Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
PLoS One ; 10(10): e0139914, 2015.
Article En | MEDLINE | ID: mdl-26452228

Klotho transgenic mice exhibit resistance to oxidative stress as measured by their urinal levels of 8-hydroxy-2-deoxyguanosine, albeit this anti-oxidant defense mechanism has not been locally investigated in the brain. Here, we tested the hypothesis that the reactive oxygen species (ROS)-sensitive apoptosis signal-regulating kinase 1 (ASK1)/p38 MAPK pathway regulates stress levels in the brain of these mice and showed that: 1) the ratio of free ASK1 to thioredoxin (Trx)-bound ASK1 is relatively lower in the transgenic brain whereas the reverse is true for the Klotho knockout mice; 2) the reduced p38 activation level in the transgene corresponds to higher level of ASK1-bound Trx, while the KO mice showed elevated p38 activation and lower level of-bound Trx; and 3) that 14-3-3ζ is hyper phosphorylated (Ser-58) in the transgene which correlated with increased monomer forms. In addition, we evaluated the in vivo robustness of the protection by challenging the brains of Klotho transgenic mice with a neurotoxin, MPTP and analyzed for residual neuron numbers and integrity in the substantia nigra pars compacta. Our results show that Klotho overexpression significantly protects dopaminergic neurons against oxidative damage, partly by modulating p38 MAPK activation level. Our data highlight the importance of ASK1/p38 MAPK pathway in the brain and identify Klotho as a possible anti-oxidant effector.


Dopaminergic Neurons/metabolism , Glucuronidase/metabolism , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Oxidative Stress , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Brain/metabolism , Brain/pathology , Dopaminergic Neurons/pathology , Enzyme Activation , Glucuronidase/genetics , Klotho Proteins , MAP Kinase Kinase Kinase 5/genetics , Mice , Mice, Knockout , Oxidation-Reduction , p38 Mitogen-Activated Protein Kinases/genetics
2.
Hum Mol Genet ; 24(20): 5805-27, 2015 Oct 15.
Article En | MEDLINE | ID: mdl-26220976

Many genes involved in brain development have been associated with human neurodevelopmental disorders, but underlying pathophysiological mechanisms remain undefined. Human genetic and mouse behavioral analyses suggest that ENGRAILED-2 (EN2) contributes to neurodevelopmental disorders, especially autism spectrum disorder. In mouse, En2 exhibits dynamic spatiotemporal expression in embryonic mid-hindbrain regions where monoamine neurons emerge. Considering their importance in neuropsychiatric disorders, we characterized monoamine systems in relation to forebrain neurogenesis in En2-knockout (En2-KO) mice. Transmitter levels of serotonin, dopamine and norepinephrine (NE) were dysregulated from Postnatal day 7 (P7) to P21 in En2-KO, though NE exhibited the greatest abnormalities. While NE levels were reduced ∼35% in forebrain, they were increased 40 -: 75% in hindbrain and cerebellum, and these patterns paralleled changes in locus coeruleus (LC) fiber innervation, respectively. Although En2 promoter was active in Embryonic day 14.5 -: 15.5 LC neurons, expression diminished thereafter and gene deletion did not alter brainstem NE neuron numbers. Significantly, in parallel with reduced NE levels, En2-KO forebrain regions exhibited reduced growth, particularly hippocampus, where P21 dentate gyrus granule neurons were decreased 16%, suggesting abnormal neurogenesis. Indeed, hippocampal neurogenic regions showed increased cell death (+77%) and unexpectedly, increased proliferation. Excess proliferation was restricted to early Sox2/Tbr2 progenitors whereas increased apoptosis occurred in differentiating (Dcx) neuroblasts, accompanied by reduced newborn neuron survival. Abnormal neurogenesis may reflect NE deficits because intra-hippocampal injections of ß-adrenergic agonists reversed cell death. These studies suggest that disruption of hindbrain patterning genes can alter monoamine system development and thereby produce forebrain defects that are relevant to human neurodevelopmental disorders.


