Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neuroscience ; 487: 166-183, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35167938

ABSTRACT

The Munc13 family of proteins is critically involved in synaptic vesicle priming and release in glutamatergic neurons in the brain. Munc13-1 binds to alcohol and, in Drosophila, modulates sedation sensitivity and self-administration. We examined the effect of alcohol consumption on the expression of Munc13-1 and Munc13-2, NMDA receptor subunits GluN1, GluN2A and GluN2B in the hippocampus-derived HT22 cells, hippocampal primary neuron culture, and wild-type and Munc13-1+/- male mouse hippocampus after ethanol consumption (Drinking in the Dark (DID) paradigm). In HT22 cells, Munc13-1 was upregulated following 25 mM ethanol treatment for 24 h. In the primary neuronal culture, however, the expression of both Munc13-1 and Munc13-2 increased after ethanol exposure. While Munc13-1 was upregulated in the hippocampus, Munc13-2 was downregulated following DID. This differential effect was found in the CA1 subfield of the hippocampus. Although Munc13-1+/- mice had approximately 50% Munc13-1 expression compared to wild-type, it was nonetheless significantly increased following DID. Munc13-1 and Munc13-2 were expressed in vesicular glutamate transporter1 (VGLUT1) immunoreactive neurons in the hippocampus, but ethanol did not alter the expression of VGLUT1. The NMDA receptor subunits, GluN1, GluN2A and GluN2B were upregulated in the hippocampal primary culture and in the CA1. Ethanol exerts a differential effect on the expression of Munc13-1 and Munc13-2 in the CA1 in male mice. Our study also found that ethanol's effect on Munc13 expression is dependent on the experimental paradigm, and both Munc13-1 and Munc13-2 could contribute to the ethanol-induced augmentation of glutamatergic neurotransmission.


Subject(s)
Alcohol Drinking , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins , Receptors, N-Methyl-D-Aspartate , Animals , Drosophila/metabolism , Ethanol/pharmacology , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Male , Mice , Nerve Tissue Proteins/biosynthesis , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission
2.
Front Pharmacol ; 12: 631375, 2021.
Article in English | MEDLINE | ID: mdl-33995031

ABSTRACT

Oxidative stress, neuroinflammation and apoptosis are some of the key etiological factors responsible for dopamin(DA)ergic degeneration during Parkinson's disease (PD), yet the downstream molecular mechanisms underlying neurodegeneration are largely unknown. Recently, a genome-wide association study revealed the FYN gene to be associated with PD, suggesting that Fyn kinase could be a pharmacological target for PD. In this study, we report that Fyn-mediated PKCδ tyrosine (Y311) phosphorylation is a key event preceding its proteolytic activation in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinsonism. MPP+/MPTP induced Fyn kinase activation in N27 DAergic neuronal cells and the mouse substantia nigra. PKCδ-Y311 phosphorylation by activated Fyn initiates the apoptotic caspase-signaling cascade during DAergic degeneration. Pharmacological attenuation of Fyn activity protected DAergic neurons from MPP+-induced degeneration in primary mesencephalic neuronal cultures. We further employed Fyn wild-type and Fyn knockout (KO) mice to confirm whether Fyn is a valid pharmacological target of DAergic neurodegeneration. Primary mesencephalic neurons from Fyn KO mice were greatly protected from MPP+-induced DAergic cell death, neurite loss and DA reuptake loss. Furthermore, Fyn KO mice were significantly protected from MPTP-induced PKCδ-Y311 phosphorylation, behavioral deficits and nigral DAergic degeneration. This study thus unveils a mechanism by which Fyn regulates PKCδ's pro-apoptotic function and DAergic degeneration. Pharmacological inhibitors directed at Fyn activation could prove to be a novel therapeutic target in the delay or halting of selective DAergic degeneration during PD.

3.
Mol Neurodegener ; 16(1): 5, 2021 01 31.
Article in English | MEDLINE | ID: mdl-33517893

ABSTRACT

BACKGROUND: Genome-wide association studies have established clusterin (CLU) as a genetic modifier for late-onset Alzheimer's disease (AD). Both protective and risk alleles have been identified which may be associated with its expression levels. However, the physiological function of clusterin in the central nervous system remains largely unknown. METHODS: We examined Clu expression in mouse brains by immunohistochemistry and high-resolution imaging. We performed electrophysiological recordings and morphological analysis of dendritic spines in wild-type and Clu knockout mice. We tested synaptic function of astrocytic Clu using neuron-glia co-cultures and by AAV-mediated astroglial Clu expression in vivo. Finally, we investigated the role of astrocytic Clu on synaptic properties and amyloid pathology in 5xFAD transgenic mouse model of AD. RESULTS: We show that astrocyte secreted Clu co-localizes with presynaptic puncta of excitatory neurons. Loss of Clu led to impaired presynaptic function and reduced spine density in vivo. Neurons co-cultured with Clu-overexpressing astrocytes or treated with conditioned media from HEK293 cells transfected with Clu displayed enhanced excitatory neurotransmission. AAV-mediated astroglial Clu expression promoted excitatory neurotransmission in wild-type mice and rescued synaptic deficits in Clu knockout mice. Overexpression of Clu in the astrocytes of 5xFAD mice led to reduced Aß pathology and fully rescued the synaptic deficits. CONCLUSION: We identify Clu as an astrocyte-derived synaptogenic and anti-amyloid factor; the combination of these activities may influence the progression of late-onset AD.


