Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
J Vet Intern Med ; 32(1): 423-427, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29130572

ABSTRACT

A 6-year-old neutered female Jack Russell terrier was investigated for sudden onset prechiasmatic bilateral blindness, left circling, reduced proprioception in the right pelvic limb and right facial allodynia. Electroretinography was normal. Magnetic resonance imaging (MRI) examination revealed that the right optic nerve and the optic chiasm were hyperintense on diffusion weighted imaging and hypointense on apparent diffusion coefficient map consistent with ischemic optic neuropathy. A concurrent lacunar infarct was detected in the left rostral colliculus. Primary systemic hypertension was diagnosed based on blood pressure measurement and no detectable abnormalities on hematology, comprehensive serum biochemistry, urinalysis including protein/creatinine and cortisol/creatinine ratios and thoracic/abdominal imaging. Prednisolone for 10 days and amlodipine long-term were administered. Vision was not recovered after 7 months. Repeat MRI supported the diagnosis of ischemic lesions and revealed a recent striatocapsular infarct. Ischemic optic neuropathy is a well-recognized cause of blindness in humans and should be included as a differential diagnosis for acute prechiasmatic blindness in dogs.


Subject(s)
Blindness/veterinary , Dog Diseases/diagnosis , Hypertension/veterinary , Optic Neuropathy, Ischemic/veterinary , Amlodipine/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Antihypertensive Agents/therapeutic use , Blindness/diagnosis , Dogs , Female , Hypertension/drug therapy , Magnetic Resonance Imaging/veterinary , Optic Neuropathy, Ischemic/diagnostic imaging , Prednisolone/therapeutic use
2.
J Small Anim Pract ; 54(4): 205-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23278915

ABSTRACT

Nematode worms were retrieved from the left eyes of two dogs presented for unilateral ocular discharge in Belgium. Morphological and molecular identification were performed and the parasites were identified as Thelazia callipaeda. The history suggested that the infection had been acquired in south-western France and southern Italy where the disease has been observed regularly for the last 6 and 12 years, respectively. In these two regions, the disease is considered endemic and spreading. To the authors' knowledge, this is the first case report of canine thelaziosis in Belgium.


Subject(s)
Dog Diseases/diagnosis , Eye Infections, Parasitic/veterinary , Spirurida Infections/veterinary , Animals , Belgium , Dogs , Eye Infections, Parasitic/diagnosis , Female , France/epidemiology , Italy/epidemiology , Male , Spirurida Infections/diagnosis , Thelazioidea/isolation & purification , Travel
3.
Vet Rec ; 171(10): 247, 2012 Sep 08.
Article in English | MEDLINE | ID: mdl-22791527

ABSTRACT

The objective of this prospective study was to investigate the success rate, practicality and complications of a new perilimbal pocket technique for the replacement of prolapsed nictitans gland in 30 dogs (44 eyes). A first incision was made in the bulbar conjunctiva, 2-3 mm from and parallel to the infero-nasal limbus, a second incision on the bulbar aspect of the nictitating membrane (NM), 2-3 mm parallel to the free edge. The gland was returned to its normal position by suturing the subconjunctival tissues of the NM to the episcleral tissues, using four to six interrupted horizontal mattress sutures. The English bulldog, Neapolitan mastiff, great dane and American cocker spaniel were commonly presented. Nictitans gland prolapse occurred prior to one year of age in 83.3 per cent of dogs, and unilaterally in 15 patients. The procedure was easy to perform, and had a 90.9 per cent success rate, with minimal complications. The median duration of follow-up, conducted by ophthalmic examination or telephone contact with the owners, was 21.5 months. Tear production and ocular health were not affected in 17 eyes with at least six months follow-up. There was a statistically significant increase between preoperative and postoperative Schirmer tear test-1 measurements.


Subject(s)
Dog Diseases/surgery , Exocrine Glands/surgery , Eyelid Diseases/veterinary , Nictitating Membrane/surgery , Age Factors , Animals , Breeding , Dog Diseases/pathology , Dogs , Exocrine Glands/pathology , Eyelid Diseases/pathology , Eyelid Diseases/surgery , Female , Male , Nictitating Membrane/pathology , Prolapse , Prospective Studies , Recurrence , Suture Techniques/veterinary , Tears/metabolism , Treatment Outcome
4.
Br J Pharmacol ; 162(4): 947-60, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21039416

