymology , Brain/physiopathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/physiopathology , Neuroglia/drug effects , Neuroglia/enzymology , Neurons/drug effects , Neurons/enzymology , Neurotoxins/antagonists & inhibitors , Parkinsonian Disorders/physiopathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Treatment Outcome
16.
Cell Calcium ; 46(3): 209-18, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19695701

ABSTRACT

Disturbances in Ca(2+) homeostasis have been implicated in a variety of neuropathological conditions including Parkinson's disease (PD). However, the importance of store-operated Ca(2+) entry (SOCE) channels in PD remains to be investigated. In the present study, we have scrutinized the significance of TRPC1 in 1-methyl-4-phenyl-1,2,3,6-tetrahyrdro-pyridine (MPTP)-induced PD using C57BL/6 animal model and PC12 cell culture model. Both sub-acute and sub-chronic treatments of MPTP significantly reduced TRPC1, and tyrosine hydroxylase levels, but not TRPC3, along with increased neuronal death. Furthermore, MPTP induces mitochondrial dysfunction, which was associated with reduced mitochondrial membrane potential, decreased level of Bcl(2), Bcl-xl, and an altered Bcl-xl/Bax ratio thereby initiating apoptosis. Importantly, TRPC1 overexpression in PC12 cells showed significant protection against MPP(+) induced neuronal apoptosis, which was attributed to the restoration of cytosolic Ca(2+) and preventing loss of mitochondrial membrane potential. Silencing of TRPC1 or addition of TRPC1 channel blockers decreased mitochondrial membrane potential, whereas activation of TRPC1 restored mitochondrial membrane potential in cells overexpressing TRPC1. TRPC1 overexpression also inhibited Bax translocation to the mitochondria and thereby prevented cytochrome c release and mitochondrial-mediated apoptosis. Overall, these results provide compelling evidence for the role of TRPC1 in either onset/progression of PD and restoration of TRPC1 levels could limit neuronal degeneration in MPTP mediated PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Apoptosis , Dopamine Agents/toxicity , TRPC Cation Channels/metabolism , Animals , Calcium/metabolism , Cell Line, Tumor , Male , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Parkinson Disease/metabolism , RNA Interference , Rats
17.
Free Radic Res ; 43(10): 975-84, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19669997

ABSTRACT

Monoamine oxidase (MAO) B is a mitochondrial enzyme selectively involved in the oxidative activation of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxin to toxic pyridinium cations producing Parkinsonism in animal models. Various synthesized 5-nitroindazoles, 6-nitroindazole and the neuroprotectant 7-nitroindazole were examined as inhibitors of MAO and as antioxidants and radical scavengers. The oxidation of MPTP by human MAO-B and mitochondria was assessed by HPLC. Simple nitroindazoles inhibited MPTP oxidation to 1-methyl-4-phenyl-2,3-dihydropyridinium (MPDP(+)) and 1-methyl-4-phenylpyridinium (MPP(+)) in a competitive and reversible manner. 5-Nitroindazole (IC(50)=0.99 microM, K(i)=0.102 microM) and 6-nitroindazole (IC(50)=2.5 microM) were better inhibitors of human MAO-B than 7-nitroindazole (IC(50)=27.8 microM). 6-Nitroindazole also inhibited MAO-A. Nitroindazole isomers were good hydroxyl radical (OH(*)) scavengers, with 5-nitro-, 6-nitro- and 7-nitroindazole showing similar activity (k approximately 10(10) M(-1) s(-1)). Neuroprotective actions of nitroindazoles (7-nitroindazole) could be linked to their MAO-inhibitory and antiradical properties besides inhibition on nitric oxide synthase (NOS). 5-Nitro- and 6-nitroindazole, previously reported as weak NOS inhibitors, were better inhibitors of human MAO-B and more active against MPTP neurotoxin oxidation (lower MPDP(+) and MPP(+) levels) than 7-nitroindazole and acted as good radical scavengers and could be potential neuroprotective agents in addition to MAO-B inhibitors.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Indazoles/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Pyridinium Compounds/metabolism , Free Radical Scavengers/pharmacology , Humans , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Neuroprotective Agents/pharmacology , Oxidation-Reduction/drug effects
18.
FEBS Lett ; 582(6): 956-60, 2008 Mar 19.
Article in English | MEDLINE | ID: mdl-18298955

ABSTRACT

Tyrosine hydroxylase immunohistochemical analysis revealed that in cultured mesencephalic dopaminergic neurons smilagenin (SMI), added prior to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPP+), protected against the drop of neuron number and neurite outgrowth length caused by MPP+. Addition of anti-GDNF and/or anti-GFR alpha 1 functional antibodies to the medium prior to SMI, eliminated mostly, though incompletely, the action of SMI. The expression of glial cell derived neurotrophic factor (GDNF) mRNA, but not GDNF receptor alpha1 (GFR alpha 1) or receptor tyrosine kinase mRNA in MPP+ intoxicated neurons was markedly elevated as early as 2h after the addition of SMI with a peak at 24-48 h. Therefore, an important route of the protective action of SMI on dopaminergic neurons is to stimulate intrinsic GDNF expression.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mesencephalon/drug effects , Neurons/drug effects , Spirostans/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cytoprotection , Dopamine/metabolism , Dopamine Agents/pharmacology , Female , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Mesencephalon/cytology , Mesencephalon/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Tyrosine 3-Monooxygenase/analysis , Tyrosine 3-Monooxygenase/metabolism
19.
J Neural Transm (Vienna) ; 115(6): 831-42, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18235988

