Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Redox Biol ; 67: 102895, 2023 11.
Article in English | MEDLINE | ID: mdl-37769522

ABSTRACT

Glutathione (GSH) depletion, and impaired redox homeostasis have been observed in experimental animal models and patients with epilepsy. Pleiotropic strategies that elevate GSH levels via transcriptional regulation have been shown to significantly decrease oxidative stress and seizure frequency, increase seizure threshold, and rescue certain cognitive deficits. Whether elevation of GSH per se alters neuronal hyperexcitability remains unanswered. We previously showed that thiols such as dimercaprol (DMP) elevate GSH via post-translational activation of glutamate cysteine ligase (GCL), the rate limiting GSH biosynthetic enzyme. Here, we asked if elevation of cellular GSH by DMP altered neuronal hyperexcitability in-vitro and in-vivo. Treatment of primary neuronal-glial cerebrocortical cultures with DMP elevated GSH and inhibited a voltage-gated potassium channel blocker (4-aminopyridine, 4AP) induced neuronal hyperexcitability. DMP increased GSH in wildtype (WT) zebrafish larvae and significantly attenuated convulsant pentylenetetrazol (PTZ)-induced acute 'seizure-like' swim behavior. DMP treatment increased GSH and inhibited convulsive, spontaneous 'seizure-like' swim behavior in the Dravet Syndrome (DS) zebrafish larvae (scn1Lab). Furthermore, DMP treatment significantly decreased spontaneous electrographic seizures and associated seizure parameters in scn1Lab zebrafish larvae. We investigated the role of the redox-sensitive mammalian target of rapamycin (mTOR) pathway due to the presence of several cysteine-rich proteins and their involvement in regulating neuronal excitability. Treatment of primary neuronal-glial cerebrocortical cultures with 4AP or l-buthionine-(S,R)-sulfoximine (BSO), an irreversible inhibitor of GSH biosynthesis, significantly increased mTOR complex I (mTORC1) activity which was rescued by pre-treatment with DMP. Furthermore, BSO-mediated GSH depletion oxidatively modified the tuberous sclerosis protein complex (TSC) consisting of hamartin (TSC1), tuberin (TSC2), and TBC1 domain family member 7 (TBC1D7) which are critical negative regulators of mTORC1. In summary, our results suggest that DMP-mediated GSH elevation by a novel post-translational mechanism can inhibit neuronal hyperexcitability both in-vitro and in-vivo and a plausible link is the redox sensitive mTORC1 pathway.


Subject(s)
Glutathione , Zebrafish , Animals , Humans , Zebrafish/metabolism , Glutathione/metabolism , Glutamate-Cysteine Ligase/metabolism , TOR Serine-Threonine Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1 , Seizures/chemically induced , Seizures/drug therapy , Buthionine Sulfoximine/pharmacology , Mammals/metabolism
2.
J Neurosci ; 43(10): 1845-1857, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36759193

ABSTRACT

Mitochondrial dysfunction is an early event in the pathogenesis of neurologic disorders and aging. Sirtuin 3 (SIRT3) regulates mitochondrial function in response to the cellular environment through the reversible deacetylation of proteins involved in metabolism and reactive oxygen species detoxification. As the primary mitochondrial deacetylase, germline, or peripheral tissue-specific deletion of SIRT3 produces mitochondrial hyperacetylation and the accelerated development of age-related diseases. Given the unique metabolic demands of neurons, the role of SIRT3 in the brain is only beginning to emerge. Using mass spectrometry-based acetylomics, high-resolution respirometry, video-EEG, and cognition testing, we report targeted deletion of SIRT3 from select neurons in the cortex and hippocampus produces altered neuronal excitability and metabolic dysfunction in female mice. Targeted deletion of SIRT3 from neuronal helix-loop-helix 1 (NEX)-expressing neurons resulted in mitochondrial hyperacetylation, female-specific superoxide dismutase-2 (SOD2) modification, increased steady-state superoxide levels, metabolic reprogramming, altered neuronal excitability, and working spatial memory deficits. Inducible neuronal deletion of SIRT3 likewise produced female-specific deficits in spatial working memory. Together, the data demonstrate that deletion of SIRT3 from forebrain neurons selectively predisposes female mice to deficits in mitochondrial and cognitive function.SIGNIFICANCE STATEMENT Mitochondrial SIRT3 is an enzyme shown to regulate energy metabolism and antioxidant function, by direct deacetylation of proteins. In this study, we show that neuronal SIRT3 deficiency renders female mice selectively vulnerable to impairment in redox and metabolic function, spatial memory, and neuronal excitability. The observed sex-specific effects on cognition and neuronal excitability in female SIRT3-deficient mice suggest that mitochondrial dysfunction may be one factor underlying comorbid neuronal diseases, such as Alzheimer's disease and epilepsy. Furthermore, the data suggest that SIRT3 dysfunction may predispose females to age-related metabolic and cognitive impairment.