ABSTRACT

BACKGROUND AND PURPOSE: By interacting with trkB receptors, brain-derived neurotrophic factor (BDNF) triggers various signalling pathways responsible for neurone survival, differentiation and modulation of synaptic transmission. Numerous reports have implicated BDNF and trkB in the pathogenesis of various central nervous system affections and in cancer, thus representing trkB as a promising therapeutic target. In this study, we used an antibody-based approach to search for trkB-selective functional reagents. EXPERIMENTAL APPROACH: Six commercially available polyclonal and monoclonal antibodies were tested on recombinant and native, human and rodent trkB receptors. Functional and pharmacological characterization was performed using a modified version of the KIRA-elisa method and radioligand binding studies. Western blot analyses and neurite outgrowth assays were carried out to determine the specificity and selectivity of antibody effects. The survival properties of one antibody were further assessed on cultured neurones in a serum-deprived paradigm. KEY RESULTS: The functional trkB-selective antibodies showed distinct pharmacological profiles, ranging from partial agonists to antagonists, acting on trkB receptors through allosteric modulations. The same diversity of effects was observed on the mitogen-activated protein kinase signalling pathway downstream of trkB and on the subsequent neurite outgrowth. One antibody with partial agonist activity demonstrated cell survival properties by activating the Akt pathway. Finally, these antibodies were functionally validated as true trkB-selective ligands because they failed activating trkA or trkC, and contrary to BDNF, none of them bind to p75(NTR). CONCLUSIONS AND IMPLICATIONS: These trkB-selective antibodies represent a novel class of pharmacological tools to explore the pathophysiological roles of trkB and its potential therapeutic relevance for the treatment of various disorders.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies/pharmacology , Receptor, trkB/metabolism , Allosteric Regulation , Animals , Antibodies/immunology , Antibodies, Monoclonal/immunology , Antibody Specificity , Brain-Derived Neurotrophic Factor/metabolism , CHO Cells , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cricetinae , Cricetulus , Embryo, Mammalian , Humans , Mice , Neurites/drug effects , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Rats , Receptor, trkB/agonists , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects
5.
J Neurochem ; 96(3): 815-24, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16405506

ABSTRACT

Cerebral ischaemia is associated with brain damage and inhibition of neuronal protein synthesis. A deficit in neuronal metabolism and altered excitatory amino acid release may both contribute to those phenomena. In the present study, we demonstrate that both NMDA and metabolic impairment by 2-deoxyglucose or inhibitors of mitochondrial respiration inhibit protein synthesis in cortical neurons through the phosphorylation of eukaryotic elongation factor (eEF-2), without any change in phosphorylation of initiation factor eIF-2alpha. eEF-2 kinase may be activated both by Ca(2+)-independent AMP kinase or by an increase in cytosolic Ca2+. Although NMDA decreases ATP levels in neurons, only the effects of 2-deoxyglucose on protein synthesis and phosphorylation of elongation factor eEF-2 were reversed by Na(+) pyruvate. Protein synthesis inhibition by 2-deoxyglucose was not as a result of a secondary release of glutamate from cortical neurons as it was not prevented by the NMDA receptor antagonist 5-methyl-10,11-dihydro-5H-dibenzo-(a,d)-cyclohepten-5,10-imine hydrogen maleate (MK 801), nor to an increase in cytosolic-free Ca2+. Conversely, 2-deoxyglucose likely activates eEF-2 kinase through a process involving phosphorylation by AMP kinase. In conclusion, we provide evidence that protein synthesis can be inhibited by NMDA and metabolic deprivation by two distinct mechanisms involving, respectively, Ca(2+)-dependent and Ca(2+)-independent eEF-2 phosphorylation.


Subject(s)
Antimetabolites/pharmacology , Deoxyglucose/pharmacology , Excitatory Amino Acid Agonists/pharmacology , N-Methylaspartate/pharmacology , Neurons/drug effects , Peptide Elongation Factor 2/metabolism , Animals , Blotting, Western/methods , Calcium/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Ionophores/pharmacology , Leucine/metabolism , Mice , Models, Biological , Neurons/physiology , Oligomycins/pharmacology , Phosphorylation/drug effects , Protein Kinases/metabolism , Protein Synthesis Inhibitors/pharmacology , Pyruvic Acid/pharmacokinetics , Sodium Azide/pharmacology , TOR Serine-Threonine Kinases , Time Factors , Tritium/metabolism
6.
Neuroscience ; 114(4): 859-67, 2002.
Article in English | MEDLINE | ID: mdl-12379242

