Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mov Disord ; 34(5): 708-716, 2019 05.
Article in English | MEDLINE | ID: mdl-30575996

ABSTRACT

BACKGROUND: Pridopidine, in development for Huntington's disease, may modulate aberrant l-dopa-induced effects including l-dopa-induced dyskinesia (LID). OBJECTIVE: This study investigated whether pridopidine could reduce LID in the MPTP macaque model of Parkinson's disease and characterized the observed behavioral effects in terms of receptor occupancy. METHODS: The pharmacokinetic profile and effects of pridopidine (15-30 mg/kg) on parkinsonism, dyskinesia, and quality of on-time, in combination with l-dopa, were assessed in MPTP macaques with LID. Pridopidine receptor occupancy was estimated using known in vitro binding affinities to σ1 and dopamine D2 receptors, in vivo PET imaging, and pharmacokinetic profiling across different species. RESULTS: Pridopidine produced a dose-dependent reduction in dyskinesia (up to 71%, 30 mg/kg) and decreased the duration of on-time with disabling dyskinesia evoked by l-dopa by 37% (20 mg/kg) and 60% (30 mg/kg). Pridopidine did not compromise the anti-parkinsonian benefit of l-dopa. Plasma exposures following the ineffective dose (15 mg/kg) were associated with full σ1 occupancy (>80%), suggesting that σ1 engagement alone is unlikely to account for the antidyskinetic benefits of pridopidine. Exposures following effective doses (20-30 mg/kg), while providing full σ1 occupancy, provide only modest dopamine D2 occupancy (<40%). However, effective pridopidine doses clearly engage a range of receptors (including adrenergic-α2C , dopamine-D3 , and serotoninergic-5-HT1A sites) to a higher degree than D2 and might contribute to the antidyskinetic actions. CONCLUSIONS: In MPTP macaques, pridopidine produced a significant decrease in LID without compromising the antiparkinsonian benefit of l-dopa. Although the actions of pridopidine were associated with full σ1 occupancy, effective exposures are more likely associated with occupancy of additional, non-sigma receptors. This complex pharmacology may underlie the effectiveness of pridopidine against LID. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , MPTP Poisoning/drug therapy , Movement/drug effects , Parkinsonian Disorders/drug therapy , Piperidines/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Brain/diagnostic imaging , Brain/metabolism , Dyskinesia, Drug-Induced/etiology , Macaca fascicularis , Parkinsonian Disorders/chemically induced , Positron-Emission Tomography , Receptor, Muscarinic M2/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Receptors, Histamine H3/metabolism , Receptors, sigma/metabolism , Sigma-1 Receptor
2.
Blood Cells Mol Dis ; 63: 62-65, 2017 03.
Article in English | MEDLINE | ID: mdl-28178599

ABSTRACT

Dexpramipexole, an orally bioavailable small molecule previously under clinical development in amyotrophic lateral sclerosis, was observed during routine safety hematology monitoring to demonstrate pronounced, dose- and time-dependent eosinophil-lowering effects, with minor reductions on other leukocyte counts. Analysis of hematology lab values across two double-blind, randomized placebo-controlled clinical trials at total daily doses ranging from 50mg to 300mg demonstrated that dexpramipexole consistently and markedly lowered peripheral blood eosinophils. This effect developed after 1month on treatment, required 3-4months to reach its maximum, remained constant throughout treatment, and partially recovered to baseline levels upon drug withdrawal. All doses tested were well tolerated. The overall adverse event rate was similar for dexpramipexole and placebo, and notably with no increase in infection-related adverse events associated with eosinophil-lowering effects. Given the reliance on and insufficiency of off-label chronic corticosteroid therapy for hypereosinophilic syndromes and other eosinophilic-associated diseases (EADs), a need exists for less toxic, more effective, targeted therapeutic alternatives. Further clinical studies are underway to assess the eosinophil-lowering effect of dexpramipexole in the peripheral blood and target tissues of EAD patients and whether such reductions, if observed, produce clinically important benefits.