Subject(s)
Sirtuin 3 , Male , Mice , Female , Animals , Sirtuin 3/genetics , Neurons/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Aging/metabolism , Acetylation
3.
Neurobiol Dis ; 158: 105470, 2021 10.
Article in English | MEDLINE | ID: mdl-34371143

ABSTRACT

Mitochondrial superoxide (O2-) production is implicated in aging, neurodegenerative disease, and most recently epilepsy. Yet the specific contribution of neuronal O2- to these phenomena is unclear. Here, we selectively deleted superoxide dismutase-2 (SOD2) in neuronal basic helix-loop-helix transcription factor (NEX)-expressing cells restricting deletion to a subset of excitatory principle neurons primarily in the forebrain (cortex and hippocampus). This resulted in nSOD2 KO mice that lived into adulthood (2-3 months) with epilepsy, selective loss of neurons, metabolic rewiring and a marked mitohormetic gene response. Surprisingly, expression of an astrocytic gene, glial fibrillary acidic protein (GFAP) was significantly increased relative to WT. Further studies in rat primary neuron-glial cultures showed that increased mitochondrial O2-, specifically in neurons, was sufficient to upregulate GFAP. These results suggest that neuron-specific mitochondrial O2- is sufficient to drive a complex and catastrophic epileptic phenotype and highlights the ability of SOD2 to act in a cell-nonautonomous manner to influence an astrocytic response.


Subject(s)
Astrocytes/pathology , Epilepsy/pathology , Glucose Metabolism Disorders/pathology , Mitochondria , Neurons , Oxidative Stress , Animals , Behavior, Animal , Electroencephalography , Epilepsy/psychology , Glial Fibrillary Acidic Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Primary Cell Culture , Rats , Superoxide Dismutase/genetics , Superoxides/metabolism
4.
J Pharmacol Exp Ther ; 377(1): 1-10, 2021 04.
Article in English | MEDLINE | ID: mdl-33500265

ABSTRACT

Oxidative stress plays a crucial role in the pathogenesis of Parkinson disease (PD), and one strategy for neuroprotective therapy for PD is to scavenge reactive species using a catalytic antioxidant. Previous studies in our laboratory revealed that pretreatment of lipophilic metalloporphyrins showed protective effects in a mouse PD model. In this study, we optimized the formulations of these metalloporphyrins to deliver them orally and tested their efficacy on disease outcomes in a second species after initiation of an insult (i.e., disease modification). In this study, a pharmaceutical formulation of two metalloporphyrin catalytic antioxidants, AEOL11207 and AEOL11114, was tested for oral drug delivery. Both compounds showed gastrointestinal absorption, achieved high plasma concentrations, and readily penetrated the blood-brain barrier after intravenous or oral delivery. AEOL11207 and AEOL11114 bioavailabilities were calculated to be 24% and 25%, respectively, at a dose of 10 mg/kg via the oral route. In addition, both compounds significantly attenuated 6-hydroxydopamine (6-OHDA)-induced neurotoxic damage, including dopamine depletion, cytokine production, and microglial activation in the striata; dopaminergic neuronal loss in the substantia nigra; oxidative/nitrative stress indices (glutathione disulfide and 3-nitrotyrosine) in the ventral midbrain; and rotation behavioral abnormality in rats. These results indicate that AEOL11207 and AEOL11114 are orally active metalloporphyrins and protect against 6-OHDA neurotoxicity 1-3 days postlesioning, suggesting disease-modifying properties and translational potential for PD. SIGNIFICANCE STATEMENT: Two catalytic antioxidants showed gastrointestinal absorption, achieved high plasma concentrations, and readily penetrated the blood-brain barrier. Both compounds significantly attenuated dopamine depletion, cytokine production, microglial activation, dopaminergic neuronal loss, oxidative/nitrative stress indices, and behavioral abnormality in a Parkinson disease rat model. The results suggest that both metalloporphyrins possess disease-modifying properties that may be useful in treating Parkinson disease.