ABSTRACT

Transient cerebral ischemia, which is accompanied by a sustained release of glutamate, strongly depresses protein synthesis. We have previously demonstrated in cortical neurons that a glutamate-induced increase in intracellular Ca(2+) is likely responsible for the blockade of the elongation step of protein synthesis. In this study, we provide evidence indicating that NMDA mobilizes a thapsigargin-sensitive pool of intracellular Ca(2+). Exposure of cortical neurons to NMDA, in the absence of external Ca(2+), produced a transient rise in intracellular Ca(2+) that was suppressed by pretreatment with thapsigargin. This rise in intracellular Ca(2+) did not result from an influx of Na(+) via reversal of the mitochondrial Na(+)/Ca(2+) exchanger since it persisted in a Na(+)-free medium or in the presence of CGP 37157, an inhibitor of the exchanger. Moreover, the NMDA-induced increase in intracellular Ca(2+) required the presence of D-serine, was blocked by D(-)-2-amino-5-phosphonopentanoic acid, but was not reduced in the presence of external Mg(2+). This unexpected non-ionotropic effect of NMDA was associated with an inhibition of protein synthesis that was also insensitive to the absence of external Ca(2+) or Na(+), or presence of Mg(2+). NMDA treatment resulted in an increase in the phosphorylation of eEF-2 in the absence or presence of external Ca(2+). The initiation step of protein synthesis was not blocked by NMDA since the phosphorylation of initiation factor eIF-2alpha subunit was not altered by NMDA treatment. In conclusion, we provide evidence indicating that NMDA can inhibit protein synthesis in cortical neurons through a process that involves the mobilization of intracellular Ca(2+) stores via a mechanism that is not linked to the ionic properties of NMDA receptors.


Subject(s)
Cerebral Cortex/cytology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Thapsigargin/pharmacology , Animals , Calcium/metabolism , Calcium/pharmacology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Excitatory Amino Acid Agonists/pharmacology , Glutamic Acid/metabolism , Mice , N-Methylaspartate/pharmacology , Phosphorylation , Protein Biosynthesis
7.
Mol Cell Neurosci ; 20(3): 415-28, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12139919

ABSTRACT

Edg-2 is a member of the G-protein-coupled seven-transmembrane receptor family recently identified in oligodendrocytes. Here we show that both in vitro and in vivo, Edg-2 transcripts are not detected during early stages of oligodendroglial development, but are expressed only in mature oligodendrocytes, shortly before the onset of myelination. Lysophosphatidic acid (LPA) has been reported to be a ligand of Edg-2 receptor in different cell types. However, in oligodendroglial cultures, LPA had no effect on survival, maturation, or cytoskeleton organization. In myelinating oligodendrocyte-neuron cocultures, LPA did not influence myelinogenesis. In addition, LPA failed to induce Ca2+ mobilization and had no effect on forskolin-induced cAMP accumulation. Phosphorylation of the ERK1/ERK2 MAP kinases was the only response elicited by LPA in oligodendrocytes. Therefore, in contrast to other cell types, in which LPA exerts pleiotropic effects, Edg-2-positive postmitotic oligodendrocytes display a restricted responsiveness to LPA.


Subject(s)
Lysophospholipids/pharmacology , Nuclear Proteins/biosynthesis , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Transcription Factors/biosynthesis , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/genetics , Oligodendroglia/cytology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Receptors, Lysophosphatidic Acid , Transcription Factors/genetics
8.
Eur J Neurosci ; 13(12): 2067-76, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11467306

ABSTRACT

Sphingosine-1-phosphate (S1P) is a potent lysophospholipid mediator mostly released by activated platelets. It is involved in several functions in peripheral tissues, but its effects in the central nervous system are poorly documented. Therefore, we have examined the effects of S1P on the proliferation of striatal astrocytes from the mouse embryo. These cells have been found to express mRNAs for the S1P receptors, Edg-1 and Edg-3. S1P stimulated thymidine incorporation and induced activation of extracellular signal-regulated kinases (Erks). Both effects were prevented by U0126, an Erk kinase inhibitor. The S1P-evoked activation of Erk1 was totally blocked in astrocytes pretreated with a combination of either phorbol ester (24 h) and LY294002, or phorbol ester (24 h) and pertussis toxin (PTX). Each individual treatment only partially inhibited Erk1 activation. This suggests that several separate mechanisms mediate this process, one involving protein kinase C and another involving Gi/Go proteins and phosphatidylinositol 3-kinase. In contrast, the stimulatory effect of S1P on astrocyte proliferation was totally blocked by either PTX or LY294002, but not by a downregulation of protein kinase C. S1P dramatically inhibited the evoked production of cyclic AMP, a response that was impaired by PTX. Finally, S1P stimulated the production of inositol phosphates and increased intracellular calcium by mobilization from thapsigargin-sensitive stores. These latter effects were mainly insensitive to PTX. Probably, Gi/Go protein activation and phosphoinositide hydrolysis are early events that regulate the activation of Erks by S1P. Altogether, these observations show that astrocytes are targets for S1P. Their proliferation in response to S1P could have physiopathological consequences at sites of brain lesions and alterations of the blood-brain barrier.