Subject(s)
Benzothiazoles/pharmacology , Eosinophils/drug effects , Benzothiazoles/adverse effects , Benzothiazoles/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Eosinophilia/drug therapy , Eosinophils/cytology , Humans , Hypereosinophilic Syndrome/drug therapy , Infections/chemically induced , Leukocyte Count , Pramipexole
3.
Hum Mol Genet ; 21(1): 163-74, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21972245

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 gene (LRRK2) are the most common genetic cause of Parkinson's disease (PD) and cause both autosomal dominant familial and sporadic PD. Currently, the physiological and pathogenic activities of LRRK2 are poorly understood. To decipher the biological functions of LRRK2, including the genes and pathways modulated by LRRK2 kinase activity in vivo, we assayed genome-wide mRNA expression in the brain and peripheral tissues from LRRK2 knockout (KO) and kinase hyperactive G2019S (G2019S) transgenic mice. Subtle but significant differences in mRNA expression were observed relative to wild-type (WT) controls in the cortex, striatum and kidney of KO animals, but only in the striatum in the G2019S model. In contrast, robust, consistent and highly significant differences were identified by the direct comparison of KO and G2019S profiles in the cortex, striatum, kidney and muscle, indicating opposite effects on mRNA expression by the two models relative to WT. Ribosomal and glycolytic biological functions were consistently and significantly up-regulated in LRRK2 G2019S compared with LRRK2 KO tissues. Genes involved in membrane-bound organelles, oxidative phosphorylation, mRNA processing and the endoplasmic reticulum were down-regulated in LRRK2 G2019S mice compared with KO. We confirmed the expression patterns of 35 LRRK2-regulated genes using quantitative reverse transcription polymerase chain reaction. These findings provide the first description of the transcriptional responses to genetically modified LRRK2 activity and provide preclinical target engagement and/or pharmacodynamic biomarker strategies for LRRK2 and may inform future therapeutic strategies for LRRK2-associated PD.


Subject(s)
Parkinson Disease/enzymology , Parkinson Disease/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Animals , Brain/enzymology , Female , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics
4.
J Pharmacol Exp Ther ; 350(3): 495-505, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24947466

ABSTRACT

There is growing evidence that activation of metabotropic glutamate receptor 4 (mGlu4) leads to anxiolytic- and antipsychotic-like efficacy in rodent models, yet its relevance to depression-like reactivity remains unclear. Here, we present the pharmacological evaluation of ADX88178 [5-methyl-N-(4-methylpyrimidin-2-yl)-4-(1H-pyrazol-4-yl)thiazol-2-amine], a novel potent, selective, and brain-penetrant positive allosteric modulator of the mGlu4 receptor in rodent models of anxiety, obsessive compulsive disorder (OCD), fear, depression, and psychosis. ADX88178 dose-dependently reduced the number of buried marbles in the marble burying test and increased open-arm exploration in the elevated plus maze (EPM) test, indicative of anxiolytic-like efficacy. Target specificity of the effect in the EPM test was confirmed using male and female mGlu4 receptor knockout mice. In mice, ADX88178 reduced the likelihood of conditioned freezing in the acquisition phase of the fear conditioning test, yet had no carryover effect in the expression phase. Also, ADX88178 dose-dependently reduced duration of immobility in the forced swim test, indicative of antidepressant-like efficacy. ADX88178 reduced DOI (2,5-dimethoxy-4-iodoamphetamine)-mediated head twitches (albeit with no dose-dependency), and MK-801 [(5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine]-induced locomotor hyperactivity in mice, but was inactive in the conditioned avoidance response test in rats. The compound showed good specificity as it had no effect on locomotor activity in mice and rats at efficacious doses. Thus, allosteric activation of mGlu4 receptors can be a promising new therapeutic approach for treatment of anxiety, OCD, fear-related disorders, and psychosis.


Subject(s)
Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/therapeutic use , Disease Models, Animal , Mental Disorders/drug therapy , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/therapeutic use , Thiazoles/chemistry , Thiazoles/therapeutic use , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Anti-Anxiety Agents/pharmacology , Female , Male , Mental Disorders/metabolism , Mental Disorders/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Pyrimidines/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/physiology , Thiazoles/metabolism
5.
J Biol Chem ; 286(7): 5215-21, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21118811