Subject(s)
Antioxidants/pharmacokinetics , Metalloporphyrins/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Parkinsonian Disorders/drug therapy , Administration, Oral , Animals , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Blood-Brain Barrier/metabolism , Male , Metalloporphyrins/administration & dosage , Metalloporphyrins/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Tissue Distribution
5.
PLoS One ; 15(10): e0240451, 2020.
Article in English | MEDLINE | ID: mdl-33052948

ABSTRACT

Cell differentiation and cell fate determination in sensory systems are essential for stimulus discrimination and coding of environmental stimuli. Color vision is based on the differential color sensitivity of retinal photoreceptors, however the developmental programs that control photoreceptor cell differentiation and specify color sensitivity are poorly understood. In Drosophila melanogaster, there is evidence that the color sensitivity of different photoreceptors in the compound eye is regulated by inductive signals between cells, but the exact nature of these signals and how they are propagated remains unknown. We conducted a genetic screen to identify additional regulators of this process and identified a novel mutation in the hibris gene, which encodes an irre cell recognition module protein (IRM). These immunoglobulin super family cell adhesion molecules include human KIRREL and nephrin (NPHS1). hibris is expressed dynamically in the developing Drosophila melanogaster eye and loss-of-function mutations give rise to a diverse range of mutant phenotypes including disruption of the specification of R8 photoreceptor cell diversity. We demonstrate that hibris is required within the retina, and that hibris over-expression is sufficient to disrupt normal photoreceptor cell patterning. These findings suggest an additional layer of complexity in the signaling process that produces paired expression of opsin genes in adjacent R7 and R8 photoreceptor cells.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Retina/growth & development , Animals , Cell Differentiation , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Imaginal Discs/metabolism , Mutation , Organ Specificity , Photoreceptor Cells, Invertebrate/cytology , Retina/metabolism
6.
Toxicol Appl Pharmacol ; 326: 34-42, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28400118

ABSTRACT

Reactive oxygen species are a well-defined therapeutic target for Parkinson's disease (PD) and pharmacological agents that catalytically scavenge reactive species are promising neuroprotective strategies for treatment. Metalloporphyrins are synthetic catalytic antioxidants that mimic the body's own antioxidant enzymes i.e. superoxide dismutases and catalase. The goal of this study was to determine if newly designed metalloporphyrins have enhanced pharmacodynamics including oral bioavailability, longer plasma elimination half-lives, penetrate the blood brain barrier, and show promise for PD treatment. Three metalloporphyrins (AEOL 11216, AEOL 11203 and AEOL 11114) were identified in this study as potential candidates for further pre-clinical development. Each of these compounds demonstrated blood brain barrier permeability by the i.p. route and two of three compounds (AEOL 11203 and AEOL 11114) were orally bioavailable. All of these compounds protected against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity, including dopamine depletion in the striatum, dopaminergic neuronal loss in the substantial nigra, and increased oxidative/nitrative stress indices (glutathione disulfide and 3-nitrotyrosine) in the ventral midbrain of the mice without inhibiting MPTP metabolism. Daily therapeutic dosing of these metalloporphyrins were well tolerated without accumulation of brain manganese levels or behavioral alterations assessed by open field and rotarod tests. The study identified two orally active metalloporphyrins and one injectable metalloporphyrin as clinical candidates for further development in PD.