Subject(s)
Astrocytes/drug effects , I-kappa B Proteins , Lysophospholipids , Neostriatum/drug effects , Receptors, G-Protein-Coupled , Signal Transduction/drug effects , Sphingosine/metabolism , Sphingosine/pharmacology , Animals , Astrocytes/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Calcium/metabolism , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cyclic AMP/metabolism , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gliosis/metabolism , Gliosis/physiopathology , Immediate-Early Proteins/genetics , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Mice , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha , Neostriatum/embryology , Neostriatum/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Lysophospholipid , Signal Transduction/physiology , Sphingosine/analogs & derivatives
9.
Eur J Neurosci ; 13(11): 2067-76, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11422447

ABSTRACT

Sphingosine-1-phosphate (S1P) is a potent lysophospholipid mediator mostly released by activated platelets. It is involved in several functions in peripheral tissues, but its effects in the central nervous system are poorly documented. Therefore, we have examined the effects of S1P on the proliferation of striatal astrocytes from the mouse embryo. These cells have been found to express mRNAs for the S1P receptors, Edg-1 and Edg-3. S1P stimulated thymidine incorporation and induced activation of extracellular signal-regulated kinases (Erks). Both effects were prevented by U0126, an Erk kinase inhibitor. The S1P-evoked activation of Erk1 was totally blocked in astrocytes pretreated with a combination of either phorbol ester (24 h) and LY294002, or phorbol ester (24 h) and pertussis toxin (PTX). Each individual treatment only partially inhibited Erk1 activation. This suggests that several separate mechanisms mediate this process, one involving protein kinase C and another involving Gi/Go proteins and phosphatidylinositol 3-kinase. In contrast, the stimulatory effect of S1P on astrocyte proliferation was totally blocked by either PTX or LY294002, but not by a downregulation of protein kinase C. S1P dramatically inhibited the evoked production of cyclic AMP, a response that was impaired by PTX. Finally, S1P stimulated the production of inositol phosphates and increased intracellular calcium by mobilization from thapsigargin-sensitive stores. These latter effects were mainly insensitive to PTX. Probably, Gi/Go protein activation and phosphoinositide hydrolysis are early events that regulate the activation of Erks by S1P. Altogether, these observations show that astrocytes are targets for S1P. Their proliferation in response to S1P could have physiopathological consequences at sites of brain lesions and alterations of the blood-brain barrier.


Subject(s)
Astrocytes/drug effects , Cell Division/drug effects , Gliosis/metabolism , I-kappa B Proteins , Lysophospholipids , Neostriatum/drug effects , Receptors, G-Protein-Coupled , Sphingosine/metabolism , Sphingosine/pharmacology , Animals , Astrocytes/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Calcium/metabolism , Cell Division/physiology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cyclic AMP/metabolism , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gliosis/physiopathology , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Heterotrimeric GTP-Binding Proteins/metabolism , Immediate-Early Proteins/genetics , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Mice , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha , Neostriatum/embryology , Neostriatum/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Lysophospholipid , Sphingosine/analogs & derivatives
10.
J Neurochem ; 76(4): 1080-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11181828

ABSTRACT

Transient cerebral ischemia, which is accompanied by a sustained release of glutamate and zinc, as well as H(2)O(2) formation during the reperfusion period, strongly depresses protein synthesis. We have previously demonstrated that the glutamate-induced increase in cytosolic Ca(2+) is likely responsible for blockade of the elongation step of protein synthesis, whereas Zn(2+) preferentially inhibits the initiation step. In this study, we provide evidence indicating that H(2)O(2) and thapsigargin mobilized a common intracellular Ca(2+) pool. H(2)O(2) treatment stimulated a slow increase in intracellular Ca(2+), and precluded the effect of thapsigargin on Ca(2+) mobilization. H(2)O(2) stimulated the phosphorylation of both eIF-2alpha and eEF-2, in a time- and dose-dependent manner, suggesting that both the blockade of the elongation and of the initiation step are responsible for the H(2)O(2)-induced inhibition of protein synthesis. However, kinetic data indicated that, at least during the first 15 min of H(2)O(2) treatment, the inhibition of protein synthesis resulted mainly from the phosphorylation of eEF-2. In conclusion, H(2)O(2) inhibits protein translation in cortical neurons by a process that involves the phosphorylation of both eIF-2alpha and eEF-2 and the relative contribution of these two events depends on the duration of H(2)O(2) treatment.