Subject(s)
Alzheimer Disease/metabolism , Astrocytes/metabolism , Clusterin/metabolism , Neurons/metabolism , Synaptic Transmission/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Clusterin/genetics , Disease Models, Animal , Genome-Wide Association Study , Humans , Mice, Transgenic , Neuropathology/methods , Synaptic Transmission/genetics
4.
Sci Transl Med ; 12(573)2020 12 09.
Article in English | MEDLINE | ID: mdl-33298560

ABSTRACT

Neuroinflammation has been increasingly recognized to play a critical role in Alzheimer's disease (AD). The epoxy fatty acids (EpFAs) are derivatives of the arachidonic acid metabolism pathway and have anti-inflammatory activities. However, their efficacy is limited because of their rapid hydrolysis by the soluble epoxide hydrolase (sEH). We report that sEH is predominantly expressed in astrocytes and is elevated in postmortem brain tissue from patients with AD and in the 5xFAD ß amyloid mouse model of AD. The amount of sEH expressed in AD mouse brains correlated with a reduction in brain EpFA concentrations. Using a specific small-molecule sEH inhibitor, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), we report that TPPU treatment protected wild-type mice against LPS-induced inflammation in vivo. Long-term administration of TPPU to the 5xFAD mouse model via drinking water reversed microglia and astrocyte reactivity and immune pathway dysregulation. This was associated with reduced ß amyloid pathology and improved synaptic integrity and cognitive function on two behavioral tests. TPPU treatment correlated with an increase in EpFA concentrations in the brains of 5xFAD mice, demonstrating brain penetration and target engagement of this small molecule. These findings support further investigation of TPPU as a potential therapeutic agent for the treatment of AD.


Subject(s)
Alzheimer Disease , Epoxide Hydrolases , Alzheimer Disease/drug therapy , Animals , Epoxy Compounds , Humans , Mice , Phenylurea Compounds , Piperidines
5.
Biochemistry ; 58(27): 3016-3030, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31243993

ABSTRACT

Bryostatin 1 is a natural macrolide shown to improve neuronal connections and enhance memory in mice. Its mechanism of action is largely attributed to the modulation of novel and conventional protein kinase Cs (PKCs) by binding to their regulatory C1 domains. Munc13-1 is a C1 domain-containing protein that shares common endogenous and exogenous activators with novel and conventional PKC subtypes. Given the essential role of Munc13-1 in the priming of synaptic vesicles and neuronal transmission overall, we explored the potential interaction between bryostatin 1 and Munc13-1. Our results indicate that in vitro bryostatin 1 binds to both the isolated C1 domain of Munc13-1 ( Ki = 8.07 ± 0.90 nM) and the full-length Munc13-1 protein ( Ki = 0.45 ± 0.04 nM). Furthermore, confocal microscopy and immunoblot analysis demonstrated that in intact HT22 cells bryostatin 1 mimics the actions of phorbol esters, a previously established class of Munc13-1 activators, and induces plasma membrane translocation of Munc13-1, a hallmark of its activation. Consistently, bryostatin 1 had no effect on the Munc13-1H567K construct that is insensitive to phorbol esters. Effects of bryostatin 1 on the other Munc13 family members, ubMunc13-2 and bMunc13-2, resembled those of Munc13-1 for translocation. Lastly, we observed an increased level of expression of Munc13-1 following a 24 h incubation with bryostatin 1 in both HT22 and primary mouse hippocampal cells. This study characterizes Munc13-1 as a molecular target of bryostatin 1. Considering the crucial role of Munc13-1 in neuronal function, these findings provide strong support for the potential role of Munc13s in the actions of bryostatin 1.


Subject(s)
Bryostatins/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Animals , Binding Sites , Cell Line , Cells, Cultured , Mice , Models, Molecular , Molecular Docking Simulation , Nerve Tissue Proteins/chemistry , Neurons/metabolism , Phorbol Esters/pharmacology , Protein Binding
6.
Neurotoxicology ; 64: 204-218, 2018 01.
Article in English | MEDLINE | ID: mdl-28539244

ABSTRACT

Chronic manganese (Mn) exposure induces neurotoxicity, which is characterized by Parkinsonian symptoms resulting from impairment in the extrapyramidal motor system of the basal ganglia. Mitochondrial dysfunction and oxidative stress are considered key pathophysiological features of Mn neurotoxicity. Recent evidence suggests astrocytes as a major target of Mn neurotoxicity since Mn accumulates predominantly in astrocytes. However, the primary mechanisms underlying Mn-induced astroglial dysfunction and its role in metal neurotoxicity are not completely understood. In this study, we examined the interrelationship between mitochondrial dysfunction and astrocytic inflammation in Mn neurotoxicity. We first evaluated whether Mn exposure alters mitochondrial bioenergetics in cultured astrocytes. Metabolic activity assessed by MTS assay revealed an IC50 of 92.68µM Mn at 24h in primary mouse astrocytes (PMAs) and 50.46µM in the human astrocytic U373 cell line. Mn treatment reduced mitochondrial mass, indicative of impaired mitochondrial function and biogenesis, which was substantiated by the significant reduction in mRNA of mitofusin-2, a protein that serves as a ubiquitination target for mitophagy. Furthermore, Mn increased mitochondrial circularity indicating augmented mitochondrial fission. Seahorse analysis of bioenergetics status in Mn-treated astrocytes revealed that Mn significantly impaired the basal mitochondrial oxygen consumption rate as well as the ATP-linked respiration rate. The effect of Mn on mitochondrial energy deficits was further supported by a reduction in ATP production. Mn-exposed primary astrocytes also exhibited a severely quiescent energy phenotype, which was substantiated by the inability of oligomycin to increase the extracellular acidification rate. Since astrocytes regulate immune functions in the CNS, we also evaluated whether Mn modulates astrocytic inflammation. Mn exposure in astrocytes not only stimulated the release of proinflammatory cytokines, but also exacerbated the inflammatory response induced by aggregated α-synuclein. The novel mitochondria-targeted antioxidant, mito-apocynin, significantly attenuated Mn-induced inflammatory gene expression, further supporting the role of mitochondria dysfunction and oxidative stress in mediating astrogliosis. Lastly, intranasal delivery of Mn in vivo elevated GFAP and depressed TH levels in the olfactory bulbs, clearly supporting the involvement of astrocytes in Mn-induced dopaminergic neurotoxicity. Collectively, our study demonstrates that Mn drives proinflammatory events in astrocytes by impairing mitochondrial bioenergetics.