Subject(s)
Antioxidants/pharmacology , Antiparkinson Agents/pharmacology , Brain/drug effects , MPTP Poisoning/prevention & control , Metalloporphyrins/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Administration, Oral , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacokinetics , Behavior, Animal/drug effects , Biological Availability , Biomarkers/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/physiopathology , Capillary Permeability , Disease Models, Animal , Dopamine/metabolism , Drug Design , Drug Evaluation, Preclinical , Half-Life , Injections, Intraperitoneal , MPTP Poisoning/etiology , MPTP Poisoning/metabolism , MPTP Poisoning/physiopathology , Male , Metalloporphyrins/administration & dosage , Metalloporphyrins/pharmacokinetics , Mice, Inbred C57BL , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Rotarod Performance Test
7.
eNeuro ; 3(2)2016.
Article in English | MEDLINE | ID: mdl-27066534

ABSTRACT

Altered metabolism is an important feature of many epileptic syndromes but has not been reported in Dravet syndrome (DS), a catastrophic childhood epilepsy associated with mutations in a voltage-activated sodium channel, Nav1.1 (SCN1A). To address this, we developed novel methodology to assess real-time changes in bioenergetics in zebrafish larvae between 4 and 6 d postfertilization (dpf). Baseline and 4-aminopyridine (4-AP) stimulated glycolytic flux and mitochondrial respiration were simultaneously assessed using a Seahorse Biosciences extracellular flux analyzer. Scn1Lab mutant zebrafish showed a decrease in baseline glycolytic rate and oxygen consumption rate (OCR) compared to controls. A ketogenic diet formulation rescued mutant zebrafish metabolism to control levels. Increasing neuronal excitability with 4-AP resulted in an immediate increase in glycolytic rates in wild-type zebrafish, whereas mitochondrial OCR increased slightly and quickly recovered to baseline values. In contrast, scn1Lab mutant zebrafish showed a significantly slower and exaggerated increase of both glycolytic rates and OCR after 4-AP. The underlying mechanism of decreased baseline OCR in scn1Lab mutants was not because of altered mitochondrial DNA content or dysfunction of enzymes in the electron transport chain or tricarboxylic acid cycle. Examination of glucose metabolism using a PCR array identified five glycolytic genes that were downregulated in scn1Lab mutant zebrafish. Our findings in scn1Lab mutant zebrafish suggest that glucose and mitochondrial hypometabolism contribute to the pathophysiology of DS.


Subject(s)
Epilepsies, Myoclonic/physiopathology , Glycolysis/genetics , Mitochondria/metabolism , Oxygen Consumption/genetics , 4-Aminopyridine/pharmacology , Animals , Animals, Genetically Modified , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Diet, Ketogenic/methods , Disease Models, Animal , Epilepsies, Myoclonic/diet therapy , Epilepsies, Myoclonic/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glycolysis/drug effects , Histocompatibility Antigens/metabolism , Larva , Mitochondria/drug effects , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Oxygen Consumption/drug effects , Potassium Channel Blockers/pharmacology , Statistics, Nonparametric , Zebrafish
8.
J Biol Chem ; 290(36): 21951-61, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26195627

ABSTRACT

The molecular mechanisms that regulate invertebrate visual pigment absorption are poorly understood. Studies of amphioxus Go-opsin have demonstrated that Glu-181 functions as the counterion in this pigment. This finding has led to the proposal that Glu-181 may function as the counterion in other invertebrate visual pigments as well. Here we describe a series of mutagenesis experiments to test this hypothesis and to also test whether other conserved acidic amino acids in Drosophila Rhodopsin 1 (Rh1) may serve as the counterion of this visual pigment. Of the 5 Glu and Asp residues replaced by Gln or Asn in our experiments, none of the mutant pigments shift the absorption of Rh1 by more than 6 nm. In combination with prior studies, these results suggest that the counterion in Drosophila Rh1 may not be located at Glu-181 as in amphioxus, or at Glu-113 as in bovine rhodopsin. Conversely, the extremely low steady state levels of the E194Q mutant pigment (bovine opsin site Glu-181), and the rhabdomere degeneration observed in flies expressing this mutant demonstrate that a negatively charged residue at this position is essential for normal rhodopsin function in vivo. This work also raises the possibility that another residue or physiologic anion may compensate for the missing counterion in the E194Q mutant.