Subject(s)
Cerebral Cortex/metabolism , Hydrogen Peroxide/pharmacology , Neurons/metabolism , Protein Biosynthesis , Protein Synthesis Inhibitors/pharmacology , Aniline Compounds , Animals , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Eukaryotic Initiation Factor-2/metabolism , Fluorescent Dyes , Intracellular Fluid/metabolism , Mice , Neurons/cytology , Neurons/drug effects , Peptide Chain Elongation, Translational/drug effects , Peptide Elongation Factor 2/metabolism , Phosphorylation/drug effects , Proteins/analysis , Thapsigargin/pharmacology , Xanthenes
11.
Eur J Neurosci ; 14(12): 1953-60, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11860490

ABSTRACT

Glutamatergic transmission is mediated by ionotropic receptors that directly gate cationic channels and metabotropic receptors that are coupled to second messenger generating systems and to ionic channels via heterotrimeric guanine-nucleotide binding- (G) proteins. This distinction cannot be made for the ionotropic receptor subclass activated by alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA), which has been shown to be physically associated with the alpha-subunit of Gi1 protein and activates this G-protein. Here, we report that, in addition to a Ca2+ influx, AMPA induces the mobilization of Ca2+ from the mitochondrial pool by reversing the mitochondrial Na+/Ca2+ exchanger in mouse neurons in primary culture. Both processes required the activation of tetrodotoxin-sensitive Na+ channels. AMPA receptor activation modified the gating properties of the Na+ channel, independently of the AMPA current, suggesting a G-protein-mediated process. Indeed, co-immunoprecipitation experiments indicated that AMPA receptor activation induced the association of Gbeta with the alpha-subunit of the Na+ channel. These results suggest that, in addition to its ionic channel function, the AMPA receptor is coupled to Na+ channels through G-proteins and that this novel metabotropic function is involved in the control of neuronal excitability.


Subject(s)
Calcium Signaling/physiology , Central Nervous System/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Sodium Channels/metabolism , Synaptic Transmission/physiology , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Fetus , Heterotrimeric GTP-Binding Proteins/drug effects , Immunohistochemistry , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/drug effects , Neural Inhibition/drug effects , Neural Inhibition/physiology , Pregnancy , Receptors, AMPA/drug effects , Sodium Channels/drug effects , Sodium-Calcium Exchanger/drug effects , Sodium-Calcium Exchanger/metabolism , Synaptic Transmission/drug effects , Tetrodotoxin/pharmacology
12.
Eur J Neurosci ; 12(1): 8-18, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10651855

ABSTRACT

Exposure of central neurons to Zn2+ triggers neuronal death. The routes of Zn2+ entry were investigated in living cortical neurons from the mouse using the specific Zn2+ fluorescent dye N-(6-methoxy-8-quinolyl)-p-toluene sulphonamide (TSQ), which preferentially detects membrane-bound Zn2+. Exposure of cortical neurons to increasing concentrations of Zn2+ (1-100 microM) induced a progressive increase in the fluorescence of TSQ. This fluorescence signal was not attenuated by the permeation of plasma membrane with digitonin. Accordingly, the major part of TSQ fluorescence (two-thirds) was associated to the particulate fraction of cortical neurons exposed to Zn2+. These results suggest that Zn2+ detected with TSQ in neurons is mainly bound to membranes. TSQ fluorescence measured in neurons exposed to 3 microM Zn2+ was enhanced by Na+-pyrithione, a Zn2+ ionophore, alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA), N-methyl-D-aspartate (NMDA) or KCl-induced depolarization. However, in the absence of any treatment, TSQ labelling of neurons exposed to 3 microM Zn2+ was only decreased by NMDA receptor antagonists, whereas it remained unaltered in the presence of antagonists of AMPA receptors or L-type voltage-gated Ca2+ channels. Zn2+ entry through NMDA receptors did not contribute to Zn2+-induced neuronal death, as it was prevented by antagonists of NMDA receptors only when they were added after the Zn2+ exposure. Finally, Zn2+ induced a delayed accumulation of extracellular glutamate which might be responsible for the delayed NMDA receptor activation that leads to neuronal death.


Subject(s)
Cerebral Cortex/physiology , Neurons/physiology , Zinc/pharmacokinetics , Adenosine Triphosphate/metabolism , Aminoquinolines , Animals , Cell Membrane/physiology , Cell Membrane Permeability , Cells, Cultured , Cerebral Cortex/cytology , Digitonin/pharmacology , Fluorescent Dyes , Glutamic Acid/metabolism , Indoles , Mice , Microscopy, Fluorescence , Microscopy, Video , Mitochondria/metabolism , Neurons/cytology , Neurons/drug effects , Neurotoxins/pharmacokinetics , Neurotoxins/toxicity , Oxygen Consumption , Tosyl Compounds , Zinc/toxicity
13.
J Biol Chem ; 274(45): 32433-8, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542287