Subject(s)
Astrocytes/drug effects , Encephalitis/chemically induced , Manganese/toxicity , Mitochondria/drug effects , Animals , Astrocytes/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Encephalitis/metabolism , Inhibitory Concentration 50 , Male , Mice, Inbred C57BL , Mitochondria/metabolism
7.
Biochim Biophys Acta Gen Subj ; 1861(11 Pt A): 2640-2651, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28713022

ABSTRACT

BACKGROUND: Resveratrol (1) is a naturally occurring polyphenol that has been implicated in neuroprotection. One of resveratrol's several biological targets is Ca2+-sensitive protein kinase C alpha (PKCα). Resveratrol inhibits PKCα by binding to its activator-binding C1 domain. Munc13-1 is a C1 domain-containing Ca2+-sensitive SNARE complex protein essential for vesicle priming and neurotransmitter release. METHODS: To test if resveratrol could also bind and inhibit Munc13-1, we studied the interaction of resveratrol and its derivatives, (E)-1,3-dimethoxy-5-(4-methoxystyryl)benzene, (E)-5,5'-(ethene-1,2-diyl)bis(benzene-1,2,3-triol), (E)-1,2-bis(3,4,5-trimethoxyphenyl)ethane, and (E)-5-(4-(hexadecyloxy)-3,5-dihydroxystyryl)benzene-1,2,3-triol with Munc13-1 by studying its membrane translocation from cytosol to plasma membrane in HT22 cells and primary hippocampal neurons. RESULTS: Resveratrol, but not the derivatives inhibited phorbol ester-induced Munc13-1 translocation from cytosol to membrane in HT22 cells and primary hippocampal neurons, as evidenced by immunoblot analysis and confocal microscopy. Resveratrol did not show any effect on Munc13-1H567K, a mutant which is not sensitive to phorbol ester. Binding studies with Munc13-1 C1 indicated that resveratrol competes with phorbol ester for the binding site. Molecular docking and dynamics studies suggested that hydroxyl groups of resveratrol interact with phorbol-ester binding residues in the binding pocket. CONCLUSIONS AND SIGNIFICANCE: This study characterizes Munc13-1 as a target of resveratrol and highlights the importance of dietary polyphenol in the management of neurodegenerative diseases.


Subject(s)
Nerve Tissue Proteins/chemistry , Neurons/metabolism , SNARE Proteins/chemistry , Stilbenes/administration & dosage , Animals , Binding Sites , Free Radical Scavengers/administration & dosage , Free Radical Scavengers/chemistry , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mice , Molecular Docking Simulation , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Phorbol Esters/administration & dosage , Phorbol Esters/chemistry , Primary Cell Culture , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/chemistry , Resveratrol , SNARE Proteins/metabolism , Synaptic Transmission/drug effects
8.
Antioxid Redox Signal ; 27(14): 1048-1066, 2017 Nov 10.
Article in English | MEDLINE | ID: mdl-28375739

ABSTRACT

AIMS: Parkinson's disease (PD) is a neurodegenerative disorder characterized by progressive motor deficits and degeneration of dopaminergic neurons. Caused by a number of genetic and environmental factors, mitochondrial dysfunction and oxidative stress play a role in neurodegeneration in PD. By selectively knocking out mitochondrial transcription factor A (TFAM) in dopaminergic neurons, the transgenic MitoPark mice recapitulate many signature features of the disease, including progressive motor deficits, neuronal loss, and protein inclusions. In the present study, we evaluated the neuroprotective efficacy of a novel mitochondrially targeted antioxidant, Mito-apocynin, in MitoPark mice and cell culture models of neuroinflammation and mitochondrial dysfunction. RESULTS: Oral administration of Mito-apocynin (10 mg/kg, thrice a week) showed excellent central nervous system bioavailability and significantly improved locomotor activity and coordination in MitoPark mice. Importantly, Mito-apocynin also partially attenuated severe nigrostriatal degeneration in MitoPark mice. Mechanistic studies revealed that Mito-apo improves mitochondrial function and inhibits NOX2 activation, oxidative damage, and neuroinflammation. INNOVATION: The properties of Mito-apocynin identified in the MitoPark transgenic mouse model strongly support potential clinical applications for Mito-apocynin as a viable neuroprotective and anti-neuroinflammatory drug for treating PD when compared to conventional therapeutic approaches. CONCLUSION: Collectively, our data demonstrate, for the first time, that a novel orally active apocynin derivative improves behavioral, inflammatory, and neurodegenerative processes in a severe progressive dopaminergic neurodegenerative model of PD. Antioxid. Redox Signal. 27, 1048-1066.


