Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Neuroimaging ; 23(3): 397-400, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23317412

ABSTRACT

BACKGROUND AND PURPOSE: Distal hyperintense vessels (DHV) on MRI FLAIR sequences in acute brain ischemia are thought to represent leptomeningeal collateral flow. We hypothesized that DHV are more common in acute stroke patients with perfusion-diffusion weighted mismatch (PDM) than in those without. METHODS: We performed a retrospective study of consecutive anterior circulation stroke patients who underwent multimodal MRI within 8 hours of onset. We correlated DHV occurrence with the presence or absence of PDM, and analyzed DHV correlates when angiography was available. RESULTS: Twenty-one patients with PDM and 28 without were included. On univariate analysis, there was no significant difference regarding demographic variables between the two groups, with the exception of a higher frequency of atrial fibrillation (33% vs. 7%; P = .02) and intravenous tissue plasminogen activator use (57% vs 25%; P = .03) in the PDM patients. The PDM group more commonly had DHV (85% vs 25%; P < .001). On multivariate analysis, DHV presence (odds ratio, 6.01; 95% confidence-interval, 1.08-33.29; P = .04) and vessel occlusion site (odds ratio, 3.17; 95% confidence-interval, 1.21-8.31; P = .01) were the only variables independently associated with PDM. Conventional angiography was useful correlating DHV presence and collateral flow in a subset of patients. CONCLUSIONS: DHV may be a surrogate marker for PDM in patients with hyperacute ischemic stroke.


Subject(s)
Cerebral Arteries/pathology , Cerebrovascular Disorders/complications , Cerebrovascular Disorders/pathology , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Angiography/methods , Stroke/etiology , Stroke/pathology , Aged , Female , Humans , Male , Middle Aged , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity
2.
Neurology ; 79(13 Suppl 1): S86-94, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-23008419

ABSTRACT

Due to the dynamic and versatile characteristics of ischemic penumbra, selecting the right acute ischemic stroke (AIS) patients for revascularization therapy (RT) based on initial available imaging can be challenging. The main patient selection criterion for RT is the size of the mismatch between the potentially salvageable tissue (penumbra) and the irreversibly damaged tissue (core). The goal of revascularization RT is to "freeze" the core and prevent it from extending to the penumbral tissue. Penumbral imaging selection of AIS patients for RT, using magnetic resonance or CT-based studies, may provide more clinical benefit to the appropriate patients, although direct evidence is pending. Not all penumbra-core mismatches beyond 3 hours are equal and need treatment, and defining which mismatches to target for RT is the current goal of ongoing clinical trials. In addition to "penumbral"-based imaging, large vessel occlusion and clot length estimation based on CT angiography and noncontrasted ultrathin CT scan has been used to identify patients who are refractory to systemic thrombolysis and may be eligible for endovascular therapy. The application of various imaging modalities in selecting and triaging AIS patients for RT is discussed in this review. Larger prospective randomized trials are needed to better understand the role of various imaging modalities in selecting AIS patients for RT and to understand its influence on clinical outcome.


Subject(s)
Cerebral Revascularization/methods , Neuroimaging/methods , Patient Selection , Stroke/diagnosis , Stroke/surgery , Animals , Fibrinolytic Agents/administration & dosage , Humans , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/administration & dosage
4.
Free Radic Biol Med ; 40(6): 1034-44, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16540399