ABSTRACT

In the central nervous system, Zn(2+) is concentrated in the cerebral cortex and hippocampus and has been found to be toxic to neurons. In this study, we show that exposure of cultured cortical neurons from mouse to increasing concentrations of Zn(2+) (10-300 microM) induces a progressive decrease in global protein synthesis. The potency of Zn(2+) was increased by about 2 orders of magnitude in the presence of Na(+)-pyrithione, a Zn(2+) ionophore. The basal rate of protein synthesis was restored 3 h after Zn(2+) removal. Zn(2+) induced a sustained increase in phosphorylation of the alpha subunit of the translation eukaryotic initiation factor-2 (eIF-2alpha), whereas it triggered a transient increase in phosphorylation of eukaryotic elongation factor-2 (eEF-2). Protein synthesis was still depressed 60 min after the onset of Zn(2+) exposure while the state of eEF-2 phosphorylation had already returned to its basal level. Moreover, Zn(2+) was less effective than glutamate to increase eEF-2 phosphorylation, whereas it induced a more profound inhibition of protein synthesis. These results suggest that Zn(2+)-induced inhibition of protein synthesis mainly correlates with the increase in eIF-2alpha phosphorylation. Supporting further that Zn(2+) acts at the initiation step of protein synthesis, it strongly decreased the amount of polyribosomes.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Neurons/metabolism , Protein Biosynthesis , Zinc/physiology , Animals , Calcium Channels/metabolism , Cells, Cultured , Leucine/metabolism , Methionine/metabolism , Mice , Phosphorylation , Polyribosomes/metabolism
14.
Eur J Neurosci ; 11(9): 3215-24, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10510185

ABSTRACT

A sustained release of glutamate contributes to neuronal loss during cerebral ischaemia. Using cultured mouse striatal neurons, we observed that glucose deprivation, which occurs in this pathological process, enhanced the N-Methyl-D-aspartate (NMDA)- or alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)-induced neurotoxicity. The end products of glycolysis, lactate and pyruvate, strongly protected neurons from these neurotoxic effects. The neuroprotective effect of pyruvate (which is more prominent in the absence of glucose) was not related to its ability to react with H2O2 by a decarboxylation process. Pyruvate and L-lactate strongly counteracted the deep decrease in the neuronal ATP content induced by NMDA, indicating that they might protect striatal neurons by rescuing cellular energy charge. Addition of MK-801 after the NMDA withdrawal completely protected neurons, suggesting that NMDA neurotoxicity resulted from a delayed NMDA receptor activation probably linked to a delayed release of an endogenous agonist in the extracellular medium. The strong accumulation of extracellular glutamate which was found in both sham and NMDA-treated cultures was markedly decreased by pyruvate. Thus, pyruvate might also exert its protecting activity by decreasing the delayed accumulation of glutamate which seemed to be neurotoxic only after a preexposure of neurons to NMDA.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , Lactic Acid/therapeutic use , N-Methylaspartate/toxicity , Neostriatum/pathology , Neurons/pathology , Neurotoxicity Syndromes/prevention & control , Pyruvic Acid/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Energy Metabolism/drug effects , Energy Metabolism/physiology , Free Radical Scavengers/pharmacology , Glucose/physiology , Glutamic Acid/metabolism , Glycolysis/physiology , Kinetics , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neostriatum/cytology , Neostriatum/drug effects , Neurons/drug effects , Neurotoxicity Syndromes/pathology , Oxygen Consumption/drug effects
15.
J Neurochem ; 73(3): 1181-8, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10461910

ABSTRACT

The present study aims to investigate the role of extracellular glutamate and NMDA receptor stimulation in the neuronal death induced by a transient exposure to H2O2 of cultured neurons originating from mouse cerebral cortex. Most of the neuronal loss following a transient exposure to H2O2 of cortical neurons results from an apoptotic process involving a secondary stimulation of NMDA receptors, which occurs after H2O2 washout. Indeed, (a) the neurotoxic effect of H2O2 was strongly reduced by antagonists of NMDA receptors, (b) the neurotoxic effect of H2O2 was enhanced in the absence of Mg2+, (c) the protective effect of MK-801 progressively decayed when it was applied with increasing delay time after H2O2 exposure, and (d), finally, the extracellular concentration of glutamate was increased after H2O2 exposure. The major part of H2O2-induced neurotoxicity is mediated by the formation of hydroxyl radicals, which might be involved in (a) the delayed accumulation of extracellular glutamate and NMDA receptor activation and (b) the poly(ADP-ribose) polymerase activation and the related NAD content decrease. The combination of these two mechanisms could lead to both an increase in ATP consumption and a decrease of ATP synthesis. The resulting large decrease in ATP content might be finally responsible for the neuronal death.