ABSTRACT

Apolipoprotein (apo) E4 is the major genetic risk factor for late-onset Alzheimer disease (AD). ApoE4 assumes a pathological conformation through an intramolecular interaction mediated by Arg-61 in the amino-terminal domain and Glu-255 in the carboxyl-terminal domain, referred to as apoE4 domain interaction. Because AD is associated with mitochondrial dysfunction, we examined the effect of apoE4 domain interaction on mitochondrial respiratory function. Steady-state amounts of mitochondrial respiratory complexes were examined in neurons cultured from brain cortices of neuron-specific enolase promoter-driven apoE3 (NSE-apoE3) or apoE4 (NSE-apoE4) transgenic mice. All subunits of mitochondrial respiratory complexes assessed were significantly lower in NSE-apoE4 neurons compared with NSE-apoE3 neurons. However, no significant differences in levels of mitochondrial complexes were detected between astrocytes expressing different apoE isoforms driven by the glial fibrillary acidic protein promoter, leading to our conclusion that the effect of apoE4 is neuron specific. In neuroblastoma Neuro-2A (N2A) cells, apoE4 expression reduced the levels of mitochondrial respiratory complexes I, IV, and V. Complex IV enzymatic activity was also decreased, lowering mitochondrial respiratory capacity. Mutant apoE4 (apoE4-Thr-61) lacking domain interaction did not induce mitochondrial dysfunction in N2A cells, indicating that the effect is specific to apoE4-expressing cells and dependent on domain interaction. Consistent with this finding, treatment of apoE4-expressing N2A cells with a small molecule that disrupts apoE4 domain interaction restored mitochondrial respiratory complex IV levels. These results suggest that pharmacological intervention with small molecules that disrupt apoE4 domain interaction is a potential therapeutic approach for apoE4-carrying AD subjects.


Subject(s)
Alzheimer Disease/metabolism , Apolipoprotein E4/metabolism , Mitochondria/metabolism , Neurons/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Apolipoprotein E4/genetics , Cell Line, Tumor , Electron Transport/genetics , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Neurons/pathology , Protein Structure, Tertiary , Risk Factors
6.
J Pharmacol Exp Ther ; 343(1): 167-77, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22787118

ABSTRACT

Positive allosteric modulators (PAMs) of metabotropic glutamate receptor 4 (mGluR4) have been proposed as a novel therapeutic approach for the treatment of Parkinson's disease. However, evaluation of this proposal has been limited by the availability of appropriate pharmacological tools to interrogate the target. In this study, we describe the properties of a novel mGluR4 PAM. 5-Methyl-N-(4-methylpyrimidin-2-yl)-4-(1H-pyrazol-4-yl)thiazol-2-amine (ADX88178) enhances glutamate-mediated activation of human and rat mGluR4 with EC(50) values of 4 and 9 nM, respectively. The compound is highly selective for mGluR4 with minimal activities at other mGluRs. Oral administration of ADX88178 in rats is associated with high bioavailability and results in cerebrospinal fluid exposure of >50-fold the in vitro EC(50) value. ADX88178 reverses haloperidol-induced catalepsy in rats at 3 and 10 mg/kg. It is noteworthy that this compound alone has no impact on forelimb akinesia resulting from a bilateral 6-hydroxydopamine lesion in rats. However, coadministration of a low dose of L-DOPA (6 mg/kg) enabled a robust, dose-dependent reversal of the forelimb akinesia deficit. ADX88178 also increased the effects of quinpirole in lesioned rats and enhanced the effects of L-DOPA in MitoPark mice. It is noteworthy that the enhancement of the actions of L-DOPA was not associated with an exacerbation of L-DOPA-induced dyskinesias in rats. ADX88178 is a novel, potent, and selective mGluR4 PAM that is a valuable tool for exploring the therapeutic potential of mGluR4 modulation. The use of this novel tool molecule supports the proposal that activation of mGluR4 may be therapeutically useful in Parkinson's disease.


Subject(s)
Disease Models, Animal , Excitatory Amino Acid Agonists/therapeutic use , Parkinson Disease/drug therapy , Receptors, Metabotropic Glutamate/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Excitatory Amino Acid Agonists/pharmacology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parkinson Disease/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/biosynthesis
7.
Biochim Biophys Acta ; 1802(1): 143-50, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19747973

ABSTRACT

Alterations in mitochondrial function may have a central role in the pathogenesis of many neurodegenerative diseases. The study of mitochondrial dysfunction has typically focused on ATP generation, calcium homeostasis and the production of reactive oxygen species. However, there is a growing appreciation of the dynamic nature of mitochondria within cells. Mitochondria are highly motile organelles, and also constantly undergo fission and fusion. This raises the possibility that impairment of mitochondrial dynamics could contribute to the pathogenesis of neuronal injury. In this review we describe the mechanisms that govern mitochondrial movement, fission and fusion. The key proteins that are involved in mitochondrial fission and fusion have also been linked to some inherited neurological diseases, including autosomal dominant optic atrophy and Charcot-Marie-Tooth disease 2A. We will discuss the evidence that altered movement, fission and fusion are associated with impaired neuronal viability. There is a growing collection of literature that links impaired mitochondrial dynamics to a number of disease models. Additionally, the concept that the failure to deliver a functional mitochondrion to the appropriate site within a neuron could contribute to neuronal dysfunction provides an attractive framework for understanding the mechanisms underlying neurologic disease. However, it remains difficult to clearly establish that altered mitochondrial dynamics clearly represent a cause of neuronal dysfunction.