ABSTRACT

Nitric oxide (*NO) was shown to stimulate the proteasomal function and the ubiquitin-proteasome pathway and to ameliorate endothelial apoptotic signaling induced by oxidants. Understanding the regulatory mechanisms by which *NO stimulates proteasomes and affords cytoprotection in endothelial cells has therapeutic implications, as many vascular diseases are characterized by a deficiency in *NO. Here we report that *NO/cGMP/cAMP-induced immunoproteasome subunit expression is responsible for the increased proteasomal activities. Cells pretreated with protein kinase G and protein kinase A inhibitors markedly attenuated *NO-dependent proteasome activation. Results show that the *NO/cGMP/cAMP signaling mechanism enhanced the phosphorylation of the transcription factor cAMP-response element-binding protein, elevated the cAMP-response element-promoter activity and induced the expression of immunoproteasomal subunits (LMP2 and LMP7). *NO-dependent proteasomal activity was abrogated in cells transfected with antisense LMP2 and LMP7 oligonucleotides. Lower levels of LMP2 and LMP7 were detected in aorta of iNOS(-/-) mice compared to wild-type controls, suggesting that endogenous production of *NO is important in the basal regulation of immunoproteasome. The *NO/cGMP/cAMP signaling pathway mitigates transferrin-iron-mediated oxidative stress and apoptosis through induction of immunoproteasomes. These results provide new insights on the regulatory mechanisms by which the *NO-mediated immunoproteasome signaling pathway affords cytoprotection in endothelial cells.


Subject(s)
Antioxidants/physiology , Nitric Oxide/pharmacology , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/physiology , Animals , Apoptosis/drug effects , Cattle , Cyclic AMP/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/physiology , Cysteine Endopeptidases/metabolism , Endothelium, Vascular/cytology , Humans , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multienzyme Complexes/metabolism , Nitric Oxide Synthase Type II/deficiency , Proteasome Endopeptidase Complex/immunology , Signal Transduction , Up-Regulation
5.
Methods Enzymol ; 396: 526-34, 2005.
Article in English | MEDLINE | ID: mdl-16291259

ABSTRACT

In this chapter, oxidant-induced transferrin receptor-mediated iron-signaling and apoptosis are described in endothelial and neuronal cells exposed to oxidants. The role of nitric oxide in the regulation of iron homeostasis and oxidant-induced apoptosis is described. The interrelationship between oxidative stress, iron-signaling, and nitric oxide-dependent proteasomal function provides a rational mechanism that connects both oxidative and nitrative modifications.


Subject(s)
Aging/physiology , Homeostasis , Iron/physiology , Neurodegenerative Diseases/physiopathology , Nitric Oxide/physiology , Proteasome Endopeptidase Complex/physiology , Animals , Cattle , Cells, Cultured , Up-Regulation
6.
Free Radic Biol Med ; 39(8): 1059-74, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16198233

ABSTRACT

In this study, we investigated the molecular mechanism of toxicity of 1-methyl-4-phenylpyridinium (MPP+), an ultimate toxic metabolite of a mitochondrial neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, that causes parkinsonism in experimental animals and humans. Using wild-type and human neuronal nitric oxide synthase (nNOS) stably transfected neuroblastoma cells (SH-SY5Y), we showed that nNOS overexpression in SH-SY5Y cells greatly enhanced proteasome activity and mitigated MPP+-induced apoptosis. During MPP+-induced oxidative stress, intracellular BH4 levels decreased, resulting in nNOS "uncoupling" (i.e., switching from nitric oxide to superoxide generation). Increasing the intracellular BH4 levels by sepiapterin supplementation restored the nNOS activity, inhibited superoxide formation, increased proteasome activity, decreased protein ubiquitination, and attenuated apoptosis in MPP+-treated cells. Implications of BH4 depletion in dopaminergic cells and sepiapterin supplementation to augment the striatal nNOS activity in the pathogenesis mechanism and treatment of Parkinson disease are discussed.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Apoptosis/drug effects , Nitric Oxide Synthase Type I/metabolism , Proteasome Endopeptidase Complex/metabolism , Pterins/pharmacology , Aconitate Hydratase/antagonists & inhibitors , Biopterins/analogs & derivatives , Biopterins/analysis , Biopterins/metabolism , Dimerization , Electron Transport Complex I/antagonists & inhibitors , Enzyme Activation , Humans , Neuroblastoma , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Superoxides/metabolism , Transfection , Tumor Cells, Cultured
7.
J Biol Chem ; 280(41): 34644-53, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16085644