Subject(s)
Apoptosis/drug effects , Glutamic Acid/metabolism , Hydrogen Peroxide/metabolism , Neurons/drug effects , Oxidants/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Adenosine Triphosphate/physiology , Animals , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Extracellular Space/metabolism , Hydrogen Peroxide/toxicity , Hydroxyl Radical/metabolism , Mice , NAD/metabolism , Oxidants/toxicity , Poly(ADP-ribose) Polymerases/metabolism
16.
J Physiol ; 510 ( Pt 2): 429-40, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9705994

ABSTRACT

Indo-1 and fluo-3 imaging techniques were used to investigate the role of gap junctions in the changes in cytosolic calcium concentrations ([Ca2+]i) induced by several receptor agonists. Subpopulations of confluent cultured astrocytes from the rat striatum were superfused with submaximal concentrations of endothelin-1 (Et1) and the alpha 1-adrenergic and muscarinic receptor agonists, methoxamine and carbachol, respectively. 2. Combined binding and autoradiographic studies indicated that all striatal astrocytes possess binding sites for Et1. In contrast, alpha 1-adrenergic and muscarinic binding sites were found to be heterogeneously distributed. In agreement with these findings, Et1 induced fast calcium responses in all cells while only subsets of striatal astrocytes responded to the application of methoxamine or carbachol. 3. Halothane, heptanol and octanol, which are commonly used as gap junction inhibitors, drastically reduced the amplitude of Et1-induced calcium responses. In contrast, 18-alpha-glycyrrhetinic acid (alpha GA) used at a concentration known to block gap junction permeability in astrocytes had no significant effect on the amplitude of these calcium responses. 4. As demonstrated by quantitative and topological analysis, Et1 application similarly increased [Ca2+]i levels in all astrocytes in both the absence and presence of alpha GA. 5. In control conditions, subpopulations of cells responding to methoxamine or carbachol exhibited two main types of calcium responses which differed in their shape and kinetic characteristics. In the presence of alpha GA the number of cells responding to these receptor agonists was significantly reduced. Indeed, responses characterized by their long latency, slow rise time and weak amplitude disappeared in the presence of alpha GA while responses with short latency and fast rise time were preserved. 6. These results indicate that permeable gap junction channels tend to attenuate the pharmacological and functional heterogeneity of populations of astrocytes, while their inhibition restricts calcium responses in astrocytes expressing high densities of transmitter receptors coupled to phospholipase C.


Subject(s)
Astrocytes/metabolism , Cell Communication/physiology , Gap Junctions/physiology , Neostriatum/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Autoradiography , Calcium/metabolism , Cell Communication/drug effects , Cells, Cultured , Endothelin-1/metabolism , Gap Junctions/drug effects , Immunohistochemistry , Kinetics , Microscopy, Fluorescence , Neostriatum/cytology , Neostriatum/drug effects , Rats , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Endothelin/drug effects
17.
J Neurosci ; 17(23): 9060-7, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9364052

ABSTRACT

Hydrogen peroxide (H2O2) is suspected to be involved in numerous brain pathologies such as neurodegenerative diseases or in acute injury such as ischemia or trauma. In this study, we examined the ability of pyruvate to improve the survival of cultured striatal neurons exposed for 30 min to H2O2, as estimated 24 hr later by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazoliumbromide assay. Pyruvate strongly protected neurons against both H2O2 added to the external medium and H2O2 endogenously produced through the redox cycling of the experimental quinone menadione. The neuroprotective effect of pyruvate appeared to result rather from the ability of alpha-ketoacids to undergo nonenzymatic decarboxylation in the presence of H2O2 than from an improvement of energy metabolism. Indeed, several other alpha-ketoacids, including alpha-ketobutyrate, which is not an energy substrate, reproduced the neuroprotective effect of pyruvate. In contrast, lactate, a neuronal energy substrate, did not protect neurons from H2O2. Optimal neuroprotection was achieved with relatively low concentrations of pyruvate (

Subject(s)
Corpus Striatum/cytology , Hydrogen Peroxide/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Pyruvic Acid/pharmacology , Animals , Butyrates/pharmacology , Cells, Cultured , Energy Metabolism/drug effects , Hydrogen-Ion Concentration , Ketoglutaric Acids/pharmacology , Lactic Acid/pharmacology , Mice , Oxaloacetates/pharmacology , Oxidation-Reduction , Oxidative Stress/drug effects , Vitamin K/metabolism
18.
J Neurosci ; 17(9): 2939-46, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9096130

ABSTRACT

During neuropathological states associated with inflammation, the levels of cytokines such as interleukin-1beta (IL-1beta) are increased. Several studies have suggested that the neuronal damage observed in pathogenesis implicating IL-1beta are caused by an alteration in the neurochemical interactions between neurons and astrocytes. We report here that treating striatal astrocytes in primary culture with IL-1beta for 22-24 hr enhances the ATP-evoked release of arachidonic acid (AA) with no effect on the ATP-induced accumulation of inositol phosphates. The molecular mechanism responsible for this effect involves the expression of P2Y2 receptors (a subtype of purinoceptor activated by ATP) and cytosolic phospholipase A2 (cPLA2, an enzyme that mediates AA release). Indeed, P2Y2 antisense oligonucleotides reduce the ATP-evoked release of AA only from IL-1beta-treated astrocytes. Further, both the amount of cPLA2 (as assessed by Western blotting) and the release of AA resulting from direct activation of cPLA2 increased fourfold in cells treated with IL-1beta. We also report evidence indicating that the coupling of newly expressed P2Y2 receptors to cPLA2 is dependent on PKC activity. These results suggest that during inflammatory conditions, IL-1beta reveals a functional P2Y2 signaling pathway in astrocytes that results in a dramatic increase in the levels of free AA. This pathway may thus contribute to the neuronal loss associated with cerebral ischemia or traumatic brain injury.