Subject(s)
Acetophenones/administration & dosage , Antioxidants/administration & dosage , DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Oxidative Stress/drug effects , Parkinson Disease/drug therapy , Acetophenones/chemistry , Acetophenones/pharmacology , Administration, Oral , Animals , Antioxidants/pharmacology , Cells, Cultured , Disease Models, Animal , Gene Knockout Techniques , Humans , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , NADPH Oxidase 2/metabolism , Parkinson Disease/metabolism
9.
Sci Transl Med ; 8(368): 368ra174, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27928028

ABSTRACT

Mitochondrial and autophagic dysfunction as well as neuroinflammation are involved in the pathophysiology of Parkinson's disease (PD). We hypothesized that targeting the mitochondrial pyruvate carrier (MPC), a key controller of cellular metabolism that influences mTOR (mammalian target of rapamycin) activation, might attenuate neurodegeneration of nigral dopaminergic neurons in animal models of PD. To test this, we used MSDC-0160, a compound that specifically targets MPC, to reduce its activity. MSDC-0160 protected against 1-methyl-4-phenylpyridinium (MPP+) insult in murine and cultured human midbrain dopamine neurons and in an α-synuclein-based Caenorhabditis elegans model. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice, MSDC-0160 improved locomotor behavior, increased survival of nigral dopaminergic neurons, boosted striatal dopamine levels, and reduced neuroinflammation. Long-term targeting of MPC preserved motor function, rescued the nigrostriatal pathway, and reduced neuroinflammation in the slowly progressive Engrailed1 (En1+/-) genetic mouse model of PD. Targeting MPC in multiple models resulted in modulation of mitochondrial function and mTOR signaling, with normalization of autophagy and a reduction in glial cell activation. Our work demonstrates that changes in metabolic signaling resulting from targeting MPC were neuroprotective and anti-inflammatory in several PD models, suggesting that MPC may be a useful therapeutic target in PD.


Subject(s)
Autophagy , Inflammation , Mitochondria/metabolism , Neurodegenerative Diseases/immunology , Parkinson Disease/immunology , Pyruvic Acid/chemistry , 1-Methyl-4-phenylpyridinium/chemistry , Animals , Behavior, Animal , Brain/metabolism , Caenorhabditis elegans , Disease Models, Animal , Dopamine/chemistry , Dopaminergic Neurons/metabolism , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Oxygen Consumption , Parkinson Disease/metabolism , Pyridines/chemistry , Signal Transduction , Substantia Nigra/metabolism , Thiazolidinediones/chemistry , alpha-Synuclein/chemistry
10.
Neurobiol Dis ; 93: 96-114, 2016 09.
Article in English | MEDLINE | ID: mdl-27151770

ABSTRACT

Chronic microglial activation has been linked to the progressive degeneration of the nigrostriatal dopaminergic neurons evidenced in Parkinson's disease (PD) pathogenesis. The exact etiology of PD remains poorly understood. Although both oxidative stress and neuroinflammation are identified as co-contributors in PD pathogenesis, signaling mechanisms underlying neurodegenerative processes have yet to be defined. Indeed, we recently identified that protein kinase C delta (PKCδ) activation is critical for induction of dopaminergic neuronal loss in response to neurotoxic stressors. However, it remains to be defined whether PKCδ activation contributes to immune signaling events driving microglial neurotoxicity. In the present study, we systematically investigated whether PKCδ contributes to the heightened microglial activation response following exposure to major proinflammatory stressors, including α-synuclein, tumor necrosis factor α (TNFα), and lipopolysaccharide (LPS). We report that exposure to the aforementioned inflammatory stressors dramatically upregulated PKCδ with a concomitant increase in its kinase activity and nuclear translocation in both BV-2 microglial cells and primary microglia. Importantly, we also observed a marked upregulation of PKCδ in the microglia of the ventral midbrain region of PD patients when compared to age-matched controls, suggesting a role for microglial PKCδ in neurodegenerative processes. Further, shRNA-mediated knockdown and genetic ablation of PKCδ in primary microglia blunted the microglial proinflammatory response elicited by the inflammogens, including ROS generation, nitric oxide production, and proinflammatory cytokine and chemokine release. Importantly, we found that PKCδ activated NFκB, a key mediator of inflammatory signaling events, after challenge with inflammatory stressors, and that transactivation of NFκB led to translocation of the p65 subunit to the nucleus, IκBα degradation and phosphorylation of p65 at Ser536. Furthermore, both genetic ablation and siRNA-mediated knockdown of PKCδ attenuated NFκB activation, suggesting that PKCδ regulates NFκB activation subsequent to microglial exposure to inflammatory stimuli. To further investigate the pivotal role of PKCδ in microglial activation in vivo, we utilized pre-clinical models of PD. We found that PKCδ deficiency attenuated the proinflammatory response in the mouse substantia nigra, reduced locomotor deficits and recovered mice from sickness behavior in an LPS-induced neuroinflammation model of PD. Likewise, we found that PKCδ knockout mice treated with MPTP displayed a dampened microglial inflammatory response. Moreover, PKCδ knockout mice exhibited reduced susceptibility to the neurotoxin-induced dopaminergic neurodegeneration and associated motor impairments. Taken together, our studies propose a pivotal role for PKCδ in PD pathology, whereby sustained PKCδ activation drives sustained microglial inflammatory responses and concomitant dopaminergic neurotoxicity consequently leading to neurobehavioral deficits. We conclude that inhibiting PKCδ activation may represent a novel therapeutic strategy in PD treatment.