ABSTRACT

We have investigated here the mechanism of dephosphorylation and activation of death-associated protein kinase (DAPK) and the role of lysosome in neuroblastoma cells (SH-SY5Y) treated with mitochondrial toxins, such as MPP(+) and rotenone. Mitochondrial respiratory chain inhibitors and uncouplers decreased mitochondrial membrane potential leading to DAPK dephosphorylation and activation. The class III phosphoinositide 3-kinase inhibitors attenuated DAPK dephosphorylation induced by mitochondrial toxins. Complex I inhibition by mitochondrial toxins (e.g. MPP(+)) resulted in mitochondrial swelling and lysosome reduction. Inhibition of class III phosphoinositide 3-kinase attenuated MPP(+)-induced lysosome reduction and cell death. The role of DAPK as a sensor of mitochondrial membrane potential in mitochondrial diseases was addressed.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Lysosomes/physiology , Membrane Potentials , Mitochondrial Membranes/metabolism , 1-Methyl-4-phenylpyridinium/pharmacology , Aconitate Hydratase/metabolism , Antimycin A/pharmacology , Antioxidants/pharmacology , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Death , Cell Line, Tumor , Death-Associated Protein Kinases , Electron Transport , Humans , Lysosomes/metabolism , Microscopy, Electron , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rotenone/pharmacology , Subcellular Fractions , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Toxins, Biological/pharmacology
8.
J Biol Chem ; 279(36): 37575-87, 2004 Sep 03.
Article in English | MEDLINE | ID: mdl-15220329

ABSTRACT

The mitochondria-targeted drugs mitoquinone (Mito-Q) and mitovitamin E (MitoVit-E) are a new class of antioxidants containing the triphenylphosphonium cation moiety that facilitates drug accumulation in mitochondria. In this study, Mito-Q (ubiquinone attached to a triphenylphosphonium cation) and MitoVit-E (vitamin E attached to a triphenylphosphonium cation) were used. The aim of this study was to test the hypothesis that mitochondria-targeted antioxidants inhibit peroxide-induced oxidative stress and apoptosis in bovine aortic endothelial cells (BAEC) through enhanced scavenging of mitochondrial reactive oxygen species, thereby blocking reactive oxygen species-induced transferrin receptor (TfR)-mediated iron uptake into mitochondria. Glucose/glucose oxidase-induced oxidative stress in BAECs was monitored by oxidation of dichlorodihydrofluorescein that was catalyzed by both intracellular H(2)O(2) and transferrin iron transported into cells. Pretreatment of BAECs with Mito-Q (1 microM) and MitoVit-E (1 microM) but not untargeted antioxidants (e.g. vitamin E) significantly abrogated H(2)O(2)- and lipid peroxide-induced 2',7'-dichlorofluorescein fluorescence and protein oxidation. Mitochondria-targeted antioxidants inhibit cytochrome c release, caspase-3 activation, and DNA fragmentation. Mito-Q and MitoVit-E inhibited H(2)O(2)- and lipid peroxide-induced inactivation of complex I and aconitase, TfR overexpression, and mitochondrial uptake of (55)Fe, while restoring the mitochondrial membrane potential and proteasomal activity. We conclude that Mito-Q or MitoVit-E supplementation of endothelial cells mitigates peroxide-mediated oxidant stress and maintains proteasomal function, resulting in the overall inhibition of TfR-dependent iron uptake and apoptosis.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Endothelium, Vascular/drug effects , Iron/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects , Peroxides/antagonists & inhibitors , Aconitate Hydratase/antagonists & inhibitors , Animals , Cattle , Cells, Cultured , Electron Transport Complex I/antagonists & inhibitors , Endothelium, Vascular/metabolism , Glucose Oxidase/pharmacology , In Situ Nick-End Labeling , Mitochondria/metabolism , Receptors, Transferrin/physiology
9.
J Biol Chem ; 279(18): 19099-112, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-14752097