Dopaminergic Neurons/metabolism , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Neurogenesis , Prosencephalon/metabolism , Serotonergic Neurons/metabolism , Animals , Dopaminergic Neurons/physiology , Doublecortin Protein , Female , Gene Deletion , Humans , Male , Mice , Mice, Knockout , Norepinephrine/metabolism , Prosencephalon/growth & development , Prosencephalon/pathology , Prosencephalon/physiopathology , Serotonergic Neurons/physiology , Swimming
3.
Exp Neurol ; 250: 376-83, 2013 Dec.
Article En | MEDLINE | ID: mdl-24184050

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by a prominent loss of nigrostriatal dopamine (DA) neurons with an accompanying neuroinflammation. The peptide angiotensin II (AngII) plays a role in oxidative-stress induced disorders and is thought to mediate its detrimental actions via activation of AngII AT1 receptors. The brain renin-angiotensin system is implicated in neurodegenerative disorders including PD. Blockade of the angiotensin converting enzyme or AT1 receptors provides protection in acute animal models of parkinsonism. We demonstrate here that treatment of mice with the angiotensin converting enzyme inhibitor captopril protects the striatum from acutely administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrine (MPTP), and that chronic captopril protects the nigral DA cell bodies from degeneration in a progressive rat model of parkinsonism created by the chronic intracerebral infusion of 1-methyl-4-phenylpyridinium (MPP+). The accompanying activation of microglia in the substantia nigra of MPP+-treated rats was reduced by the chronic captopril treatment. These findings indicate that captopril is neuroprotective for nigrostriatal DA neurons in both acute and chronic rodent PD models. Targeting the brain AngII pathway may be a feasible approach to slowing neurodegeneration in PD.


Angiotensin-Converting Enzyme Inhibitors/pharmacology , Brain/drug effects , Captopril/pharmacology , Dopaminergic Neurons/drug effects , MPTP Poisoning/drug therapy , Animals , Brain/metabolism , Brain/pathology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Immunohistochemistry , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Mice , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Rats
4.
Toxicol Appl Pharmacol ; 273(2): 355-64, 2013 Dec 01.
Article En | MEDLINE | ID: mdl-24070585

Persistent neuroinflammation and microglial activation play an integral role in the pathogenesis of many neurological disorders. We investigated the role of voltage-gated sodium channels (VGSC) and Na(+)/H(+) exchangers (NHE) in the activation of immortalized microglial cells (BV-2) after lipopolysaccharide (LPS) exposure. LPS (10 and 100 ng/ml) caused a dose- and time-dependent accumulation of intracellular sodium [(Na(+))i] in BV-2 cells. Pre-treatment of cells with the VGSC antagonist tetrodotoxin (TTX, 1 µM) abolished short-term Na(+) influx, but was unable to prevent the accumulation of (Na(+))i observed at 6 and 24h after LPS exposure. The NHE inhibitor cariporide (1 µM) significantly reduced accumulation of (Na(+))i 6 and 24h after LPS exposure. Furthermore, LPS increased the mRNA expression and protein level of NHE-1 in a dose- and time-dependent manner, which was significantly reduced after co-treatment with TTX and/or cariporide. LPS increased production of TNF-α, ROS, and H2O2 and expression of gp91(phox), an active subunit of NADPH oxidase, in a dose- and time-dependent manner, which was significantly reduced by TTX or TTX+cariporide. Collectively, these data demonstrate a closely-linked temporal relationship between VGSC and NHE-1 in regulating function in activated microglia, which may provide avenues for therapeutic interventions aimed at reducing neuroinflammation.


Inflammation/metabolism , Inflammation/pathology , Microglia/metabolism , Sodium-Hydrogen Exchangers/physiology , Voltage-Gated Sodium Channels/physiology , Animals , Cell Line, Transformed , Dose-Response Relationship, Drug , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Voltage-Gated Sodium Channel Blockers/pharmacology
5.
Neurotherapeutics ; 10(1): 143-53, 2013 Jan.
Article En | MEDLINE | ID: mdl-23296837

Consumption of coffee is associated with reduced risk of Parkinson's disease (PD), an effect that has largely been attributed to caffeine. However, coffee contains numerous components that may also be neuroprotective. One of these compounds is eicosanoyl-5-hydroxytryptamide (EHT), which ameliorates the phenotype of α-synuclein transgenic mice associated with decreased protein aggregation and phosphorylation, improved neuronal integrity and reduced neuroinflammation. Here, we sought to investigate if EHT has an effect in the MPTP model of PD. Mice fed a diet containing EHT for four weeks exhibited dose-dependent preservation of nigral dopaminergic neurons following MPTP challenge compared to animals given control feed. Reductions in striatal dopamine and tyrosine hydroxylase content were also less pronounced with EHT treatment. The neuroinflammatory response to MPTP was markedly attenuated, and indices of oxidative stress and JNK activation were significantly prevented with EHT. In cultured primary microglia and astrocytes, EHT had a direct anti-inflammatory effect demonstrated by repression of lipopolysaccharide-induced NFκB activation, iNOS induction, and nitric oxide production. EHT also exhibited a robust anti-oxidant activity in vitro. Additionally, in SH-SY5Y cells, MPP(+)-induced demethylation of phosphoprotein phosphatase 2A (PP2A), the master regulator of the cellular phosphoregulatory network, and cytotoxicity were ameliorated by EHT. These findings indicate that the neuroprotective effect of EHT against MPTP is through several mechanisms including its anti-inflammatory and antioxidant activities as well as its ability to modulate the methylation and hence activity of PP2A. Our data, therefore, reveal a strong beneficial effect of a novel component of coffee in multiple endpoints relevant to PD.