Subject(s)
Aspartic Acid/genetics , Drosophila Proteins/genetics , Glutamic Acid/genetics , Mutation , Rhodopsin/genetics , Animals , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Blotting, Western , Conserved Sequence/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Microspectrophotometry , Opsins/classification , Opsins/genetics , Opsins/metabolism , Phylogeny , Protein Structure, Secondary , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism
9.
Neurobiol Dis ; 75: 151-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25600213

ABSTRACT

Metabolic alterations have been implicated in the etiology of temporal lobe epilepsy (TLE), but whether or not they have a functional impact on cellular energy producing pathways (glycolysis and/or oxidative phosphorylation) is unknown. The goal of this study was to determine if alterations in cellular bioenergetics occur using real-time analysis of mitochondrial oxygen consumption and glycolytic rates in an animal model of TLE. We hypothesized that increased steady-state levels of reactive oxygen species (ROS) initiated by epileptogenic injury result in impaired mitochondrial respiration. We established methodology for assessment of bioenergetic parameters in isolated synaptosomes from the hippocampus of Sprague-Dawley rats at various times in the kainate (KA) model of TLE. Deficits in indices of mitochondrial respiration were observed at time points corresponding with the acute and chronic phases of epileptogenesis. We asked if mitochondrial bioenergetic dysfunction occurred as a result of increased mitochondrial ROS and if it could be attenuated in the KA model by pharmacologically scavenging ROS. Increased steady-state ROS in mice with forebrain-specific conditional deletion of manganese superoxide dismutase (Sod2(fl/fl)NEX(Cre/Cre)) in mice resulted in profound deficits in mitochondrial oxygen consumption. Pharmacological scavenging of ROS with a catalytic antioxidant restored mitochondrial respiration deficits in the KA model of TLE. Together, these results demonstrate that mitochondrial respiration deficits occur in experimental TLE and ROS mechanistically contribute to these deficits. Furthermore, this study provides novel methodology for assessing cellular metabolism during the entire time course of disease development.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Glycolysis/physiology , Hippocampus/metabolism , Mitochondria/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Acute Disease , Animals , Antioxidants/pharmacology , Cell Respiration/drug effects , Cell Respiration/physiology , Chronic Disease , Disease Models, Animal , Female , Hippocampus/drug effects , Kainic Acid , Male , Mice, Knockout , Mitochondria/drug effects , Rats, Sprague-Dawley , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics
10.
J Neurochem ; 118(1): 79-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21517855

ABSTRACT

Mitochondrial oxidative stress is a contributing factor in the etiology of numerous neuronal disorders. However, the precise mechanism(s) by which mitochondrial reactive oxygen species modify cellular targets to induce neurotoxicity remains unknown. In this study, we determined the role of mitochondrial aconitase (m-aconitase) in neurotoxicity by decreasing its expression. Incubation of the rat dopaminergic cell line, N27, with paraquat (PQ(2+) ) resulted in aconitase inactivation, increased hydrogen peroxide (H(2) O(2) ) and increased ferrous iron (Fe(2+) ) at times preceding cell death. To confirm the role of m-aconitase in dopaminergic cell death, we knocked down m-aconitase expression via RNA interference. Incubation of m-aconitase knockdown N27 cells with PQ(2+) resulted in decreased H(2) O(2) production, Fe(2+) accumulation, and cell death compared with cells expressing basal levels of m-aconitase. To determine the metabolic role of m-aconitase in mediating neuroprotection, we conducted a complete bioenergetic profile. m-Aconitase knockdown N27 cells showed a global decrease in metabolism (glycolysis and oxygen consumption rates) which blocked PQ(2+) -induced H(+) leak and respiratory capacity deficiency. These findings suggest that dopaminergic cells are protected from death by decreasing release of H(2) O(2) and Fe(2+) in addition to decreased cellular metabolism.