ABSTRACT

In this study, we investigated the molecular mechanisms of toxicity of 1-methyl-4-phenylpyridinium (MPP(+)), an ultimate toxic metabolite of a mitochondrial neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, that causes Parkinson-like symptoms in experimental animals and humans. We used rat cerebellar granule neurons as a model cell system for investigating MPP(+) toxicity. Results show that MPP(+) treatment resulted in the generation of reactive oxygen species from inhibition of complex I of the mitochondrial respiratory chain, and inactivation of aconitase. This, in turn, stimulated transferrin receptor (TfR)-dependent iron signaling via activation of the iron-regulatory protein/iron-responsive element interaction. MPP(+) caused a time-dependent depletion of tetrahydrobiopterin (BH(4)) that was mediated by H(2)O(2) and transferrin iron. Depletion of BH(4) decreased the active, dimeric form of neuronal nitric-oxide synthase (nNOS). MPP(+)-mediated "uncoupling" of nNOS decreased *NO and increased superoxide formation. Pretreatment of cells with sepiapterin to promote BH(4) biosynthesis or cell-permeable iron chelator and TfR antibody to prevent iron-catalyzed BH(4) decomposition inhibited MPP(+) cytotoxicity. Preincubation of cerebellar granule neurons with nNOS inhibitor exacerbated MPP(+)-induced iron uptake, BH(4) depletion, proteasomal inactivation, and apoptosis. We conclude that MPP(+)-dependent aconitase inactivation, Tf-iron uptake, and oxidant generation result in the depletion of intracellular BH(4), leading to the uncoupling of nNOS activity. This further exacerbates reactive oxygen species-mediated oxidative damage and apoptosis. Implications of these results in unraveling the molecular mechanisms of neurodegenerative diseases (Parkinson's and Alzheimer's disease) are discussed.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Apoptosis/drug effects , Biopterins/analogs & derivatives , Interneurons/cytology , Nitric Oxide Synthase/physiology , Receptors, Transferrin/physiology , 1-Methyl-4-phenylpyridinium/pharmacology , Aconitate Hydratase/antagonists & inhibitors , Animals , Antioxidants/metabolism , Biopterins/metabolism , Cerebellum/cytology , Electron Transport Complex I/antagonists & inhibitors , Humans , Iron/physiology , Mitochondrial Proteins , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Parkinson Disease , Rats , Reactive Oxygen Species/metabolism , Receptors, Transferrin/biosynthesis , Signal Transduction , Superoxides/metabolism
10.
J Neurochem ; 85(2): 358-67, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12675912

ABSTRACT

1-Methyl-4-phenylpyridinium (MPP+), the toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, induces apoptosis in cerebellar granule neurons (CGNs). We have tested the hypothesis that organic cation transporter (OCT) 3 mediates the accumulation and, hence, the toxicity of MPP+ in CGNs. CGNs in primary culture express OCT3 but do not express mRNA for OCT1, OCT2 or the dopamine transporter. Cerebellar astrocytes are negative for OCT3 protein by immunocytochemistry. [3H]MPP+ accumulation by CGNs exhibits first-order kinetics, and a Kt value of 5.3 +/- 1.2 micro m and a Tmax of 0.32 +/- 0.02 pmol per min per 106 cells. [3H]MPP+ accumulation is inhibited by corticosterone, beta-estradiol and decynium 22 with Ki values of 0.25 micro m, 0.17 micro m and 4.0 nm respectively. [3H]MPP+ accumulation is also inhibited by desipramine, dopamine, serotonin and norepinephrine, but is not affected by carnitine (10 mm), mazindol (9 micro m) or GBR 12909 (1 micro m). MPP+-induced caspase-3-like activation and cell death are prevented by pretreatment with 5 micro mbeta-estradiol. In contrast, the neurotoxic effects of rotenone are unaffected by beta-estradiol. Interestingly, GBR 12909 protects CGNs from both MPP+ and rotenone toxicity. In summary, CGNs accumulate MPP+ in manner that is consistent with uptake via OCT3 and the presence of this protein in CGNs explains their sensitivity to MPP+ toxicity.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacokinetics , Neurons/metabolism , Organic Cation Transport Proteins/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , Adrenergic Uptake Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Biological Transport/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , Corticosterone/pharmacology , Desipramine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drug Antagonism , Estradiol/pharmacology , Female , Male , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/genetics , Piperazines/pharmacology , Quinolines/pharmacology , RNA, Messenger/biosynthesis , Rats , Rotenone/toxicity , Temperature
11.
Biochem J ; 371(Pt 1): 151-64, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12523938