Anti-Inflammatory Agents/pharmacology , Coffee/chemistry , MPTP Poisoning/prevention & control , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , MPTP Poisoning/metabolism , Male , Mice , Mice, Inbred C57BL
6.
Exp Neurol ; 234(2): 482-7, 2012 Apr.
Article En | MEDLINE | ID: mdl-22309831

Parkinson's disease (PD) is characterized by a prominent degeneration of nigrostriatal dopamine (DA) neurons with an accompanying neuroinflammation. Despite clinical and preclinical studies of neuroprotective strategies for PD, there is no effective treatment for preventing or slowing the progression of neurodegeneration. The inverse correlation between caffeine consumption and risk of PD suggests that caffeine may exert neuroprotection. Whether caffeine is neuroprotective in a chronic progressive model of PD has not been evaluated nor is it known if delayed caffeine treatment can stop DA neuronal loss. We show that a chronic unilateral intra-cerebroventricular infusion of 1-methyl-4-phenylpyridinium in the rat brain for 28 days produces a progressive loss of DA and tyrosine hydroxylase in the ipsilateral striatum and a loss of DA cell bodies and microglial activation in the ipsilateral substantia nigra. Chronic caffeine consumption prevented the degeneration of DA cell bodies in the substantia nigra. Importantly, neuroprotection was still apparent when caffeine was introduced after the onset of the neurodegenerative process. These results add to the clinical relevance for adenosine receptors as a disease-modifying drug target for PD.


Caffeine/pharmacology , Dopaminergic Neurons/drug effects , Nerve Degeneration/drug therapy , Parkinson Disease/drug therapy , Substantia Nigra/drug effects , Animals , Caffeine/therapeutic use , Cell Count , Disease Models, Animal , Disease Progression , Dopaminergic Neurons/pathology , Male , Nerve Degeneration/pathology , Parkinson Disease/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/pathology
7.
PLoS One ; 7(1): e29935, 2012.
Article En | MEDLINE | ID: mdl-22253830

Apoptosis signal-regulating kinase 1 (ASK1), a member of the mitogen-activated protein kinase 3 family, is activated by oxidative stress. The death-signaling pathway mediated by ASK1 is inhibited by DJ-1, which is linked to recessively inherited Parkinson's disease (PD). Considering that DJ-1 deficiency exacerbates the toxicity of the mitochondrial complex I inhibitor 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), we sought to investigate the direct role and mechanism of ASK1 in MPTP-induced dopamine neuron toxicity. In the present study, we found that MPTP administration to wild-type mice activates ASK1 in the midbrain. In ASK1 null mice, MPTP-induced motor impairment was less profound, and striatal dopamine content and nigral dopamine neuron counts were relatively preserved compared to wild-type littermates. Further, microglia and astrocyte activation seen in wild-type mice challenged with MPTP was markedly attenuated in ASK⁻/⁻ mice. These data suggest that ASK1 is a key player in MPTP-induced glial activation linking oxidative stress with neuroinflammation, two well recognized pathogenetic factors in PD. These findings demonstrate that ASK1 is an important effector of MPTP-induced toxicity and suggest that inhibiting this kinase is a plausible therapeutic strategy for protecting dopamine neurons in PD.


MAP Kinase Kinase Kinase 5/metabolism , MPTP Poisoning/enzymology , Neuroglia/enzymology , Neuroglia/pathology , Animals , Behavior, Animal/drug effects , Cells, Cultured , Cytoprotection , Dopaminergic Neurons/metabolism , Enzyme Activation , Humans , MAP Kinase Kinase Kinase 5/deficiency , Mice , Mice, Inbred C57BL , Nerve Endings/metabolism , Nerve Endings/pathology , Oxidative Stress , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Substantia Nigra/pathology
8.
Curr Mol Pharmacol ; 2011 Jun 15.
Article En | MEDLINE | ID: mdl-21675953

Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with the loss of catecholaminergic neurons in several brain regions. The motor symptoms of the disease are related to degeneration of the midbrain dopaminergic neurons, which occurs some time after the disease has begun. Both the innate and adaptive immune systems appear to play a role in the neurodegenerative process, and may contribute to disease progression. Here we review the neuropathology of PD with attention focused on the involvement of the innate immune cells (microglia) and the adaptive immune cells (T lymphocytes). In addition, we discuss animal models of the disease with emphasis on a progressive rat model which allows a detailed analysis of how the immune system contributes to neurodegeneration both during early and late stages of degeneration. Finally, for the early detection and treatment of PD, we discuss immunotherapy approaches.

9.
J Neurosci ; 31(19): 6963-71, 2011 May 11.
Article En | MEDLINE | ID: mdl-21562258

α-Synuclein (α-Syn) is a key protein that accumulates as hyperphosphorylated aggregates in pathologic hallmark features of Parkinson's disease (PD) and other neurodegenerative disorders. Phosphorylation of this protein at serine 129 is believed to promote its aggregation and neurotoxicity, suggesting that this post-translational modification could be a therapeutic target. Here, we demonstrate that phosphoprotein phosphatase 2A (PP2A) dephosphorylates α-Syn at serine 129 and that this activity is greatly enhanced by carboxyl methylation of the catalytic C subunit of PP2A. α-Syn-transgenic mice raised on a diet supplemented with eicosanoyl-5-hydroxytryptamide, an agent that enhances PP2A methylation, dramatically reduced both α-Syn phosphorylation at Serine 129 and α-Syn aggregation in the brain. These biochemical changes were associated with enhanced neuronal activity, increased dendritic arborizations, and reduced astroglial and microglial activation, as well as improved motor performance. These findings support the notion that serine 129 phosphorylation of α-Syn is of pathogenetic significance and that promoting PP2A activity is a viable disease-modifying therapeutic strategy for α-synucleinopathies such as PD.


Neurodegenerative Diseases/metabolism , Neurons/metabolism , Phosphoprotein Phosphatases/metabolism , Serotonin/analogs & derivatives , alpha-Synuclein/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Blotting, Western , Brain/metabolism , Brain/pathology , Cell Line , Cells, Cultured , Dendrites/genetics , Dendrites/metabolism , Dendrites/pathology , Disease Models, Animal , Immunohistochemistry , Methylation , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/pathology , Phosphorylation/physiology , Serotonin/metabolism , alpha-Synuclein/genetics
10.
Exp Neurol ; 221(2): 329-34, 2010 Feb.
Article En | MEDLINE | ID: mdl-19948168

Zonisamide is an FDA-approved antiepileptic drug that blocks voltage-dependent Na(+) channels and T-type Ca(2+) channels and improves clinical outcome in Parkinson's disease (PD) patients when used as an adjunct to other PD therapies. Zonisamide also modifies dopamine (DA) activity, provides protection in ischemia models and influences antioxidant systems. Thus, we tested it for its ability to protect DA neurons in a mouse model of PD and investigated mechanisms underlying its protection. Concurrent treatment of mice with zonisamide and 1-methyl-4-phenyl-1,2,3,6-tetraydropyridine (MPTP) attenuated the reduction in striatal contents of DA, its metabolite DOPAC and tyrosine hydroxylase (TH). We also discovered that zonisamide inhibited monoamine oxidase B (MAO-B) activity in vitro with an IC(50) of 25 muM, a concentration that is well within the therapeutic range used for treating epilepsy in humans. Moreover, the irreversible binding of systemically administered selegiline to MAO-B in mouse brain was attenuated by zonisamide as measured by ex vivo assays. Zonisamide treatment alone did not produce any lasting effects on ex vivo MAO-B activity, indicating that it is a reversible inhibitor of the enzyme. Consistent with the effects of zonisamide on MAO-B, the striatal content of 1-methyl-4-phenylpyridinium (MPP(+)), which is derived from the administered MPTP via MAO-B actions, was substantially reduced in mice treated with MPTP and zonisamide. The potency and reversibility with which zonisamide blocks MAO-B may contribute to the ability of the drug to improve clinical symptoms in PD patients. The results also suggest that caution in its use may be necessary, especially when administered with other drugs, in the treatment of epilepsy or PD.


