Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
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
2.
Exp Mol Med ; 49(3): e298, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28255166

ABSTRACT

The effects of capsaicin (CAP), a transient receptor potential vanilloid subtype 1 (TRPV1) agonist, were determined on nigrostriatal dopamine (DA) neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD). The results showed that TRPV1 activation by CAP rescued nigrostriatal DA neurons, enhanced striatal DA functions and improved behavioral recovery in MPTP-treated mice. CAP neuroprotection was associated with reduced expression of proinflammatory cytokines (tumor necrosis factor-α and interleukin-1ß) and reactive oxygen species/reactive nitrogen species from activated microglia-derived NADPH oxidase, inducible nitric oxide synthase or reactive astrocyte-derived myeloidperoxidase. These beneficial effects of CAP were reversed by treatment with the TRPV1 antagonists capsazepine and iodo-resiniferatoxin, indicating TRPV1 involvement. This study demonstrates that TRPV1 activation by CAP protects nigrostriatal DA neurons via inhibition of glial activation-mediated oxidative stress and neuroinflammation in the MPTP mouse model of PD. These results suggest that CAP and its analogs may be beneficial therapeutic agents for the treatment of PD and other neurodegenerative disorders that are associated with neuroinflammation and glial activation-derived oxidative damage.


Subject(s)
Antioxidants/pharmacology , Capsaicin/pharmacology , Dopaminergic Neurons/drug effects , MPTP Poisoning/metabolism , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress , Animals , Antioxidants/therapeutic use , Capsaicin/therapeutic use , Dopaminergic Neurons/metabolism , MPTP Poisoning/drug therapy , Male , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Neuroglia/metabolism , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism , TRPV Cation Channels/metabolism
3.
Exp Mol Med ; 48(1): e205, 2016 01 22.
Article in English | MEDLINE | ID: mdl-27534533

ABSTRACT

The cannabinoid (CB2) receptor type 2 has been proposed to prevent the degeneration of dopamine neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. However, the mechanisms underlying CB2 receptor-mediated neuroprotection in MPTP mice have not been elucidated. The mechanisms underlying CB2 receptor-mediated neuroprotection of dopamine neurons in the substantia nigra (SN) were evaluated in the MPTP mouse model of Parkinson's disease (PD) by immunohistochemical staining (tyrosine hydroxylase, macrophage Ag complex-1, glial fibrillary acidic protein, myeloperoxidase (MPO), and CD3 and CD68), real-time PCR and a fluorescein isothiocyanate-labeled albumin assay. Treatment with the selective CB2 receptor agonist JWH-133 (10 µg kg(-1), intraperitoneal (i.p.)) prevented MPTP-induced degeneration of dopamine neurons in the SN and of their fibers in the striatum. This JWH-133-mediated neuroprotection was associated with the suppression of blood-brain barrier (BBB) damage, astroglial MPO expression, infiltration of peripheral immune cells and production of inducible nitric oxide synthase, proinflammatory cytokines and chemokines by activated microglia. The effects of JWH-133 were mimicked by the non-selective cannabinoid receptor WIN55,212 (10 µg kg(-1), i.p.). The observed neuroprotection and inhibition of glial-mediated neurotoxic events were reversed upon treatment with the selective CB2 receptor antagonist AM630, confirming the involvement of the CB2 receptor. Our results suggest that targeting the cannabinoid system may be beneficial for the treatment of neurodegenerative diseases, such as PD, that are associated with glial activation, BBB disruption and peripheral immune cell infiltration.


Subject(s)
Blood-Brain Barrier/pathology , Dopaminergic Neurons/pathology , Parkinson Disease, Secondary/pathology , Receptor, Cannabinoid, CB2/immunology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Blood-Brain Barrier/immunology , Cytokines/analysis , Cytokines/immunology , Disease Models, Animal , Dopaminergic Neurons/immunology , Glial Fibrillary Acidic Protein/analysis , Glial Fibrillary Acidic Protein/immunology , Macrophages/immunology , Macrophages/pathology , Male , Mice, Inbred C57BL , Neuroprotection , Parkinson Disease, Secondary/immunology , Substantia Nigra/immunology , Substantia Nigra/pathology
4.
Brain ; 138(Pt 12): 3610-22, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26490328