ABSTRACT

1-Methyl-4-phenylpyridinium (MPP(+)) is a neurotoxin used in cellular models of Parkinson's Disease. Although intracellular iron plays a crucial role in MPP(+)-induced apoptosis, the molecular signalling mechanisms linking iron, reactive oxygen species (ROS) and apoptosis are still unknown. We investigated these aspects using cerebellar granule neurons (CGNs) and human SH-SY5Y neuroblastoma cells. MPP(+) enhanced caspase 3 activity after 24 h with significant increases as early as 12 h after treatment of cells. Pre-treatment of CGNs and neuroblastoma cells with the metalloporphyrin antioxidant enzyme mimic, Fe(III)tetrakis(4-benzoic acid)porphyrin (FeTBAP), completely prevented the MPP(+)-induced caspase 3 activity as did overexpression of glutathione peroxidase (GPx1) and pre-treatment with a lipophilic, cell-permeable iron chelator [N, N '-bis-(2-hydroxybenzyl)ethylenediamine-N, N '-diacetic acid, HBED]. MPP(+) treatment increased the number of TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick-end-labelling)-positive cells which was completely blocked by pre-treatment with FeTBAP. MPP(+) treatment significantly decreased the aconitase and mitochondrial complex I activities; pre-treatment with FeTBAP, HBED and GPx1 overexpression reversed this effect. MPP(+) treatment increased the intracellular oxidative stress by 2-3-fold, as determined by oxidation of dichlorodihydrofluorescein and dihydroethidium (hydroethidine). These effects were reversed by pre-treatment of cells with FeTBAP and HBED and by GPx1 overexpression. MPP(+)-treatment enhanced the cell-surface transferrin receptor (TfR) expression, suggesting a role for TfR-induced iron uptake in MPP(+) toxicity. Treatment of cells with anti-TfR antibody (IgA class) inhibited MPP(+)-induced caspase activation. Inhibition of nitric oxide synthase activity did not affect caspase 3 activity, apoptotic cell death or ROS generation by MPP(+). Overall, these results suggest that MPP(+)-induced cell death in CGNs and neuroblastoma cells proceeds via apoptosis and involves mitochondrial release of ROS and TfR-dependent iron.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Apoptosis/drug effects , Edetic Acid/analogs & derivatives , Hydrogen Peroxide/metabolism , Iron/metabolism , Mitochondria/metabolism , Receptors, Transferrin/metabolism , Aconitate Hydratase/drug effects , Aconitate Hydratase/metabolism , Animals , Antioxidants/pharmacology , Caspase 3 , Caspases/drug effects , Caspases/metabolism , Cells, Cultured , Deferoxamine/pharmacology , Edetic Acid/pharmacology , Female , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Humans , Iron Chelating Agents/pharmacology , Male , Metalloporphyrins/pharmacology , Mitochondria/drug effects , NADH, NADPH Oxidoreductases/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidants/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism , Receptors, Transferrin/drug effects , Receptors, Transferrin/immunology , Staining and Labeling/methods , Transferrin/metabolism , Glutathione Peroxidase GPX1
SELECTION OF CITATIONS
SEARCH DETAIL
...