Subject(s)
Dopaminergic Neurons/metabolism , Microglia/metabolism , Parkinson Disease/metabolism , Protein Kinase C-delta/metabolism , Animals , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mesencephalon/cytology , Mice, Knockout , Microglia/drug effects , Models, Theoretical , Nerve Degeneration/pathology , Parkinson Disease/pathology , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Up-Regulation/drug effects
11.
J Neuroimmune Pharmacol ; 11(2): 259-78, 2016 06.
Article in English | MEDLINE | ID: mdl-26838361

ABSTRACT

Mitochondrial dysfunction, oxidative stress and neuroinflammation have been implicated as key mediators contributing to the progressive degeneration of dopaminergic neurons in Parkinson's disease (PD). Currently, we lack a pharmacological agent that can intervene in all key pathological mechanisms, which would offer better neuroprotective efficacy than a compound that targets a single degenerative mechanism. Herein, we investigated whether mito-apocynin (Mito-Apo), a newly-synthesized and orally available derivative of apocynin that targets mitochondria, protects against oxidative damage, glial-mediated inflammation and nigrostriatal neurodegeneration in cellular and animal models of PD. Mito-Apo treatment in primary mesencephalic cultures significantly attenuated the 1-methyl-4-phenylpyridinium (MPP(+))-induced loss of tyrosine hydroxylase (TH)-positive neuronal cells and neurites. Mito-Apo also diminished MPP(+)-induced increases in glial cell activation and inducible nitric oxide synthase (iNOS) expression. Additionally, Mito-Apo decreased nitrotyrosine (3-NT) and 4-hydroxynonenol (4-HNE) levels in primary mesencephalic cultures. Importantly, we assessed the neuroprotective property of Mito-Apo in the MPTP mouse model of PD, wherein it restored the behavioral performance of MPTP-treated mice. Immunohistological analysis of nigral dopaminergic neurons and monoamine measurement further confirmed the neuroprotective effect of Mito-Apo against MPTP-induced nigrostriatal dopaminergic neuronal loss. Mito-Apo showed excellent brain bioavailability and also markedly attenuated MPTP-induced oxidative markers in the substantia nigra (SN). Furthermore, oral administration of Mito-Apo significantly suppressed MPTP-induced glial cell activation, upregulation of proinflammatory cytokines, iNOS and gp91phox in IBA1-positive cells of SN. Collectively, these results demonstrate that the novel mitochondria-targeted compound Mito-Apo exhibits profound neuroprotective effects in cellular and pre-clinical animal models of PD by attenuating oxidative damage and neuroinflammatory processes.


Subject(s)
Acetophenones/therapeutic use , Antioxidants/therapeutic use , Dopaminergic Neurons/drug effects , Inflammation Mediators/antagonists & inhibitors , Mitochondria/drug effects , Parkinsonian Disorders/prevention & control , Acetophenones/chemistry , Acetophenones/pharmacology , Animals , Animals, Newborn , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/chemistry , Antioxidants/pharmacology , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Treatment Outcome
12.
Neurobiol Dis ; 73: 70-82, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25281317

ABSTRACT

Current research on Parkinson's disease (PD) pathogenesis requires relevant animal models that mimic the gradual and progressive development of neuronal dysfunction and degeneration that characterizes the disease. Polymorphisms in engrailed 1 (En1), a homeobox transcription factor that is crucial for both the development and survival of mesencephalic dopaminergic neurons, are associated with sporadic PD. This suggests that En1 mutant mice might be a promising candidate PD model. Indeed, a mouse that lacks one En1 allele exhibits decreased mitochondrial complex I activity and progressive midbrain dopamine neuron degeneration in adulthood, both features associated with PD. We aimed to further characterize the disease-like phenotype of these En1(+/-) mice with a focus on early neurodegenerative changes that can be utilized to score efficacy of future disease modifying studies. We observed early terminal defects in the dopaminergic nigrostriatal pathway in En1(+/-) mice. Several weeks before a significant loss of dopaminergic neurons in the substantia nigra could be detected, we found that striatal terminals expressing high levels of dopaminergic neuron markers TH, VMAT2, and DAT were dystrophic and swollen. Using transmission electron microscopy, we identified electron dense bodies consistent with abnormal autophagic vacuoles in these terminal swellings. In line with these findings, we detected an up-regulation of the mTOR pathway, concurrent with a downregulation of the autophagic marker LC3B, in ventral midbrain and nigral dopaminergic neurons of the En1(+/-) mice. This supports the notion that autophagic protein degradation is reduced in the absence of one En1 allele. We imaged the nigrostriatal pathway using the CLARITY technique and observed many fragmented axons in the medial forebrain bundle of the En1(+/-) mice, consistent with axonal maintenance failure. Using in vivo electrochemistry, we found that nigrostriatal terminals in the dorsal striatum were severely deficient in dopamine release and reuptake. Our findings support a progressive retrograde degeneration of En1(+/-) nigrostriatal neurons, akin to what is suggested to occur in PD. We suggest that using the En1(+/-) mice as a model will provide further key insights into PD pathogenesis, and propose that axon terminal integrity and function can be utilized to estimate dopaminergic neuron health and efficacy of experimental PD therapies.


Subject(s)
Corpus Striatum/metabolism , Corpus Striatum/pathology , Homeodomain Proteins/genetics , Nerve Degeneration/etiology , Parkinson Disease , Substantia Nigra/pathology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Autophagy/genetics , Disease Models, Animal , Disease Progression , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Dopaminergic Neurons/ultrastructure , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homovanillic Acid/metabolism , Mice , Mice, Transgenic , Parkinson Disease/complications , Parkinson Disease/genetics , Parkinson Disease/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Substantia Nigra/metabolism , TOR Serine-Threonine Kinases/metabolism , Time Factors , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
13.
J Biol Chem ; 289(50): 34743-67, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25342743