ABSTRACT

Currently there is no neuroprotective or neurorestorative therapy for Parkinson's disease. Here we report that transient receptor potential vanilloid 1 (TRPV1) on astrocytes mediates endogenous production of ciliary neurotrophic factor (CNTF), which prevents the active degeneration of dopamine neurons and leads to behavioural recovery through CNTF receptor alpha (CNTFRα) on nigral dopamine neurons in both the MPP(+)-lesioned or adeno-associated virus α-synuclein rat models of Parkinson's disease. Western blot and immunohistochemical analysis of human post-mortem substantia nigra from Parkinson's disease suggests that this endogenous neuroprotective system (TRPV1 and CNTF on astrocytes, and CNTFRα on dopamine neurons) might have relevance to human Parkinson's disease. Our results suggest that activation of astrocytic TRPV1 activates endogenous neuroprotective machinery in vivo and that it is a novel therapeutic target for the treatment of Parkinson's disease.


Subject(s)
Astrocytes/metabolism , Ciliary Neurotrophic Factor/metabolism , Dopaminergic Neurons/metabolism , Neuroprotection , Parkinson Disease/metabolism , Parkinson Disease/pathology , Substantia Nigra/metabolism , Animals , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Disease Models, Animal , Dopaminergic Neurons/pathology , Female , Humans , Nerve Regeneration , Parkinson Disease/physiopathology , Rats , Substantia Nigra/cytology , Substantia Nigra/pathology , TRPV Cation Channels/metabolism
5.
J Biol Chem ; 288(51): 36717-32, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24235151

ABSTRACT

Calpains are a family of calcium-dependent cysteine proteases that are ubiquitously expressed in mammals and play critical roles in neuronal death by catalyzing substrate proteolysis. Here, we developed two-dimensional gel electrophoresis-based protease proteomics to identify putative calpain substrates. To accomplish this, cellular lysates from neuronal cells were first separated by pI, and the immobilized sample on a gel strip was incubated with a recombinant calpain and separated by molecular weight. Among 25 altered protein spots that were differentially expressed by at least 2-fold, we confirmed that arsenical pump-driving ATPase, optineurin, and peripherin were cleaved by calpain using in vitro and in vivo cleavage assays. Furthermore, we found that all of these substrates were cleaved in MN9D cells treated with either ionomycin or 1-methyl-4-phenylpyridinium, both of which cause a calcium-mediated calpain activation. Their cleavage was blocked by calcium chelator or calpain inhibitors. In addition, calpain-mediated cleavage of these substrates and its inhibition by calpeptin were confirmed in a middle cerebral artery occlusion model of cerebral ischemia, as well as a stereotaxic brain injection model of Parkinson disease. Transient overexpression of each protein was shown to attenuate 1-methyl-4-phenylpyridinium-induced cell death, indicating that these substrates may confer protection of varying magnitudes against dopaminergic injury. Taken together, the data indicate that our protease proteomic method has the potential to be applicable for identifying proteolytic substrates affected by diverse proteases. Moreover, the results described here will help us decipher the molecular mechanisms underlying the progression of neurodegenerative disorders where protease activation is critically involved.


Subject(s)
Calpain/metabolism , Dopaminergic Neurons/metabolism , Proteome/metabolism , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Arsenite Transporting ATPases/genetics , Arsenite Transporting ATPases/metabolism , Calpain/antagonists & inhibitors , Cell Death , Cell Line , Dipeptides/pharmacology , Dipeptides/therapeutic use , Dopaminergic Neurons/drug effects , Electrophoresis, Gel, Two-Dimensional/methods , Glycine/analogs & derivatives , Glycine/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Ionomycin/pharmacology , Peripherins/genetics , Peripherins/metabolism , Proteomics/methods , Rats , Rats, Sprague-Dawley
6.
J Neuroimmunol ; 265(1-2): 36-42, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24090651