Subject(s)
Aconitate Hydratase/metabolism , Dopamine/metabolism , Hydrogen Peroxide/metabolism , Iron/metabolism , Mitochondria/metabolism , Aconitate Hydratase/deficiency , Aconitate Hydratase/genetics , Analysis of Variance , Animals , Antimycin A/pharmacology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Death/drug effects , Cell Line, Transformed , Fumarate Hydratase/metabolism , Gene Expression Regulation/drug effects , Herbicides/toxicity , Ionophores/pharmacology , L-Lactate Dehydrogenase/metabolism , Mitochondria/drug effects , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Paraquat/toxicity , Protons , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Time Factors , Transfection/methods
11.
J Neurosci ; 27(16): 4326-33, 2007 Apr 18.
Article in English | MEDLINE | ID: mdl-17442816

ABSTRACT

Parkinson's disease (PD) is an age-related neurodegenerative disease in which the role of reactive oxygen species (ROS) is strongly implicated. The presence of oxidative stress has been detected in human and experimental PD using both direct and indirect indices. Scavenging ROS is, therefore, an important therapeutic avenue for the treatment of PD. Manganic porphyrins are catalytic antioxidants that scavenge a wide range of ROS. In this study, we tested the therapeutic effects of a compound [5,15-bis(methoxycarbonyl)-10,20-bis-trifluoromethyl-porphyrinato manganese (III) chloride (AEOL11207)] belonging to a new generation of lipophilic manganic porphyrins for neuroprotection and oral bioavailability in the mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of parkinsonism. Groups of adult C57BL/6 mice were administered MPTP with varying subcutaneous or oral dosing regimens of AEOL11207. Neurotoxicity was assessed by measurement of striatal dopamine levels and quantification of tyrosine hydroxylase-positive neurons in the substantial nigra pars compacta one week after the first dose of MPTP. Glutathione depletion, lipid peroxidation, and 3-nitrotyrosine (3-NT) formation were measured as indicators of oxidative stress in the ventral midbrain in vivo. AEOL11207 administered either by subcutaneous or oral routes protected against MPTP-induced dopamine depletion in the striatum as well as dopaminergic neuronal loss, glutathione depletion, lipid peroxidation, and 3-NT formation in the ventral midbrain. Neuroprotection correlated with brain metalloporphyrin concentrations. This is the first demonstration of neuroprotection by an orally active catalytic antioxidant in the MPTP mouse model and suggests its potential clinical utility for the treatment of chronic neurodegenerative diseases such as PD.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Metalloporphyrins/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinson Disease/prevention & control , Administration, Oral , Animals , Blood-Brain Barrier/metabolism , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Glutathione/metabolism , Glutathione Disulfide/metabolism , Half-Life , Injections, Subcutaneous , MPTP Poisoning/complications , Male , Metalloporphyrins/pharmacokinetics , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Parkinson Disease/etiology
12.
Popul Trends ; (122): 18-26, 2005.
Article in English | MEDLINE | ID: mdl-16411621

ABSTRACT

In each decade, population estimates are rebased using data from the most recent census. However, this would lead to a step change in the population estimates series. To avoid this discontinuity the backseries for 1992 to 2000, was revised to bring it into line with the 2001 Census. This article discusses the methodology used to produce the final revised backseries for 1992 to 2000 published by ONS in October 2004. The final estimates were produced after a long period of research into the best methodology to use. Traditionally, the backseries have been revised using an interim simple period method, followed by a final simple cohort method. The approach taken following the 2001 Census was much more comprehensive. This article outlines this approach, summarises the range of methods available and describes in detail the final method selected.


Subject(s)
Censuses/history , Data Interpretation, Statistical , Population Growth , Cohort Studies , England , History, 20th Century , Humans , Wales
SELECTION OF CITATIONS
SEARCH DETAIL
...