Subject(s)
Mitochondria/metabolism , Mitochondria/ultrastructure , Neurodegenerative Diseases/pathology , Neurons/ultrastructure , Adenosine Triphosphate/metabolism , Animals , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurons/metabolism
8.
J Neurochem ; 118(6): 1016-31, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21736568

ABSTRACT

The EphA4 receptor and its ephrin ligands are involved in astrocytic gliosis following CNS injury. Therefore, a strategy aimed at the blockade of EphA4 signaling could have broad therapeutic interest in brain disorders. We have identified novel small molecule inhibitors of EphA4 kinase in specific enzymatic and cell-based assays. In addition, we have demonstrated in two in vitro models of scratch injury that EphA4 receptor kinase is activated through phosphorylation and is involved in the repopulation of the wound after the scratch. A potent EphA4 kinase inhibitor significantly inhibited wound closure and reduced the accumulation of the reactive astrocytes inside the scratch. We have also shown that after the transient focal cerebral ischemia in rats, a large glial scar is formed by the accumulation of astrocytes and chondroitin sulfate proteoglycan surrounding the infarcted tissue at 7 days and 14 days of reperfusion. EphA4 protein expression is highly up-regulated in the same areas at these time points, supporting its potential role in the glial scar formation and maintenance. Taken together, these results suggest that EphA4 kinase inhibitors might interfere with the astrogliosis reaction and thereby lead to improved neurological outcome after ischemic injury.


Subject(s)
Gliosis/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptor, EphA4/antagonists & inhibitors , Wounds and Injuries/pathology , Animals , Astrocytes/pathology , Blotting, Western , CHO Cells , Cell Movement/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Gliosis/pathology , Humans , Immunohistochemistry , Ischemic Attack, Transient/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley , Small Molecule Libraries , Wound Healing/drug effects
9.
Biomedicines ; 9(5)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946782

ABSTRACT

Zinc is a highly abundant cation in the brain, essential for cellular functions, including transcription, enzymatic activity, and cell signaling. However, zinc can also trigger injurious cascades in neurons, contributing to the pathology of neurodegenerative diseases. Mitochondria, critical for meeting the high energy demands of the central nervous system (CNS), are a principal target of the deleterious actions of zinc. An increasing body of work suggests that intracellular zinc can, under certain circumstances, contribute to neuronal damage by inhibiting mitochondrial energy processes, including dissipation of the mitochondrial membrane potential (MMP), leading to ATP depletion. Additional consequences of zinc-mediated mitochondrial damage include reactive oxygen species (ROS) generation, mitochondrial permeability transition, and excitotoxic calcium deregulation. Zinc can also induce mitochondrial fission, resulting in mitochondrial fragmentation, as well as inhibition of mitochondrial motility. Here, we review the known mechanisms responsible for the deleterious actions of zinc on the organelle, within the context of neuronal injury associated with neurodegenerative processes. Elucidating the critical contributions of zinc-induced mitochondrial defects to neurotoxicity and neurodegeneration may provide insight into novel therapeutic targets in the clinical setting.

10.
Front Neurosci ; 14: 778, 2020.
Article in English | MEDLINE | ID: mdl-32792905

ABSTRACT

This special issue of Frontiers in Neuroscience-Neurodegeneration celebrates the 50th anniversary of John Olney's seminal work introducing the concept of excitotoxicity as a mechanism for neuronal cell death. Since that time, fundamental research on the pathophysiological activation of glutamate receptors has played a central role in our understanding of excitotoxic cellular signaling pathways, leading to the discovery of many potential therapeutic targets in the treatment of acute or chronic/progressive neurodegenerative disorders. Importantly, excitotoxic signaling processes have been found repeatedly to be closely intertwined with oxidative cellular cascades. With this in mind, this review looks back at long-standing collaborative efforts by the authors linking cellular redox status and glutamate neurotoxicity, focusing first on the discovery of the redox modulatory site of the N-methyl-D-aspartate (NMDA) receptor, followed by the study of the oxidative conversion of 3,4-dihydroxyphenylalanine (DOPA) to the non-NMDA receptor agonist and neurotoxin 2,4,5-trihydroxyphenylalanine (TOPA) quinone. Finally, we summarize our work linking oxidative injury to the liberation of zinc from intracellular metal binding proteins, leading to the uncovering of a signaling mechanism connecting excitotoxicity with zinc-activated cell death-signaling cascades.