ABSTRACT

The present study examined whether Interleukin-13 (IL-13) or IL-4, an anti-inflammatory cytokine, could induce cell death of activated microglia by prothrombin kringle-2 (pKr-2) which is a domain of prothrombin distinct from thrombin. Microglia cell death was detected at eight days after co-treatment of pKr-2 with IL-13/IL-4 in vitro. This cell death was assessed by live assay, dead assay, TUNEL and MTT assay. In parallel, reactive oxygen species (ROS) production was evident as assessed by superoxide assay, WST-1 and analyzing DCF in combination of pKr-2 and IL-13 or IL-4 treated microglia. The IL-13/IL-4-enhanced ROS production and cell death in pKr-2 activated microglia was partially inhibited by an NADPH oxidase inhibitor, apocynin and/or by several antioxidants. Moreover, Western blot analysis showed a significant increase in cyclooxygenase-2 (COX-2) expression in combination of pKr-2 and IL-13 or IL-4 treated microglia, which was partially inhibited by apocynin and an antioxidant, trolox. Additional studies demonstrated that microglia cell death was reversed by treatment with COX-2 inhibitor, NS398. Our data strongly suggest that oxidative stress and COX-2 activation through NADPH oxidase may contribute to IL-13/IL-4 induced cell death of pKr-2 activated microglia.


Subject(s)
Interleukin-13/pharmacology , Interleukin-4/pharmacology , Kringles , Microglia/drug effects , Microglia/metabolism , Oxidative Stress/drug effects , Prothrombin/chemistry , Acetophenones/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/pharmacology , In Situ Nick-End Labeling , NADPH Oxidases/metabolism , Nitrobenzenes/pharmacology , Prothrombin/pharmacology , Rats , Sulfonamides/pharmacology , Tetrazolium Salts , Thiazoles
7.
Brain Res Bull ; 89(3-4): 92-6, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22796104

ABSTRACT

The present study examined whether capsaicin (CAP), an agonist of transient receptor potential vanilloid subtype 1 (TRPV1) can prevent 1-methyl-4-phenylpyridinium (MPP(+))-induced dopaminergic (DA) neuronal death in the substantia nigra (SN). Unilateral injection of MPP(+) into the median forebrain bundle of rat brain resulted in a significant loss of nigral DA neurons, assessed by tyrosine hydroxylase (TH) immunostaining. In parallel, activation of microglia, visualized by OX-42 and OX-6 immunostaining were also observed in the SN, where degeneration of nigral neurons was found. By contrast, MPP(+) neurotoxicity was partially inhibited by co-treatment with MPP(+) and CAP. Interestingly, CAP significantly decreased not only immunoreactivity of OX-42 and OX-6 but also production of microglia-derived reactive oxygen species (ROS) in the SN of MPP(+)-treated rats. In experiments designed to further verify effectiveness of CAP against microglia-derived neurotoxicity, CAP inhibited ROS production and blocked MPP(+)-induced death of DA neurons in co-cultures of mesencephalic neurons and microglia, but not in microglia-free, neuron-enriched mesencephalic cultures. This beneficial effect was reversed by capsazepine, an antagonist of TRPV1, expressed in microglia, indicating TRPV1 involvement. Our data demonstrate for the first time that CAP may inhibit microglial activation-mediated oxidative stress via TRPV1, suggesting that CAP and its analogs may have therapeutic value by inhibiting microglial activation and/or ROS generation that occurs in Parkinson's disease.


Subject(s)
Dopaminergic Neurons/drug effects , Mesencephalon/cytology , Microglia/physiology , Nerve Degeneration/metabolism , Oxidative Stress/physiology , TRPV Cation Channels/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Analysis of Variance , Animals , Antigens, CD/metabolism , Capsaicin/therapeutic use , Cell Count , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Drug Interactions , Female , Functional Laterality , Microglia/drug effects , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neurotoxins/pharmacology , Oxidative Stress/drug effects , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sensory System Agents/therapeutic use , Substantia Nigra/drug effects , Tyrosine 3-Monooxygenase/metabolism
8.
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
9.
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
10.
Neuropharmacology ; 60(6): 963-74, 2011 May.
Article in English | MEDLINE | ID: mdl-21288472