ABSTRACT

There is growing evidence indicating that reactive nitrogen species (RNS) and reactive oxygen species (ROS) are a major contributor to the pathogenesis and progression of Parkinson's disease. Here we investigated whether edaravone (free radical scavenger), minocycline (inducible nitric oxide synthase, iNOS inhibitor), 7-nitroindazole (neuronal NOS, nNOS inhibitor), fluvastatin (endothelial NOS, eNOS activator) and pitavastatin (eNOS activator) can protect against MPTP neurotoxicity in mice under the same condition. The present study showed that 7-nitroindazole could protect dose-dependently against the striatal dopamine depletions in mice 5 days after MPTP treatment. In contrast, edaravone, minocycline, fluvastatin and pitavastatin did not show the neuroprotective effect on MPTP-induced striatal dopamine depletion. Our immunohistochemical study showed that TH (tyrosine hydroxylase) and DAT (dopamine transporter) immunoreactivity was decreased significantly in the striatum and substantia nigra 5 days after MPTP treatment. The administration of 7-nitroindazole showed a protective effect against the severe reductions in levels of TH and DAT immunoreactivity in the striatum and substantia nigra 5 days after MPTP treatment. Furthermore, our Western blot analyses study showed the remarkable loss of TH protein levels in the striatum 5 days after MPTP treatment. In contrast, 7-nitroindazole prevented a significant loss in TH protein levels in the striatum 5 days after MPTP treatment. On the other hand, GFAP (glial fibrillary acidic protein) immunoreactivity increased significantly in the striatum and substantia nigra, 5 days after MPTP treatment. 7-Nitroindazole ameliorated severe increases in number of GFAP immunoreactive astrocytes in the striatum and substantia nigra 5 days after MPTP treatment. Furthermore, our Western blot analyses study showed the increase of GFAP protein levels in the striatum 5 days after MPTP treatment. 7-Nitroindazole prevented a significant increase in the GFAP protein levels in the striatum 5 days after MPTP treatment. The present results indicate that 7-nitroindazole can protect dose-dependently against the striatal dopamine depletions in mice 5 days after MPTP treatment. In contrast, edaravone, minocycline, fluvastatin and pitavastatin did not show the neuroprotective effect on MPTP-induced striatal dopamine depletions. These findings demonstrate that the overexpression of nNOS may play a major role in the neurotoxic processes of MPTP, as compared to the production of ROS, the overexpression of iNOS and the modulation of eNOS. Thus, our findings provide strong evidence for neuroprotective properties of nNOS inhibitor in this animal model of Parkinson's disease.


Subject(s)
Indazoles/pharmacology , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress/physiology , Parkinsonian Disorders/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Cytoprotection/drug effects , Disease Models, Animal , Dopamine/deficiency , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Glial Fibrillary Acidic Protein/drug effects , Glial Fibrillary Acidic Protein/metabolism , Gliosis/chemically induced , Gliosis/metabolism , Gliosis/physiopathology , Homovanillic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III , Oxidative Stress/drug effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/physiopathology , Tyrosine 3-Monooxygenase/metabolism
20.
Proc Natl Acad Sci U S A ; 105(5): 1716-21, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18218782

ABSTRACT

PTEN-induced putative kinase 1 (Pink1) is a recently identified gene linked to a recessive form of familial Parkinson's disease (PD). The kinase contains a mitochondrial localization sequence and is reported to reside, at least in part, in mitochondria. However, neither the manner by which the loss of Pink1 contributes to dopamine neuron loss nor its impact on mitochondrial function and relevance to death is clear. Here, we report that depletion of Pink1 by RNAi increased neuronal toxicity induced by MPP(+). Moreover, wild-type Pink1, but not the G309D mutant linked to familial PD or an engineered kinase-dead mutant K219M, protects neurons against MPTP both in vitro and in vivo. Intriguingly, a mutant that contains a deletion of the putative mitochondrial-targeting motif was targeted to the cytoplasm but still provided protection against 1-methyl-4-phenylpyridine (MPP(+))/1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced toxicity. In addition, we also show that endogenous Pink1 is localized to cytosolic as well as mitochondrial fractions. Thus, our findings indicate that Pink1 plays a functional role in the survival of neurons and that cytoplasmic targets, in addition to its other actions in the mitochondria, may be important for this protective effect.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Neurons/enzymology , Neurotoxins/antagonists & inhibitors , Parkinson Disease/enzymology , Protein Kinases/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Survival/genetics , Cytoplasm/enzymology , Dopamine Agents/toxicity , Mice , Mice, Inbred Strains , Mitochondria/enzymology , Mutation , Neurons/drug effects , Neurotoxins/toxicity , Parkinson Disease/genetics , Protein Kinases/analysis , Protein Kinases/genetics , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL
...