11.
J Neurochem ; 106(5): 2184-93, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18624907

ABSTRACT

Liberation of zinc from intracellular stores contributes to oxidant-induced neuronal injury. However, little is known regarding how endogenous oxidant systems regulate intracellular free zinc ([Zn(2+)](i)). Here we simultaneously imaged [Ca(2+)](i) and [Zn(2+)](i) to study acute [Zn(2+)](i) changes in cultured rat forebrain neurons after glutamate receptor activation. Neurons were loaded with fura-2FF and FluoZin-3 to follow [Ca(2+)](i) and [Zn(2+)](i), respectively. Neurons treated with glutamate (100 microM) for 10 min gave large Ca(2+) responses that did not recover after termination of the glutamate stimulus. Glutamate also increased [Zn(2+)](i), however glutamate-induced [Zn(2+)](i) changes were completely dependent on Ca(2+) entry, appeared to arise entirely from internal stores, and were substantially reduced by co-application of the membrane-permeant chelator TPEN during the glutamate treatment. Pharmacological maneuvers revealed that a number of endogenous oxidant producing systems, including nitric oxide synthase, phospholipase A(2), and mitochondria all contributed to glutamate-induced [Zn(2+)](i) changes. We found no evidence that mitochondria buffered [Zn(2+)](i) during acute glutamate receptor activation. We conclude that glutamate-induced [Zn(2+)](i) transients are caused in part by [Ca(2+)](i)-induced reactive oxygen species that arises from both cytosolic and mitochondrial sources.


Subject(s)
Calcium Signaling/physiology , Glutamic Acid/metabolism , Nerve Degeneration/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Zinc/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Cytosol/drug effects , Cytosol/metabolism , Fluorescent Dyes , Fura-2 , Glutamic Acid/pharmacology , Intracellular Fluid/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/metabolism , Oxidants/biosynthesis , Oxidative Stress/drug effects , Polycyclic Compounds , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Zinc/pharmacology
12.
Prog Neurobiol ; 80(5): 241-68, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17188795

ABSTRACT

Mitochondria are the primary generators of ATP and are important regulators of intracellular calcium homeostasis. These organelles are dynamically transported along lengthy neuronal processes, presumably for appropriate distribution to cellular regions of high metabolic demand and elevated intracellular calcium, such as synapses. The removal of damaged mitochondria that produce harmful reactive oxygen species and promote apoptosis is also thought to be mediated by transport of mitochondria to autophagosomes. Mitochondrial trafficking is therefore important for maintaining neuronal and mitochondrial health while cessation of movement may lead to neuronal and mitochondrial dysfunction. Mitochondrial morphology is also dynamic and is remodeled during neuronal injury and disease. Recent studies reveal different manifestations and mechanisms of impaired mitochondrial movement and altered morphology in injured neurons. These are likely to cause varied courses toward neuronal degeneration and death. The goal of this review is to provide an appreciation of the full range of mitochondrial function, morphology and trafficking, and the critical role these parameters play in neuronal physiology and pathophysiology.


Subject(s)
Biological Transport/physiology , Mitochondria/pathology , Mitochondria/physiology , Neurons , Animals , Brain/pathology , Brain/physiology , Brain Injuries/pathology , Brain Injuries/physiopathology , Calcium/metabolism , Humans , Neurons/pathology , Neurons/physiology , Neurons/ultrastructure , Reactive Oxygen Species/metabolism
13.
J Neurosci ; 26(26): 7035-45, 2006 Jun 28.
Article in English | MEDLINE | ID: mdl-16807333

ABSTRACT

Functional synapses require mitochondria to supply ATP and regulate local [Ca2+]i for neurotransmission. Mitochondria are thought to be transported to specific cellular regions of increased need such as synapses. However, little is known about how this occurs, including the spatiotemporal distribution of mitochondria relative to presynaptic and postsynaptic sites, whether mitochondria are dynamically recruited to synapses, and how synaptic activity affects these trafficking patterns. We used primary cortical neurons in culture that form synaptic connections and show spontaneous synaptic activity under normal conditions. Neurons were cotransfected with a mitochondrially targeted cyan fluorescent protein and an enhanced yellow fluorescent protein-tagged synaptophysin or postsynaptic density-95 plasmid to label presynaptic or postsynaptic structures, respectively. Fluorescence microscopy revealed longer dendritic mitochondria that occupied a greater fraction of neuronal process length than axonal mitochondria. Mitochondria were significantly more likely to be localized at synaptic sites. Although this localization was unchanged by inhibition of synaptic activity by tetrodotoxin, it increased in dendritic synapses and decreased in axonal synapses during overactivity by veratridine. Mitochondrial movement and recruitment to synapses also differed between axons and dendrites under basal conditions and when synaptic activity was altered. Additionally, we show that movement of dendritic mitochondria can be selectively impaired by glutamate and zinc. We conclude that mitochondrial trafficking to synapses is dynamic in neurons and is modulated by changes in synaptic activity. Furthermore, mitochondrial morphology and distribution may be optimized differentially to best serve the synaptic distributions in axons and dendrites. Last, selective cessation of mitochondrial movement in dendrites suggests early postsynaptic dysfunction in neuronal injury and degeneration.