ABSTRACT

Parkinson's disease (PD) is characterized by degeneration of nigrostriatal dopaminergic (DA) neurons. Mice treated with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) exhibit microglial activation-induced oxidative stress and inflammation, and nigrostriatal DA neuronal damage, and thus serve as an experimental model of PD. Here, we report that fluoxetine, one of the most commonly prescribed antidepressants, prevents MPTP-induced degeneration of nigrostriatal DA neurons and increases striatal dopamine levels with the partial motor recovery. This was accompanied by inhibiting transient expression of proinflammatory cytokines and inducible nitric oxide synthase; and attenuating microglial NADPH oxidase activation, reactive oxygen species/reactive nitrogen species production, and consequent oxidative damage. Interestingly, fluoxetine was found to protect DA neuronal damage from 1-methyl-4-phenyl-pyridinium (MPP(+)) neurotoxicity in co-cultures of mesencephalic neurons and microglia but not in neuron-enriched mesencephalic cultures devoid of microglia. The present in vivo and in vitro findings show that fluoxetine may possess anti-inflammatory properties and inhibit glial activation-mediated oxidative stress. Therefore, we carefully propose that neuroprotection of fluoxetine might be associated with its anti-inflammatory properties and could be employed as novel therapeutic agents for PD and other disorders associated with neuroinflammation and microglia-derived oxidative damage.


Subject(s)
Dopamine/metabolism , Fluoxetine/therapeutic use , MPTP Poisoning/prevention & control , Microglia/metabolism , Nerve Degeneration/prevention & control , Neurons/pathology , Recovery of Function/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacokinetics , 1-Methyl-4-phenylpyridinium/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Cell Count/methods , Coculture Techniques/methods , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cytokines/metabolism , Fluoxetine/pharmacology , MPTP Poisoning/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Nerve Degeneration/chemically induced , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Rotarod Performance Test/methods , Substantia Nigra
11.
Brain Res ; 1363: 143-50, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-20858471

ABSTRACT

Lipopolysaccharide (LPS)-induced microglial activation causes degeneration of nigral dopaminergic (DA) neurons. Here, we examined whether fluoxetine prevents LPS-induced degeneration of DA in the rat substantia nigra (SN) in vivo. Seven days after LPS injection into the SN, immunostaining for tyrosine hydroxylase (TH) revealed a significant loss of nigral DA neurons. Parallel activation of microglia (visualized by OX-42 and ED1 immunohistochemistry), production of reactive oxygen species (ROS) (assessed by hydroethidine histochemistry), and degeneration of nigral DA neurons were also observed in the SN. Western blot analyses and double-label immunohistochemistry showed an increase in the expression of inducible nitric oxide synthase (iNOS) within activated microglia. LPS also induced translocation of p67(phox), the cytosolic component of NADPH oxidase, to the membrane of SN microglia, indicating activation of NADPH oxidase. The LPS-induced loss of nigral DA neurons was partially inhibited by fluoxetine, and the observed neuroprotective effects were associated with fluoxetine-mediated suppression of microglial NADPH oxidase activation and iNOS upregulation, and decreased ROS generation and oxidative stress. These results suggest that fluoxetine and analogs thereof may be beneficial for the treatment of neurodegenerative diseases, such as PD, that are associated with microglia-derived oxidative damage.


Subject(s)
Fluoxetine/pharmacology , Microglia/drug effects , Oxidative Stress/drug effects , Parkinsonian Disorders/drug therapy , Substantia Nigra/drug effects , Animals , Enzyme Activation/drug effects , Female , Lipopolysaccharides/toxicity , Microglia/enzymology , Microglia/pathology , NADPH Oxidases/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Nitric Oxide Synthase Type II/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Rats , Rats, Sprague-Dawley , Selective Serotonin Reuptake Inhibitors/pharmacology , Substantia Nigra/metabolism , Substantia Nigra/pathology
12.
BMC Neurosci ; 11: 74, 2010 Jun 12.
Article in English | MEDLINE | ID: mdl-20540782

ABSTRACT

BACKGROUND: Gangliosides, sialic acid-containing glycosphingolipids exist in mammalian cell membranes particularly neuronal membranes. The trisialoganglioside (GT1b) is one of the major brain gangliosides and acts as an endogenous regulator in the brain. We previously showed GT1b induces mesencephalic dopaminergic (DA) neuronal death, both in vivo and in vitro. We further investigate the underlying mechanisms of GT1b neurotoxicity. RESULTS: Consistent with earlier findings, GT1b attenuated the DA neuron number and dopamine uptake level in mesencephalic cultures. Morphological evidence revealed GT1b-induced chromatin condensation and nuclear fragmentation as well as an increased number of TUNEL-positive cells, compared to control cultures. Interestingly, while GT1b enhanced caspase-3 activity, DEVD, a caspase-3 inhibitor, failed to rescue DA neuronal death. Immunoblot analysis revealed that GT1b inactivates Akt through dephosphorylation at both Ser473 and Thr308, subsequent dephosphorylation of GSK-3beta, a substrate of Akt, and hyperphosphorylation of tau, downstream of GSK-3beta. Moreover, a GSK-3beta specific inhibitor, L803-mt, attenuated tau phosphorylation and rescued DA neurons from cell death in mesencephalic cultures. CONCLUSION: Our data provide novel evidence that a Akt/GSK-3beta/tau-dependent, but not caspase-3 signaling pathway plays a pivotal role in GT1b-mediated neurotoxic actions on mesencephalic DA neurons.