Antioxidants/pharmacology , Corpus Striatum/drug effects , Isoxazoles/pharmacology , MPTP Poisoning , Monoamine Oxidase/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Analysis of Variance , Animals , Antioxidants/therapeutic use , Corpus Striatum/enzymology , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Glutathione/metabolism , Isoxazoles/therapeutic use , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Male , Mass Spectrometry , Mice , Regression, Psychology , Tyrosine 3-Monooxygenase/metabolism , Zonisamide
11.
Parkinsonism Relat Disord ; 14 Suppl 2: S116-8, 2008.
Article En | MEDLINE | ID: mdl-18583172

Animal models of Parkinson's disease (PD) that more closely exhibit the chronic neuropathology seen in the human condition are needed in order to reveal processes involved with progressive neurodegeneration and for testing potential interventions for retarding dopamine (DA) neuronal loss. Here we describe the recently developed chronic rat model of PD in which 1-methyl-4-phenylpyridinium ion (MPP(+)) is infused chronically into the lateral cerebral ventricle. We review features of this model that include loss of nigral DA neurons, swollen and abnormal mitochondria, striatal inclusion-like bodies and microgliosis. Advantages as well as limitations of the model are addressed.


1-Methyl-4-phenylpyridinium/administration & dosage , Disease Models, Animal , Herbicides/administration & dosage , Parkinson Disease/etiology , Animals , Drug Administration Schedule , Humans , Injections, Intraventricular/methods , Parkinson Disease/pathology
12.
J Neurochem ; 106(1): 231-43, 2008 Jul.
Article En | MEDLINE | ID: mdl-18363831

Na(+)/H(+) exchanger (NHE) proteins are involved in intracellular pH and volume regulation and may indirectly influence neurotransmission. The abundant NHE isoform 1 (NHE1) has also been linked to brain cell damage during metabolic stress. It is not known, however, whether NHE1 or other NHE isoforms play a role in striatal dopamine (DA) neurotransmission under normal or metabolic stress conditions. Our study tested the hypothesis that NHE inhibition with cariporide mesilate (HOE-642) modifies striatal DA overflow and DAergic terminal damage in mice caused by the mitochondrial inhibitor malonate. We also explored the expression of NHE1-5 in the striatum and substantia nigra. Reverse microdialysis of HOE-642 elicited a transient elevation followed by a reduction in DA overflow accompanied by a decline in striatal DA content. HOE-642 pre-treatment diminished the malonate-induced DA overflow without reducing the intensity of the metabolic stress or subsequent DAergic axonal damage. Although NHE isoforms 1-5 are expressed in the striatum and midbrain, NHE1 protein was not co-located on nigrostriatal DAergic neurons. The absence of NHE1 co-location on DAergic neurons suggests that the effects of HOE-642 on striatal DA overflow are either mediated via NHE1 located on other cell types or that HOE-642 is acting through multiple NHE isoforms.


Brain Diseases, Metabolic/metabolism , Cation Transport Proteins/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Oxidative Stress/physiology , Sodium-Hydrogen Exchangers/metabolism , Synaptic Transmission/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Brain Diseases, Metabolic/physiopathology , Cation Transport Proteins/antagonists & inhibitors , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Guanidines/pharmacology , Male , Malonates/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/chemically induced , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/physiopathology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Sulfones/pharmacology , Synaptic Transmission/physiology , Wallerian Degeneration/chemically induced , Wallerian Degeneration/metabolism , Wallerian Degeneration/physiopathology
13.
Acta Neuropathol ; 115(4): 385-98, 2008 Apr.
Article En | MEDLINE | ID: mdl-18273623

Parkinson's disease (PD) is a progressive neurodegenerative disorder whose etiology is not understood. This disease occurs both sporadically and through inheritance of single genes, although the familial types are rare. Over the past decade or so, experimental and clinical data suggest that PD could be a multifactorial, neurodegenerative disease that involves strong interactions between the environment and genetic predisposition. Our understanding of the pathophysiology and motor deficits of the disease relies heavily on fundamental research on animal models and the last few years have seen an explosion of toxin-, inflammation-induced and genetically manipulated models. The insight gained from the use of such models has strongly advanced our understanding of the progression and stages of the disease. The models have also aided the development of novel therapies to improve symptomatic management, and they are critical for the development of neuroprotective strategies. This review critically evaluates these in vivo models and the roles they play in mimicking the progression of PD.


Disease Models, Animal , Parkinson Disease/pathology , Animals , Disease Progression , Humans
14.
Prog Neurobiol ; 83(5): 310-31, 2007 Dec.
Article En | MEDLINE | ID: mdl-18023959

This review summarizes recent developments that have contributed to understand how adenosine receptors, particularly A2A receptors, modulate brain injury in various animal models of neurological disorders, including Parkinson's disease (PD), stroke, Huntington's disease (HD), multiple sclerosis, Alzheimer's disease (AD) and HIV-associated dementia. It is clear that extracellular adenosine acting at adenosine receptors influences the functional outcome in a broad spectrum of brain injuries, indicating that A2A Rs may modulate some general cellular processes to affect neuronal cells death. Pharmacological, neurochemical and molecular/genetic approaches to the complex actions of A2A receptors in different cellular elements suggest that A2A receptor activation can be detrimental or protective after brain insults, depending on the nature of brain injury and associated pathological conditions. An interesting concept that emerges from these studies is A2A R's ability to fine tune neuronal and glial functions to produce neuroprotective effects. While the data presented here clearly highlight the complexity of using adenosinergic agents therapeutically in PD and other neurodegenerative disorders and point out many areas for further inquiry, they also confirm that adenosine receptor ligands, particularly A2A receptor ligands, have many promising characteristics that encourage the pursuit of their therapeutic potential.


Adenosine/metabolism , Brain Diseases/metabolism , Brain/metabolism , Cytoprotection/physiology , Nerve Degeneration/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Brain/drug effects , Brain/physiopathology , Brain Diseases/drug therapy , Brain Diseases/genetics , Brain Injury, Chronic/genetics , Brain Injury, Chronic/metabolism , Brain Injury, Chronic/physiopathology , Cell Survival/drug effects , Cell Survival/genetics , Cytoprotection/drug effects , Humans , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neuroprotective Agents/pharmacology
15.
J Pharmacol Exp Ther ; 320(2): 747-56, 2007 Feb.
Article En | MEDLINE | ID: mdl-17090704

Endogenous striatal dopamine (DA) overflow has been associated with neuropathological conditions resulting from ischemia, psychostimulants, and metabolic inhibition. Malonate, a reversible inhibitor of succinate dehydrogenase, models the effects of energy impairment in neurodegenerative disorders. We have previously reported that the striatal DA efflux and damage to DA nerve terminals resulting from intrastriatal malonate infusions is prevented by prior DA depletion, suggesting that DA plays a role in the neuronal damage. We presently report that the malonate-induced DA efflux is partially mediated by reverse transport of DA from the cytosol to the extracellular space via the DA transporter (DAT). Pharmacological blockade of the DAT with a series of structurally different inhibitors [cocaine, mazindol, 1-(2-(bis(4-fluophenyl methoxy) ethyl)-4-(3-(4-fluorophenyl)-propyl)piperazine) dimethane sulfonate (GBR 13098) and methyl(-)-3beta-(p-fluorophenyl)-1alphaH,5alphaH-tropane-2beta-carboxylate1,5-naphthalene (Win 35,428)] attenuated malonate-induced DA overflow in vivo and protected mice against subsequent damage to DA nerve terminals. Consistent with these findings, the DAT inhibitors prevented malonate-induced damage to DA neurons in mesencephalic cultures and also protected against the loss of GABA neurons in this system. The DAT inhibitors did not modify malonate-induced formation of reactive oxygen species or lactate production, indicating that the DAT inhibitors neither exert antioxidant effects nor interfere with the actions of malonate. Taken together, these findings provide direct evidence that mitochondrial impairment and metabolic stress cause striatal DA efflux via the DAT and suggest that disruptions in DA homeostasis resulting from energy impairment may contribute to the pathogenesis of neurodegenerative diseases.


Corpus Striatum/drug effects , Dopamine Plasma Membrane Transport Proteins/physiology , Dopamine/metabolism , Malonates/toxicity , Mesencephalon/drug effects , Mitochondria/drug effects , Animals , Corpus Striatum/pathology , Free Radicals , Lactic Acid/biosynthesis , Male , Mesencephalon/pathology , Mice , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/analysis , gamma-Aminobutyric Acid/physiology
16.
J Neurosci ; 23(34): 10982-7, 2003 Nov 26.
Article En | MEDLINE | ID: mdl-14645494

Mitochondrial dysfunction may contribute to dopaminergic (DAergic) cell death in Parkinson's disease and GABAergic cell death in Huntington's disease. In the present work, we tested whether blocking A1 receptors could enhance the damage to DAergic and GABAergic neurons caused by mitochondrial inhibition, and whether blocking A2a receptors could protect against damage in this model. Animals received an intraperitoneal injection of 8-cyclopentyl-1,3-dipropylxanthine (CPX) (A1 antagonist) or 3,7-dimethyl-1-propargylxanthine (DMPX) (A2a antagonist) 30 min before intrastriatal infusion of malonate (mitochondrial complex II inhibitor). Damage was assessed 1 week later by measuring striatal dopamine, tyrosine hydroxylase (TH), and GABA content. In mice and rats, malonate-induced depletion of striatal dopamine, TH, or GABA was potentiated by pretreatment with 1 mg/kg CPX and attenuated by pretreatment with 5 mg/kg DMPX. To determine the location of the A1 and A2a receptors mediating these effects, CPX or DMPX was infused directly into the striatum or substantia nigra of rats 30 min before intrastriatal infusion of malonate. When infused into the striatum, CPX (20 ng) potentiated, whereas DMPX (50 ng) prevented malonate-induced GABA loss, but up to 100 ng of CPX or 500 ng of DMPX did not alter malonate-induced striatal dopamine loss. Intranigral infusion of CPX (100 ng) or DMPX (500 ng), however, did exacerbate and protect, respectively, against malonate-induced striatal dopamine loss. Thus, A1 receptor blockade enhances and A2a receptor blockade protects against damage to DAergic and GABAergic neurons caused by mitochondrial inhibition. Interestingly, these effects are mediated by A1 and A2a receptors located in the substantia nigra for DAergic neurons and in the striatum for GABAergic neurons.


Adenosine/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Mitochondria/metabolism , Substantia Nigra/metabolism , Theobromine/analogs & derivatives , gamma-Aminobutyric Acid/metabolism , Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Antagonists , Animals , Corpus Striatum/drug effects , Drug Administration Routes , Drug Synergism , Electron Transport Complex II/antagonists & inhibitors , Huntington Disease/metabolism , Male , Malonates/pharmacology , Mice , Mitochondria/drug effects , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/metabolism , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Theobromine/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Xanthines/pharmacology
17.
J Comp Neurol ; 452(1): 24-37, 2002 Oct 07.
Article En | MEDLINE | ID: mdl-12205707

Firing during sensorimotor exam was used to categorize single neurons in the lateral striatum of awake, unrestrained rats. Five rats received unilateral injection of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle to deplete striatal dopamine (DA; >98% depletion, postmortem assay). Three months after treatment, rats exhibited exaggerated rotational behavior induced by L-dihydroxyphenylalanine (L-DOPA) and contralateral sensory neglect. Electrode track "depth profiles" on the DA-depleted side showed fragmented clustering of neurons related to sensorimotor activity of single body parts (SBP neurons). Clusters were smaller than normal, and more SBP neurons were observed in isolation, outside of clusters. More body parts were represented per unit volume. No recovery in these measures was observed up to one year post lesion. Overall distributions of neurons related to different body parts were not altered. The fragmentation of SBP clusters after DA depletion indicates that a percentage of striatal SBP neurons switched responsiveness from one body part to one or more different body parts. Because the specific firing that characterizes striatal SBP neurons is mediated by corticostriatal inputs (Liles and Updyke [1985] Brain Res. 339:245-255), the data indicate that DA depletion resulted in a reorganization of corticostriatal connections, perhaps via unmasking or sprouting of connections to adjacent clusters of striatal neurons. After reorganization, sensory activity in a localized body part activates striatal neurons that have switched to that body part. In turn, switched signals sent from basal ganglia to premotor and motor neurons, which likely retain their original connections, would create mismatches in these normally precise topographic connections. Switched signals could partially explain parkinsonian deficits in motor functions involving somatosensory guidance and their intractability to L-DOPA therapy-particularly if the switching involves sprouting.


Corpus Striatum/physiopathology , Dopamine/deficiency , Neurons/pathology , Neurons/physiology , Parkinsonian Disorders/physiopathology , Adrenergic Agents/pharmacology , Animals , Behavior, Animal/physiology , Dopamine/metabolism , Electrophysiology , Male , Oxidopamine/pharmacology , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Serotonin/metabolism
18.
Exp Neurol ; 176(1): 193-202, 2002 Jul.
Article En | MEDLINE | ID: mdl-12093096

Previous work has shown that overstimulation of GABA(A) receptors can potentiate neuronal cell damage during excitotoxic or metabolic stress in vitro and that GABA(A) antagonists or GABA transport blockers are neuroprotective under these situations. Malonate, a reversible succinate dehydrogenase/mitochondrial complex II inhibitor, is frequently used in animals to model cell loss in neurodegenerative diseases such as Parkinson's and Huntington's diseases. To determine if GABA transporter blockade during mitochondrial impairment can protect neurons in vivo as compared with in vitro studies, rats received a stereotaxic infusion of malonate (2 micromol) into the left striatum to induce a metabolic stress. The nonsubstrate GABA transport blocker, NO711 (20 nmol) was infused in some rats 30 min before and 3 h following malonate infusion. After 1 week, dopamine and GABA levels in the striata were measured. Malonate caused a significant loss of striatal dopamine and GABA. Blockade of the GABA transporter significantly attenuated GABA, but not dopamine loss. In contrast with several in vitro reports, GABA(A) receptors were not a downstream mediator of protection by NO711. Intrastriatal infusion of malonate (2 micromol) plus or minus the GABA(A) receptor agonist muscimol (1 micromol), the GABA(A) Cl- binding site antagonist picrotoxin (50 nmol) or the GABA(B) receptor antagonist saclofen (33 nmol) did not modify loss of striatal dopamine or GABA when examined 1 week following infusion. These data show that GABA transporter blockade during mitochondrial impairment in the striatum provides protection to GABAergic neurons. GABA transporter blockade, which is currently a pharmacological strategy for the treatment of epilepsy, may thus also be beneficial in the treatment of acute and chronic conditions involving energy inhibition such as stroke/ischemia or Huntington's disease. These findings also point to fundamental differences between immature and adult neurons in the downstream involvement of GABA receptors during metabolic insult.


Carrier Proteins/antagonists & inhibitors , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Malonates/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Transport Proteins , Nipecotic Acids/pharmacology , Organic Anion Transporters , Oximes/pharmacology , Animals , Catheterization , Corpus Striatum/chemistry , Dopamine/analysis , Dopamine/metabolism , Drug Administration Routes , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , GABA Plasma Membrane Transport Proteins , GABA-A Receptor Agonists , GABA-A Receptor Antagonists , Male , Malonates/administration & dosage , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Nipecotic Acids/administration & dosage , Oximes/administration & dosage , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/analysis , gamma-Aminobutyric Acid/metabolism
19.
J Biol Chem ; 277(39): 36040-4, 2002 Sep 27.
Article En | MEDLINE | ID: mdl-12130655

Caffeine and more specific antagonists of the adenosine A(2A) receptor recently have been found to be neuroprotective in the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) model of Parkinson's disease. Here we show that 8-(3-chlorostyryl)caffeine (CSC), a specific A(2A) antagonist closely related to caffeine, also attenuates MPTP-induced neurotoxicity. Because the neurotoxicity of MPTP relies on its oxidative metabolism to the mitochondrial toxin MPP(+), we investigated the actions of CSC on striatal MPTP metabolism in vivo. CSC elevated striatal levels of MPTP but lowered levels of the oxidative intermediate MPDP(+) and of MPP(+), suggesting that CSC blocks the conversion of MPTP to MPDP(+) in vivo. In assessing the direct effects of CSC and A(2A) receptors on monoamine oxidase (MAO) activity, we found that CSC potently and specifically inhibited mouse brain mitochondrial MAO-B activity in vitro with a K(i) value of 100 nm, whereas caffeine and another relatively specific A(2A) antagonist produced little or no inhibition. The A(2A) receptor independence of MAO-B inhibition by CSC was further supported by the similarity of brain MAO activities derived from A(2A) receptor knockout and wild-type mice and was confirmed by demonstrating potent inhibition of A(2A) receptor knockout-derived MAO-B by CSC. Together, these data indicate that CSC possesses dual actions of MAO-B inhibition and A(2A) receptor antagonism, a unique combination suggesting a new class of compounds with the potential for enhanced neuroprotective properties.


1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Caffeine/analogs & derivatives , Caffeine/pharmacology , Dopamine Agents/pharmacology , Drug Interactions , Monoamine Oxidase Inhibitors/pharmacology , Neurons/drug effects , Receptors, Purinergic P1/metabolism , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A , Time Factors
...