Subject(s)
Cerebral Cortex/physiology , Mitochondria/physiology , Neurons/physiology , Synapses/physiology , Animals , Axons/ultrastructure , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/ultrastructure , Dendrites/ultrastructure , Mitochondria/drug effects , Mitochondria/ultrastructure , Neurons/ultrastructure , Neurotoxins/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors
14.
J Clin Invest ; 113(7): 981-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15057304

ABSTRACT

HIV infection in humans and simian immunodeficiency virus (SIV) infection in macaques result in encephalitis in approximately one-quarter of infected individuals and is characterized by infiltration of the brain with infected and activated macrophages. 1-(2-chlorphenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinoline-carboxamide (PK11195) is a ligand specific for the peripheral benzodiazepine receptor abundant on macrophages and is expressed in low levels in the noninfected brain. We hypothesized that positron-emission tomography (PET) with the carbon-11-labeled, R-enantiomer form of PK11195 ([(11)C](R)-PK11195) could image brain macrophages and hence the development of encephalitis in vivo. [(11)C](R)-PK11195 binding was assessed in the brain using PET in 11 SIV infected macaques, six of which showed increased binding in vivo. Postmortem examination of the brain in these six macaques demonstrated encephalitis, while macaques that did not show an increase in [(11)C](R)-PK11195 binding did not develop SIV encephalitis. Brain tissue from SIV encephalitic macaques also showed increased [(3)H](R)-PK11195 binding compared with binding in nonencephalitic macaques. Increased PK11195 binding in vivo and in postmortem brain tissue correlated with abundance of macrophages but not astrocytes. Our results suggest that PET [(11)C](R)-PK11195 imaging can detect the presence of macrophages in SIV encephalitis in vivo and may be useful to predict the development of HIV encephalitis and in studies of the pathogenesis and treatment of HIV dementia.


Subject(s)
Brain/diagnostic imaging , Macaca/metabolism , Macrophages/diagnostic imaging , Receptors, GABA-A/metabolism , Tomography, Emission-Computed , Acquired Immunodeficiency Syndrome/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Macrophages/metabolism , Magnetic Resonance Imaging , Simian Immunodeficiency Virus/metabolism
15.
J Cell Biol ; 216(4): 1091-1105, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28254829

ABSTRACT

Glutamate is the dominant excitatory neurotransmitter in the brain, but under conditions of metabolic stress it can accumulate to excitotoxic levels. Although pharmacologic modulation of excitatory amino acid receptors is well studied, minimal consideration has been given to targeting mitochondrial glutamate metabolism to control neurotransmitter levels. Here we demonstrate that chemical inhibition of the mitochondrial pyruvate carrier (MPC) protects primary cortical neurons from excitotoxic death. Reductions in mitochondrial pyruvate uptake do not compromise cellular energy metabolism, suggesting neuronal metabolic flexibility. Rather, MPC inhibition rewires mitochondrial substrate metabolism to preferentially increase reliance on glutamate to fuel energetics and anaplerosis. Mobilizing the neuronal glutamate pool for oxidation decreases the quantity of glutamate released upon depolarization and, in turn, limits the positive-feedback cascade of excitotoxic neuronal injury. The finding links mitochondrial pyruvate metabolism to glutamatergic neurotransmission and establishes the MPC as a therapeutic target to treat neurodegenerative diseases characterized by excitotoxicity.