Subject(s)
Dopamine/metabolism , Gangliosides/pharmacology , Glycogen Synthase Kinase 3/metabolism , Mesencephalon/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , tau Proteins/metabolism , Analysis of Variance , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Count , Cells, Cultured , Female , Immunohistochemistry , In Situ Nick-End Labeling , Mesencephalon/cytology , Mesencephalon/drug effects , Microscopy, Immunoelectron , Neurons/cytology , Neurons/drug effects , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
13.
J Immunol ; 185(2): 1230-7, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20566832

ABSTRACT

The present study examined whether the antidepressant paroxetine 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. Real-time PCR, Western blotting, and immunohistochemistry showed upregulation of proinflammatory cytokines, activation of microglial NADPH oxidase and astroglial myeloperoxidase, and subsequent reactive oxygen species production and oxidative DNA damage in the MPTP-treated substantia nigra. Treatment with paroxetine prevented degeneration of nigrostriatal DA neurons, increased striatal dopamine levels, and improved motor function. This neuroprotection afforded by paroxetine was associated with the suppression of astroglial myeloperoxidase expression and/or NADPH oxidase-derived reactive oxygen species production and reduced expression of proinflammatory cytokines, including IL-1beta, TNF-alpha, and inducible NO synthase, by activated microglia. The present findings show that paroxetine may possess anti-inflammatory properties and inhibit glial activation-mediated oxidative stress, suggesting that paroxetine and its analogues may have therapeutic value in the treatment of aspects of Parkinson's disease related to neuroinflammation.


Subject(s)
Brain/drug effects , Neurons/drug effects , Parkinson Disease, Secondary/prevention & control , Paroxetine/pharmacology , Substantia Nigra/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antidepressive Agents, Second-Generation/pharmacology , Blotting, Western , Brain/metabolism , Brain/pathology , Dopamine/metabolism , Enzyme Activation/drug effects , Immunohistochemistry , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/physiopathology , Peroxidase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Substantia Nigra/cytology , Substantia Nigra/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
Neurochem Int ; 57(1): 16-32, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20403401

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder caused by selective degeneration of the dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). Although mitochondrial abnormality, oxidative stress and proteasomal dysfunction are recognized as major contributors to the progression of PD, there is a limited understanding of the key molecular events that provoke degeneration of DA neurons. Using a proteomic approach, we attempted to identify profiles of proteins with altered expression levels in rats following unilateral stereotaxic injection of 6-hydroxydopamine into the SNc. Protein expression profiles of these proteins in the substantia nigra and the striatum were made using two-dimensional gel electrophoresis in conjunction with a mass spectrometry. More than 70 identified proteins displayed significant differences in their temporal and spatial expression pattern between experimental and vehicle-operated control groups. Based on the identity of the proteins, we further searched for potential binding partners using biological databases available on the web and constructed a protein interaction network. Among several interconnected proteins in the network, we verified the interaction between prohibitin and the NADH-ubiquinone oxidoreductase 30kDa subunit (NDUFS3 subunit; a mitochondrial complex I subunit) by co-immunoprecipitation. We also confirmed, using immunohistochemical localization, that both prohibitin and the NDUFS3 subunit were increased in the dying DA neurons, suggesting its potential role in regulating mitochondrial function in dying DA neurons. Furthermore, knockdown of prohibitin accelerated 6-hydroxydopamine-induced cell death in SH-SY5Y cells. Our results raise the possibility that interconnected proteins in the network may positively or negatively impact the progression of DA neuronal death.


Subject(s)
Brain/drug effects , Brain/pathology , Disease Models, Animal , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Protein Interaction Mapping/methods , Proteomics/methods , Animals , Cell Line, Tumor , Gene Regulatory Networks/drug effects , Gene Silencing/drug effects , Humans , Male , Neuroblastoma/metabolism , Neuroblastoma/pathology , Parkinsonian Disorders/genetics , Prohibitins , Random Allocation , Rats , Rats, Sprague-Dawley , Repressor Proteins/deficiency , Repressor Proteins/genetics
15.
J Immunol ; 183(7): 4666-74, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19752235

ABSTRACT

In the present study, we investigated the effects of IL-13, a well-known anti-inflammatory cytokine, on the thrombin-treated hippocampus in vivo. NeuN immunohistochemistry and Nissl staining revealed significant loss of hippocampal CA1 neurons upon intrahippocampal injection of thrombin. This neurotoxicity was accompanied by substantial microglial activation, as evident from OX-42 immunohistochemistry results. In parallel, Western blot analysis and hydroethidine histochemistry disclosed activation of NADPH oxidase, generation of reactive oxygen species, and oxidative damage in the hippocampal CA1 area showing hippocampal neuron degeneration. Interestingly, immunohistochemical and biochemical experiments showed that intrahippocampal injection of thrombin increased IL-13 immunoreactivity and IL-13 levels as early as 8 h after thrombin, reaching a peak at 7 days, which was maintained up to 14 days. Moreover, double-label immunohistochemistry revealed IL-13 immunoreactivity exclusively in activated microglia. IL-13-neutralizing Abs significantly rescued CA1 hippocampal neurons from thrombin neurotoxicity. In parallel, neutralization of IL-13 inhibited activation of NADPH oxidase, reactive oxygen species production, and oxidative damage. Additionally, IL-13 neutralization suppressed the expression of inducible NO synthase and several proinflammatory cytokines. To our knowledge, the present study is the first to show that IL-13 triggers microglial NADPH oxidase-derived oxidative stress, leading to the degeneration of hippocampal neurons in vivo, as occurs in cases of Alzheimer's disease.


Subject(s)
Apoptosis/immunology , Hippocampus/immunology , Interleukin-13/physiology , Microglia/enzymology , Microglia/immunology , NADPH Oxidases/metabolism , Neurons/immunology , Oxidative Stress/immunology , Animals , Enzyme Activation/immunology , Female , Gene Expression Regulation, Enzymologic/immunology , Hippocampus/enzymology , Hippocampus/pathology , Interleukin-13/biosynthesis , Interleukin-13/genetics , Microglia/pathology , NADPH Oxidases/biosynthesis , NADPH Oxidases/physiology , Neurons/enzymology , Neurons/pathology , Rats , Rats, Sprague-Dawley , Thrombin/administration & dosage , Thrombin/toxicity , Up-Regulation/immunology
16.
J Neuroimmunol ; 214(1-2): 83-92, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19660816

ABSTRACT

In the present study, we examine whether prothrombin kringle-2 (pKr-2), a domain of prothrombin distinct from thrombin and a potent microglial activator induces reactive oxygen species (ROS) generation through stimulation of microglial NADPH oxidase activity, and whether this phenomenon contributes to oxidative damage and consequent neurodegeneration. Intracortical injection of pKr-2 caused significant loss of cortical neurons in vivo after seven days, as evident from Nissl staining and immunohistochemical analysis using the neuronal-specific nuclear protein (NeuN) antibody. In parallel, pKr-2-activated microglia and ROS production were observed in rat cortex displaying degeneration of cortical neurons. Reverse transcription-PCR at various time points after pKr-2 administration disclosed early and transient expression of inducible nitric oxide synthase (iNOS) and proinflammatory cytokines, such as interleukin 1beta (IL-1beta). Co-localization of iNOS, IL-1beta, and TNF-alpha within microglia was evident with double-label immunohistochemistry. Additionally, pKr-2 induced upregulation of cytosolic components of NADPH oxidase (p67(phox)), translocation of cytosolic p67(phox) protein to the membrane, and p67(phox) expression in microglia in the cortex in vivo, signifying NADPH oxidase activation. The pKr-2-induced oxidation of proteins and loss of cortical neurons were partially inhibited by DPI, an NADPH oxidase inhibitor, and trolox, an antioxidant. Consistent with our hypothesis, following treatment with pKr-2 in vitro, neurotoxicity was detected exclusively in co-cultures of cortical neurons and microglia, but not in microglia-free neuron-enriched cortical cultures, indicating that microglia are required for pKr-2 neurotoxicity. Our results strongly suggest that pKr-2 as an endogenous compound participates in cortical neuron death through microglial NADPH oxidase-mediated oxidative stress.