ABSTRACT

The oxidative stress-sensitive protein kinase Cδ (PKCδ) has been implicated in dopaminergic neuronal cell death. However, little is known about the epigenetic mechanisms regulating PKCδ expression in neurons. Here, we report a novel mechanism by which the PKCδ gene can be regulated by histone acetylation. Treatment with histone deacetylase (HDAC) inhibitor sodium butyrate (NaBu) induced PKCδ expression in cultured neurons, brain slices, and animal models. Several other HDAC inhibitors also mimicked NaBu. The chromatin immunoprecipitation analysis revealed that hyperacetylation of histone H4 by NaBu is associated with the PKCδ promoter. Deletion analysis of the PKCδ promoter mapped the NaBu-responsive element to an 81-bp minimal promoter region. Detailed mutagenesis studies within this region revealed that four GC boxes conferred hyperacetylation-induced PKCδ promoter activation. Cotransfection experiments and Sp inhibitor studies demonstrated that Sp1, Sp3, and Sp4 regulated NaBu-induced PKCδ up-regulation. However, NaBu did not alter the DNA binding activities of Sp proteins or their expression. Interestingly, a one-hybrid analysis revealed that NaBu enhanced transcriptional activity of Sp1/Sp3. Overexpression of the p300/cAMP-response element-binding protein-binding protein (CBP) potentiated the NaBu-mediated transactivation potential of Sp1/Sp3, but expressing several HDACs attenuated this effect, suggesting that p300/CBP and HDACs act as coactivators or corepressors in histone acetylation-induced PKCδ up-regulation. Finally, using genetic and pharmacological approaches, we showed that NaBu up-regulation of PKCδ sensitizes neurons to cell death in a human dopaminergic cell model and brain slice cultures. Together, these results indicate that histone acetylation regulates PKCδ expression to augment nigrostriatal dopaminergic cell death, which could contribute to the progressive neuropathogenesis of Parkinson disease.


Subject(s)
Dopaminergic Neurons/pathology , Epigenesis, Genetic , Histones/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Kinase C-delta/metabolism , Up-Regulation , Acetylation/drug effects , Animals , Brain/pathology , Butyric Acid/pharmacology , Cell Death/drug effects , Cell Line , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Mice , Oxidative Stress/drug effects , Parkinson Disease/metabolism , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Kinase C-delta/genetics , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sp Transcription Factors/chemistry , Sp Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Up-Regulation/drug effects , p300-CBP Transcription Factors/metabolism
14.
Biochim Biophys Acta ; 1842(8): 1282-94, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24060637

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disease in the elderly, and no cure or disease-modifying therapies exist. Several lines of evidence suggest that mitochondrial dysfunction and oxidative stress have a central role in the dopaminergic neurodegeneration of Parkinson's disease. In this context, mitochondria-targeted therapies that improve mitochondrial function may have great promise in the prevention and treatment of Parkinson's disease. In this review, we discuss the recent developments in mitochondria-targeted antioxidants and their potential beneficial effects as a therapy for ameliorating mitochondrial dysfunction in Parkinson's disease.


Subject(s)
Antioxidants/therapeutic use , Clinical Trials as Topic , Mitochondria/metabolism , Parkinson Disease/drug therapy , Antioxidants/chemistry , Antioxidants/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/pathology , Oxidative Stress/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism
15.
Neurosci Lett ; 549: 57-62, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23721786

ABSTRACT

The most prominent mechanism proposed for death of dopaminergic neurons in Parkinson's disease (PD) is elevated generation of reactive oxygen/nitrogen species (ROS/RNS). Recent studies suggest that ROS produced during PD pathogenesis may contribute to cytotoxicity in cell culture models of PD. We hypothesized that inhibition of ROS production would prevent PD symptoms in the LRRK2(R1441G) transgenic (tg) mouse model of PD. These mice overexpress a mutant form of leucine-rich repeat kinase 2 (LRRK2) and are reported to develop PD-like symptoms at approximately 10 months of age. Despite similar expression of the transgene, our colony did not recapitulate the same type of motor dysfunction originally reported. However, tests of motor coordination (pole test, Rotor-Rod) revealed a significant defect in LRRK2(R1441G) mice by 16 months of age. LRRK2(R1441G) tg mice, or wild type littermates, were given diapocynin (200mg/kg, a proposed NADPH oxidase inhibitor) three times per week by oral gavage starting at 12 weeks of age. Decreased performance on the pole test and Rotor-Rod in the LRRK2(R1441G) mice was prevented with diapocynin treatment. No loss in open field movement or rearing was found. As expected, tyrosine hydroxylase staining was similar in both the substantia nigra and striatum in all treatment groups. Together these data demonstrate that diapocynin is a viable agent for protection of neurobehavioral function.


Subject(s)
Acetophenones/pharmacology , Biphenyl Compounds/pharmacology , Gait/drug effects , Motor Activity/drug effects , Parkinson Disease/prevention & control , Protein Serine-Threonine Kinases/genetics , Animals , Disease Models, Animal , Gait/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Transgenic , Motor Activity/genetics , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Rotarod Performance Test
16.
J Biol Chem ; 288(30): 21955-71, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23754278

ABSTRACT

Parkinson disease (PD) is a chronic neurodegenerative disease characterized by a slow and progressive degeneration of dopaminergic neurons in substantia nigra. The pathophysiological mechanisms underlying PD remain unclear. Pin1, a major peptidyl-prolyl isomerase, has recently been associated with certain diseases. Notably, Ryo et al. (Ryo, A., Togo, T., Nakai, T., Hirai, A., Nishi, M., Yamaguchi, A., Suzuki, K., Hirayasu, Y., Kobayashi, H., Perrem, K., Liou, Y. C., and Aoki, I. (2006) J. Biol. Chem. 281, 4117-4125) implicated Pin1 in PD pathology. Therefore, we sought to systematically characterize the role of Pin1 in PD using cell culture and animal models. To our surprise we observed a dramatic up-regulation of Pin1 mRNA and protein levels in dopaminergic MN9D neuronal cells treated with the parkinsonian toxicant 1-methyl-4-phenylpyridinium (MPP(+)) as well as in the substantia nigra of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. Notably, a marked expression of Pin1 was also observed in the substantia nigra of human PD brains along with a high co-localization of Pin1 within dopaminergic neurons. In functional studies, siRNA-mediated knockdown of Pin1 almost completely prevented MPP(+)-induced caspase-3 activation and DNA fragmentation, indicating that Pin1 plays a proapoptotic role. Interestingly, multiple pharmacological Pin1 inhibitors, including juglone, attenuated MPP(+)-induced Pin1 up-regulation, α-synuclein aggregation, caspase-3 activation, and cell death. Furthermore, juglone treatment in the MPTP mouse model of PD suppressed Pin1 levels and improved locomotor deficits, dopamine depletion, and nigral dopaminergic neuronal loss. Collectively, our findings demonstrate for the first time that Pin1 is up-regulated in PD and has a pathophysiological role in the nigrostriatal dopaminergic system and suggest that modulation of Pin1 levels may be a useful translational therapeutic strategy in PD.


Subject(s)
Apoptosis/physiology , Dopaminergic Neurons/metabolism , Peptidylprolyl Isomerase/metabolism , Up-Regulation , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Gene Expression/drug effects , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/pharmacology , Neurotoxins/pharmacology , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/metabolism , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/genetics , RNA Interference , RNA-Directed DNA Polymerase , Substantia Nigra/drug effects , Substantia Nigra/metabolism
17.
J Biol Chem ; 288(29): 20843-20855, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23744073

ABSTRACT

Although Parkinson disease (PD) is a progressive neurodegenerative disorder, available animal models do not exhibit irreversible neurodegeneration, and this is a major obstacle in finding out an effective drug against this disease. Here we delineate a new irreversible model to study PD pathogenesis. The model is based on simple castration of young male mice. Levels of inducible nitric-oxide synthase (iNOS), glial markers (glial fibrillary acidic protein and CD11b), and α-synuclein were higher in nigra of castrated male mice than normal male mice. On the other hand, after castration, the level of glial-derived neurotrophic factor (GDNF) markedly decreased in the nigra of male mice. Accordingly, castration also induced the loss of tyrosine hydroxylase-positive neurons in the nigra and decrease in tyrosine hydroxylase-positive fibers and neurotransmitters in the striatum. Reversal of nigrostriatal pathologies in castrated male mice by subcutaneous implantation of 5α-dihydrotestosterone pellets validates an important role of male sex hormone in castration-induced nigrostriatal pathology. Interestingly, castration was unable to cause glial activation, decrease nigral GDNF, augment the death of nigral dopaminergic neurons, induce the loss of striatal fibers, and impair neurotransmitters in iNOS(-/-) male mice. Furthermore, we demonstrate that iNOS-derived NO is responsible for decreased expression of GDNF in activated astrocytes. Together, our results suggest that castration induces nigrostriatal pathologies via iNOS-mediated decrease in GDNF. These results are important because castrated young male mice may be used as a simple, toxin-free, and nontransgenic animal model to study PD-related nigrostriatal pathologies, paving the way for easy drug screening against PD.


Subject(s)
Aging/pathology , Castration/adverse effects , Nitric Oxide Synthase Type II/metabolism , Parkinson Disease/enzymology , Parkinson Disease/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Down-Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hippocampus/pathology , Hormone Replacement Therapy , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/enzymology , Neostriatum/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/deficiency , Parkinson Disease/etiology , Parkinson Disease/physiopathology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Testosterone/administration & dosage , Testosterone/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation/drug effects , alpha-Synuclein/metabolism
18.
J Neuroinflammation ; 9: 241, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23092448

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a devastating neurodegenerative disorder characterized by progressive motor debilitation, which affects several million people worldwide. Recent evidence suggests that glial cell activation and its inflammatory response may contribute to the progressive degeneration of dopaminergic neurons in PD. Currently, there are no neuroprotective agents available that can effectively slow the disease progression. Herein, we evaluated the anti-inflammatory and antioxidant efficacy of diapocynin, an oxidative metabolite of the naturally occurring agent apocynin, in a pre-clinical 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. METHODS: Both pre-treatment and post-treatment of diapocynin were tested in the MPTP mouse model of PD. Diapocynin was administered via oral gavage to MPTP-treated mice. Following the treatment, behavioral, neurochemical and immunohistological studies were performed. Neuroinflammatory markers, such as ionized calcium binding adaptor molecule 1 (Iba-1), glial fibrillary acidic protein (GFAP), gp91phox and inducible nitric oxide synthase (iNOS), were measured in the nigrostriatal system. Nigral tyrosine hydroxylase (TH)-positive neurons as well as oxidative markers 3-nitrotyrosine (3-NT), 4-hydroxynonenal (4-HNE) and striatal dopamine levels were quantified for assessment of the neuroprotective efficacy of diapocynin. RESULTS: Oral administration of diapocynin significantly attenuated MPTP-induced microglial and astroglial cell activation in the substantia nigra (SN). MPTP-induced expression of gp91phox and iNOS activation in the glial cells of SN was also completely blocked by diapocynin. Notably, diapocynin markedly inhibited MPTP-induced oxidative markers including 3-NT and 4-HNE levels in the SN. Treatment with diapocynin also significantly improved locomotor activity, restored dopamine and its metabolites, and protected dopaminergic neurons and their nerve terminals in this pre-clinical model of PD. Importantly, diapocynin administered 3 days after initiation of the disease restored the neurochemical deficits. Diapocynin also halted the disease progression in a chronic mouse model of PD. CONCLUSIONS: Collectively, these results demonstrate that diapocynin exhibits profound neuroprotective effects in a pre-clinical animal model of PD by attenuating oxidative damage and neuroinflammatory responses. These findings may have important translational implications for treating PD patients.


Subject(s)
Acetophenones/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Encephalitis/drug therapy , MPTP Poisoning/drug therapy , Neuroprotective Agents/administration & dosage , Acetophenones/metabolism , Animals , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/metabolism , Chromatography, High Pressure Liquid , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Disease Progression , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dose-Response Relationship, Drug , Encephalitis/etiology , Fluoresceins , MPTP Poisoning/complications , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , NADPH Oxidases/metabolism , Neuroglia/drug effects , Neurotransmitter Agents/metabolism , Nitric Oxide Synthase Type II/metabolism , Organic Chemicals , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Tyrosine 3-Monooxygenase/metabolism
19.
PLoS One ; 7(6): e38113, 2012.
Article in English | MEDLINE | ID: mdl-22723850

ABSTRACT

Neuroinflammation and oxidative stress underlie the pathogenesis of various neurodegenerative disorders. Here we demonstrate that sodium phenylbutyrate (NaPB), an FDA-approved therapy for reducing plasma ammonia and glutamine in urea cycle disorders, can suppress both proinflammatory molecules and reactive oxygen species (ROS) in activated glial cells. Interestingly, NaPB also decreased the level of cholesterol but involved only intermediates, not the end product of cholesterol biosynthesis pathway for these functions. While inhibitors of both geranylgeranyl transferase (GGTI) and farnesyl transferase (FTI) inhibited the activation of NF-κB, inhibitor of GGTI, but not FTI, suppressed the production of ROS. Accordingly, a dominant-negative mutant of p21(rac), but not p21(ras), attenuated the production of ROS from activated microglia. Inhibition of both p21(ras) and p21(rac) activation by NaPB in microglial cells suggests that NaPB exerts anti-inflammatory and antioxidative effects via inhibition of these small G proteins. Consistently, we found activation of both p21(ras) and p21(rac)in vivo in the substantia nigra of acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. Oral administration of NaPB reduced nigral activation of p21(ras) and p21(rac), protected nigral reduced glutathione, attenuated nigral activation of NF-κB, inhibited nigral expression of proinflammatory molecules, and suppressed nigral activation of glial cells. These findings paralleled dopaminergic neuronal protection, normalized striatal neurotransmitters, and improved motor functions in MPTP-intoxicated mice. Consistently, FTI and GGTI also protected nigrostriata in MPTP-intoxicated mice. Furthermore, NaPB also halted the disease progression in a chronic MPTP mouse model. These results identify novel mode of action of NaPB and suggest that NaPB may be of therapeutic benefit for neurodegenerative disorders.


Subject(s)
Antioxidants/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Parkinson Disease/metabolism , Phenylbutyrates/pharmacology , Animals , Antioxidants/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Male , Mevalonic Acid/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , NF-kappa B/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oncogene Protein p21(ras)/metabolism , Parkinson Disease/genetics , Phenylbutyrates/administration & dosage , Reactive Oxygen Species/metabolism
20.
J Neurosci ; 31(6): 2035-51, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21307242

ABSTRACT

We recently demonstrated that protein kinase Cδ (PKCδ), an important member of the novel PKC family, is a key oxidative stress-sensitive kinase that can be activated by caspase-3-dependent proteolytic cleavage to induce dopaminergic neuronal cell death. We now report a novel association between α-synuclein (αsyn), a protein associated with the pathogenesis of Parkinson's disease, and PKCδ, in which αsyn negatively modulates the p300- and nuclear factor-κB (NFκB)-dependent transactivation to downregulate proapoptotic kinase PKCδ expression and thereby protects against apoptosis in dopaminergic neuronal cells. Stable expression of human wild-type αsyn at physiological levels in dopaminergic neuronal cells resulted in an isoform-dependent transcriptional suppression of PKCδ expression without changes in the stability of mRNA and protein or DNA methylation. The reduction in PKCδ transcription was mediated, in part, through the suppression of constitutive NFκB activity targeted at two proximal PKCδ promoter κB sites. This occurred independently of NFκB/IκBα (inhibitor of κBα) nuclear translocation but was associated with decreased NFκB-p65 acetylation. Also, αsyn reduced p300 levels and its HAT (histone acetyltransferase) activity, thereby contributing to diminished PKCδ transactivation. Importantly, reduced PKCδ and p300 expression also were observed within nigral dopaminergic neurons in αsyn-transgenic mice. These findings expand the role of αsyn in neuroprotection by modulating the expression of the key proapoptotic kinase PKCδ in dopaminergic neurons.


Subject(s)
Apoptosis/physiology , Dopamine/metabolism , Neurons/physiology , Protein Kinase C-delta/metabolism , alpha-Synuclein/metabolism , p300-CBP Transcription Factors/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Apoptosis/genetics , Caspase 3/metabolism , Cell Line, Transformed , Cell Line, Tumor , Chromatin Immunoprecipitation/methods , Computational Biology , Dopamine Agents/pharmacology , Electrophoretic Mobility Shift Assay/methods , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Expression Regulation, Enzymologic/physiology , Humans , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Transgenic , Neurons/drug effects , Protein Kinase C-delta/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Substantia Nigra/cytology , Transfection/methods , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...