Subject(s)
Cell Death/physiology , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Neurons/metabolism , Neurons/physiology , Pyruvic Acid/metabolism , Animals , Energy Metabolism/physiology , Glutamic Acid/metabolism , Mitochondrial Proteins , Monocarboxylic Acid Transporters , Neurodegenerative Diseases/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Solute Carrier Proteins
16.
J Neurosci ; 25(41): 9507-14, 2005 Oct 12.
Article in English | MEDLINE | ID: mdl-16221861

ABSTRACT

Mitochondria have been identified as targets of the neurotoxic actions of zinc, possibly through decreased mitochondrial energy production and increased reactive oxygen species accumulation. It has been hypothesized that impairment of mitochondrial trafficking may be a mechanism of neuronal injury. Here, we report that elevated intraneuronal zinc impairs mitochondrial trafficking. At concentrations just sufficient to cause injury, zinc rapidly inhibited mitochondrial movement without altering morphology. Zinc chelation initially restored movement, but the actions of zinc became insensitive to chelator in <10 min. A search for downstream signaling events revealed that inhibitors of phosphatidylinositol (PI) 3-kinase prevented this zinc effect on movement. Moreover, transient inhibition of PI 3-kinase afforded neuroprotection against zinc-mediated toxicity. These data illustrate a novel mechanism that regulates mitochondrial trafficking in neurons and also suggest that mitochondrial trafficking may be closely coupled to neuronal viability.


Subject(s)
Enzyme Activation/physiology , Mitochondria/drug effects , Mitochondria/enzymology , Neurons/drug effects , Neurons/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Zinc/pharmacology , Animals , Cations, Divalent , Cells, Cultured , Chlorides/pharmacology , Chlorides/physiology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Neural Inhibition/drug effects , Neural Inhibition/physiology , Rats , Rats, Sprague-Dawley , Zinc/physiology , Zinc Compounds/pharmacology
17.
Biochim Biophys Acta ; 1709(2): 127-37, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16112074

ABSTRACT

Ca(2+)-induced mitochondrial depolarization was studied in single isolated rat brain and liver mitochondria. Digital imaging techniques and rhodamine 123 were used for mitochondrial membrane potential measurements. Low Ca(2+) concentrations (about 30--100 nM) initiated oscillations of the membrane potential followed by complete depolarization in brain mitochondria. In contrast, liver mitochondria were less sensitive to Ca(2+); 20 microm Ca(2+) was required to depolarize liver mitochondria. Ca(2+) did not initiate oscillatory depolarizations in liver mitochondria, where each individual mitochondrion depolarized abruptly and irreversibly. Adenine nucleotides dramatically reduced the oscillatory depolarization in brain mitochondria and delayed the onset of the depolarization in liver mitochondria. In both type of mitochondria, the stabilizing effect of adenine nucleotides completely abolished by an inhibition of adenine nucleotide translocator function with carboxyatractyloside, but was not sensitive to bongkrekic acid. Inhibitors of mitochondrial permeability transition cyclosporine A and bongkrekic acid also delayed Ca(2+)-depolarization. We hypothesize that the oscillatory depolarization in brain mitochondria is associated with the transient conformational change of the adenine nucleotide translocator from a specific transporter to a non-specific pore, whereas the non-oscillatory depolarization in liver mitochondria is caused by the irreversible opening of the pore.


Subject(s)
Calcium/pharmacology , Mitochondria, Liver/drug effects , Mitochondria/drug effects , Adenine Nucleotides/pharmacology , Animals , Atractyloside/analogs & derivatives , Atractyloside/pharmacology , Brain/cytology , Cations, Divalent/pharmacology , Cyclosporine/pharmacology , Membrane Potentials/drug effects , Mitochondria/physiology , Mitochondrial ADP, ATP Translocases/antagonists & inhibitors , Rats
18.
Cell Calcium ; 37(3): 225-32, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15670869

ABSTRACT

Elevation of intracellular free zinc ([Zn2+]i) probably contributes to cell death in injury paradigms involving calcium deregulation and oxidative stress such as glutamate excitotoxicity. However, it is difficult to monitor both ions simultaneously in live cells. Here we present a new method using fluorescence microscopy and the ion sensitive indicators fura-2FF and FluoZin-3 to monitor both [Ca2+]i and [Zn2+]i in primary cortical neurons. We show that the new single wavelength dye FluoZin-3 responds robustly to small zinc loads, is insensitive to high Ca2+ or Mg2+, and is relatively unaffected by low pH or oxidants. The ratiometric indicator fura-2FF is sensitive to both Ca2+ and Zn2+. However, in conditions analogous to excitotoxic glutamate exposure where [Ca2+]i is high relative to [Zn2+]i, we found that fura-2FF responds mostly to [Ca2+]i but is relatively unaffected by low [Zn2+]i. Moreover, fura-2FF ratio changes caused by high [Ca2+]i or high [Zn2+]i could be distinguished because each ion produces a different spectral response. Finally, dual dye experiments showed that FluoZin-3 and fura-2FF respond robustly to [Zn2+]i and [Ca2+]j, respectively, in the same neurons during intense glutamate exposure. These studies provide a novel method for the simultaneous detection of both calcium and zinc in cells.