Subject(s)
Apoptosis , Cerebral Cortex/metabolism , Kringles , Microglia/enzymology , NADPH Oxidases/metabolism , Neurons/metabolism , Oxidative Stress , Prothrombin/metabolism , Animals , Antioxidants/pharmacology , Blotting, Western , Chromans/pharmacology , Coculture Techniques , Enzyme Activation , Female , Fluorescent Antibody Technique , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Singlet Oxygen/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
J Biol Chem ; 283(15): 9986-98, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18250162

ABSTRACT

The aim of this study was to investigate changes in protein profiles during the early phase of dopaminergic neuronal death using two-dimensional gel electrophoresis in conjunction with mass spectrometry. Several protein spots were identified whose expression was significantly altered following treatment of MN9D dopaminergic neuronal cells with 6-hydroxydopamine (6-OHDA). In particular, we detected oxidative modification of thioredoxin-dependent peroxidases (peroxiredoxins; PRX) in treated MN9D cells. Oxidative modification of PRX induced by 6-OHDA was blocked in the presence of N-acetylcysteine, suggesting that reactive oxygen species (ROS) generated by 6-OHDA induce oxidation of PRX. These findings were confirmed in primary cultures of mesencephalic neurons and in rat brain injected stereotaxically. Overexpression of PRX1 in MN9D cells (MN9D/PRX1) exerted neuroprotective effects against death induced by 6-OHDA through scavenging of ROS. Consequently, generation of both superoxide anion and hydrogen peroxide following 6-OHDA treatment was decreased in MN9D/PRX1. Furthermore, overexpression of PRX1 protected cells against 6-OHDA-induced activation of p38 MAPK and subsequent activation of caspase-3. In contrast, 6-OHDA-induced apoptotic death signals were enhanced by RNA interference-targeted reduction of PRX1 in MN9D cells. Taken together, our data suggest that the redox state of PRX may be intimately involved in 6-OHDA-induced dopaminergic neuronal cell death and also provide a molecular mechanism by which PRX1 exerts a protective role in experimental models of Parkinson disease.


Subject(s)
Adrenergic Agents/toxicity , Apoptosis/drug effects , Mesencephalon/metabolism , Oxidopamine/toxicity , Parkinson Disease, Secondary/metabolism , Peroxiredoxins/metabolism , Signal Transduction/drug effects , Acetylcysteine/pharmacology , Animals , Caspase 3/metabolism , Cell Line , Disease Models, Animal , Enzyme Activation/drug effects , Neurons/metabolism , Oxidation-Reduction/drug effects , Parkinson Disease, Secondary/chemically induced , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Neurosci Res ; 86(5): 1053-63, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18183616

ABSTRACT

The present study investigated whether thrombin can induce the production of reactive oxygen species (ROS) through activation of neuronal NADPH oxidase and whether this contributes to oxidative damage and consequently to neurodegeneration. Immunocytochemical and biochemical evidence demonstrated that, in neuron-enriched hippocampal cultures, thrombin induces neurodegeneration in a dose-dependent manner. In parallel, ROS production was evident as assessed by analyzing DCF and hydroethidine. Real-time PCR analysis, at various time points after thrombin treatment, also demonstrated that expression of NADPH oxidase subunits (p47(phox) and p67(phox)) occurs. In addition, Western blot analysis and double-label immunocytochemistry showed an up-regulation in the expression of cytosolic components (Rac 1 and p67(phox)), the translocation of cytosolic proteins (p47(phox) and p67(phox)) to the membrane, and the localization of gp91(phox) or p47(phox) expression in hippocampal neurons of cultures and CA1 layer. The thrombin-induced ROS production, protein oxidation, and loss of cultured hippocampal neurons were partially attenuated by an NADPH oxidase inhibitor and/or by several antioxidants. Collectively, the present study is the first to demonstrate that, in cultured hippocampal neurons, thrombin-induced neurotoxicity is, at least in part, caused by neuronal NADPH oxidase-mediated oxidative stress. This strongly suggests that thrombin can act as an endogenous neurotoxin, and inhibitors of thrombin and/or antioxidants can be useful agents for treating oxidative stress-mediated hippocampal neurodegenerative diseases, such as Alzheimer's disease.


Subject(s)
Hippocampus/metabolism , NADPH Oxidases/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Thrombin/toxicity , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Heredodegenerative Disorders, Nervous System/drug therapy , Heredodegenerative Disorders, Nervous System/metabolism , Heredodegenerative Disorders, Nervous System/physiopathology , Hippocampus/drug effects , Hippocampus/physiopathology , NADPH Oxidases/drug effects , NADPH Oxidases/genetics , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurotoxins/antagonists & inhibitors , Neurotoxins/metabolism , Neurotoxins/toxicity , Oxidative Stress/drug effects , Phosphoproteins/genetics , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Thrombin/antagonists & inhibitors , Thrombin/metabolism
19.
Curr Aging Sci ; 1(3): 192-201, 2008 Dec.
Article in English | MEDLINE | ID: mdl-20021392

ABSTRACT

We investigated the effects of interleukin-4 (IL-4), a well-known anti-inflammatory cytokine, on thrombin-treated rat hippocampi in vivo. Intrahippocampal injection of thrombin resulted in a significant loss of hippocampal CA1 neurons, as determined by Nissl staining and NeuN immunohistochemistry. Thrombin-induced neurotoxicity was accompanied by substantial microglial activation, as demonstrated by OX-42 immunohistochemistry. In parallel, Western blot analysis and hydroethidine histochemistry revealed activation of NADPH oxidase (as demonstrated by increased translocation of the cytosolic proteins p67(phox) and p47(phox)), generation of reactive oxygen species (ROS), and oxidative damage in the hippocampal CA1 area, where degeneration of hippocampal neurons was evident. Interestingly, immunohistochemical and biochemical analysis demonstrated that intrahippocampal injection of thrombin increased immunoreactivity and levels of IL-4 as early as 8 h post-treatment, reaching a peak at 7 days that was maintained for up to 14 days. Moreover, double-label immunohistochemistry detected IL-4 immunoreactivity solely in activated microglia. In experiments to explore the involvement of IL-4 in neurotoxicity, IL-4-neutralizing antibodies significantly increased the survival of CA1 hippocampal neurons at 7 days post-thrombin treatment. Consistent with these results, IL-4 neutralization inhibited activation of NADPH oxidase, ROS production and oxidative damage. Thus, the present study is the first to demonstrate that IL-4 generates microglial NADPH oxidase-derived oxidative stress and leads to the degeneration of hippocampal neurons in vivo, as occurs in Alzheimer's disease.


Subject(s)
Hippocampus/metabolism , Hippocampus/pathology , Interleukin-4/metabolism , NADPH Oxidases/metabolism , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Cell Count , Cell Death/drug effects , Cell Death/physiology , Female , Hippocampus/drug effects , Humans , Microglia/drug effects , Microglia/enzymology , Nerve Degeneration/pathology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Thrombin/administration & dosage
20.
Mol Neurobiol ; 35(3): 245-54, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17917113

ABSTRACT

Transient receptor potential vanilloid subtype 1 (TRPV1), also known as vanilloid receptor 1 (VR1), is a nonselective cation channel that is activated by a variety of ligands, such as exogenous capsaicin (CAP) or endogenous anandamide (AEA), as well as products of lipoxygenases. Cannabinoid type 1 (CB1) receptor belongs to the G protein-coupled receptor superfamily and is activated by cannabinoids such as AEA and exogenous Delta-9-tetrahydrocannabinol (THC). TRPV1 and CB1 receptors are widely expressed in the brain and play many significant roles in various brain regions; however, the issue of whether TRPV1 or CB1 receptors mediate neuroprotection or neurotoxicity remains controversial. Furthermore, functional crosstalk between these two receptors has been recently reported. It is therefore timely to review current knowledge regarding the functions of these two receptors and to consider new directions of investigation on their roles in the brain.


Subject(s)
Brain/metabolism , Receptor, Cannabinoid, CB1/metabolism , TRPV Cation Channels/metabolism , Animals , Arachidonic Acids/metabolism , Brain/anatomy & histology , Cannabinoid Receptor Modulators/metabolism , Capsaicin/chemistry , Capsaicin/metabolism , Endocannabinoids , Neuroprotective Agents/metabolism , Polyunsaturated Alkamides/metabolism , Sensory System Agents/chemistry , Sensory System Agents/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...