Subject(s)
Adenosine Triphosphate/metabolism , Arachidonic Acids/metabolism , Astrocytes/drug effects , Interleukin-1/pharmacology , Animals , Dose-Response Relationship, Drug , Mice
19.
J Neurosci ; 17(10): 3445-54, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9133370

ABSTRACT

Postischemic delayed neuronal death is attributed to excitotoxic activation of glutamate receptors. It is preceded by a persistent inhibition of protein synthesis, the molecular basis of which is not known. Here we have examined in cortical neurons in culture the regulation by glutamate of phosphorylation of eukaryotic elongation factor-2 (eEF-2) by eEF-2 kinase, a Ca2+/calmodulin-dependent enzyme. Using a phosphorylation state-specific antibody, we show that glutamate, which triggers a large influx of Ca2+, enhances dramatically the phosphorylation of eEF-2. On the basis of kinetic and pharmacological analysis, we demonstrate a close correlation among the increase in cytosolic Ca2+ concentration, the degree of eEF-2 phosphorylation, and the inhibition of protein synthesis. A 30 min treatment with NMDA induced a transient phosphorylation of eEF-2 and delayed neuronal death. However, pharmacological inhibition of protein translation was not neurotoxic by itself and protected neurons against the toxicity evoked by low concentrations of NMDA. Thus, phosphorylation of eEF-2 and the resulting depression of protein translation may have protective effects against excitotoxicity and open new perspectives for understanding long-term effects of glutamate.


Subject(s)
Glutamic Acid/pharmacology , Neurons/metabolism , Peptide Elongation Factors/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Antibody Specificity , Calcium/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured/chemistry , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cerebral Cortex/cytology , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Mice , Nerve Tissue Proteins/metabolism , Neurons/chemistry , Neurons/cytology , Neurotoxins/pharmacology , Peptide Elongation Factor 2 , Peptide Elongation Factors/immunology , Phosphorylation , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Receptors, AMPA/agonists , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/physiology
20.
J Pharmacol Exp Ther ; 280(3): 1277-83, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9067314

ABSTRACT

A protective effect of nicotine against glutamate-induced neurotoxicity has previously been reported in cultured striatal and cortical neurons. The aim of this study was to investigate whether nicotine also inhibits glutamate-evoked arachidonic acid release from cultured striatal neurons. (-)-Nicotine selectively inhibited the release of [3H]-arachidonic acid induced by the joint stimulation of alpha-amino-3-isoxazol-5-propionic acid and metabotropic receptors, whereas the response evoked by the sole activation of N-methyl-D-aspartate receptors remained unchanged. The inhibitory effect of (-)-nicotine was not mediated by nicotinic receptors because it was neither reproduced by acetylcholine (in the presence of atropine) or 1,1-dimethyl-4-phenyl piperazinium, nor reversed by dihydro-beta-erythroidine or hexamethonium, two central nicotinic receptor antagonists. (-)-Nicotine, which induced rapidly desensitizing inward currents in 17% of striatal neurons, did not alter the alpha-amino-3-isoxazol-5-propionic acid-evoked currents. Moreover, (-)-nicotine did not inhibit the accumulation of inositol phosphate derivatives induced by agonists of glutamate metabotropic receptors. In fact, using the fluorogenic phospholipase A2 substrate 1,2-bis-(1-pyrenedecanoyl)-sn-glycero-3-phosphocholine, (-)-nicotine was found to inhibit both particulate and soluble phospholipase A2 activities from striatal neurons. Therefore, (-)-nicotine can modulate a neuronal response (arachidonic acid release) evoked by glutamate but this process is not involved in the neuroprotective effect of the drug on glutamate-induced neurotoxicity.


Subject(s)
Neurons/drug effects , Nicotine/pharmacology , Phospholipases A/antagonists & inhibitors , Animals , Arachidonic Acid/metabolism , Cells, Cultured , Inositol Phosphates/metabolism , Mice , Neurons/enzymology , Neurons/metabolism , Phospholipases A2 , Receptors, AMPA/drug effects , Receptors, Metabotropic Glutamate/drug effects , Receptors, Nicotinic/metabolism , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...