Subject(s)
Calcium/analysis , Intracellular Fluid/chemistry , Zinc/analysis , Animals , Cells, Cultured , Fluorescent Dyes , Fura-2/analogs & derivatives , Neurons/metabolism , Polycyclic Compounds , Prosencephalon/cytology , Prosencephalon/embryology , Rats , Spectrometry, Fluorescence
19.
J Neurosci ; 22(14): 5848-55, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12122047

ABSTRACT

Iron is an essential element for cells but may also be an important cytotoxin. However, very little is known about iron transport, redox status, or toxicity specifically inside cells. In this study, we exploited the sensitivity of fura-2 to quenching by ferrous iron (Fe(2+)) to detect intracellular free iron ([Fe(2+)](i)) in neurons, astrocytes, and oligodendrocytes in primary culture. All cell types exposed to Fe(2+) in the presence of the ionophore pyrithione rapidly accumulated Fe(2+) to a similar extent. The heavy-metal chelators bipyridyl and N,N,N',N'-tetrakis(2-pyridalmethyl)ethyl-enediamine rapidly reversed the increase in [Fe(2+)](i), whereas desferrioxamine had little effect. Interestingly, the Fe(2+)-mediated quenching of fura-2 fluorescence was reversed in a concentration-dependent manner by hydrogen peroxide. This was likely caused by the oxidation of Fe(2+) to Fe(3+) inside the cell. Acute exposure of cells to Fe(2+) was only toxic when the metal was applied together with pyrithione, showing that Fe(2+) is only toxic when elevated inside cells. Interestingly, only neurons and oligodendrocytes were injured by this elevation in [Fe(2+)](i), whereas astrocytes were unaffected, although [Fe(2+)](i) was elevated to the same degree in each cell type. These studies provide a novel approach for detecting [Fe(2+)](i) in a manner sensitive to the redox state of the metal. These studies also provide a model system for the study of the toxic consequences of elevated [Fe(2+)](i) in neural cells.


Subject(s)
Astrocytes/metabolism , Intracellular Fluid/metabolism , Iron/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cell Survival/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Fluorescent Dyes , Fura-2 , Hydrogen Peroxide/pharmacology , Ion Transport/drug effects , Ionophores/pharmacology , Iron/pharmacology , Iron Chelating Agents/pharmacology , Neurons/cytology , Neurons/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oxidants/pharmacology , Oxidation-Reduction , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Thiones
20.
J Neurosci ; 23(21): 7881-8, 2003 Aug 27.
Article in English | MEDLINE | ID: mdl-12944518

ABSTRACT

Mitochondria are essential to maintain neuronal viability. In addition to the generation of ATP and maintenance of calcium homeostasis, the effective delivery of mitochondria to the appropriate location within neurons is also likely to influence their function. In this study we examined mitochondrial movement and morphology in primary cultures of rat forebrain using a mitochondrially targeted enhanced yellow fluorescent protein (mt-eYFP). Mt-eYFP-labeled mitochondria display a characteristic elongated phenotype and also move extensively. Application of glutamate to cultures results in a rapid diminution of movement and also an alteration from elongated to rounded morphology. This effect required the entry of calcium and was mediated by activation of the NMDA subtype of glutamate receptor. Treatment of cultures with an uncoupler or blocking ATP synthesis with oligomycin also stopped movement but did not alter morphology. Interestingly, application of glutamate together with the uncoupler did not prevent the changes in movement or shape but facilitated recovery after washout of the stimuli. This suggests that the critical target for calcium in this paradigm is cytosolic. These studies demonstrate that in addition to altering the bioenergetic properties of mitochondria, neurotoxins can also alter mitochondrial movement and morphology. We speculate that neurotoxin-mediated impairment of mitochondrial delivery may contribute to the injurious effects of neurotoxins.


Subject(s)
Glutamic Acid/pharmacology , Mitochondria/ultrastructure , Neurons/ultrastructure , Prosencephalon/ultrastructure , Animals , Bacterial Proteins/analysis , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cells, Cultured , Dendrites/ultrastructure , Luminescent Proteins/analysis , Microscopy, Fluorescence , Mitochondria/drug effects , Movement/drug effects , Neurons/cytology , Neurons/drug effects , Prosencephalon/cytology , Rats , Uncoupling Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL