Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
Add more filters










Publication year range
1.
Br J Anaesth ; 91(3): 390-6, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12925480

ABSTRACT

BACKGROUND: We studied the potential neuroprotective effects of halothane and desflurane, compared with the awake state, on infarct size following 2 h of intraluminal middle cerebral artery occlusion (MCAo) and 22 h of reperfusion. METHODS: Male Sprague-Dawley rats were anaesthetized with desflurane or halothane, intubated, and mechanically ventilated. Mean arterial pressure (MAP), blood gases, and pH were controlled. Body temperature was maintained at 37.5-38 degrees C. Animals were assigned to one of four groups according to the anaesthetic type (halothane or desflurane) and the duration of anaesthesia: "short-duration", during the preparation only; "long-duration", during both preparation and ischaemia. Twenty-four hours after MCAo, infarcts were visualized by staining with 2,3,5-triphenyltetrazolium chloride. Two additional groups of rats were subjected to the same protocol as that of long-duration halothane and long-duration desflurane with additional pericranial temperature measurements made. RESULTS: Physiological parameters were comparable between the groups but MAP was higher (P<0.0001) in the short-duration groups. In the short-duration groups, cerebral infarct volumes were not significantly different between anaesthetics (short-duration halothane: 288 (61) mm(3), mean (SD); short-duration desflurane: 269 (71) mm(3), P>0.56). Compared with the awake state (short-duration groups), halothane and desflurane significantly reduced infarct volumes (long-duration halothane: 199 (54) mm(3), P<0.0047 vs short-duration halothane; long-duration desflurane: 121 (55) mm(3), P<0.0001 vs short-duration desflurane). The mean infarct volume in the long-duration desflurane group was significantly lower than that in the long-duration halothane group (P<0.0053). Pericranial temperatures were similar in the desflurane and halothane long-duration groups (P>0.17). CONCLUSIONS: In rats, desflurane-induced neuroprotection against focal cerebral ischaemia was greater than that conferred by halothane.


Subject(s)
Anesthetics, Inhalation/therapeutic use , Cerebral Infarction/prevention & control , Halothane/therapeutic use , Isoflurane/analogs & derivatives , Isoflurane/therapeutic use , Neuroprotective Agents/therapeutic use , Analysis of Variance , Animals , Blood Pressure/drug effects , Brain Ischemia/complications , Carbon Dioxide/blood , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Desflurane , Drug Administration Schedule , Hydrogen-Ion Concentration/drug effects , Male , Oxygen/blood , Partial Pressure , Rats , Rats, Sprague-Dawley
2.
FASEB J ; 17(3): 443-5, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12514118

ABSTRACT

Although angiopoietin-1 (Ang-1) is recognized as an endothelial growth factor, its presence in brain following an ischemic event suggests a role in the evolution of neuronal damage. Using primary neuronal cultures, we showed that neurons express Ang-1 and possess the functional angiopoietin-receptor Tie-2, which is phosphorylated in the presence of Ang-1. We further investigated in vitro whether Ang-1 could protect neurons against either excitotoxic necrosis or apoptosis induced by serum deprivation (SD). A neuroprotective effect for Ang-1 was detected exclusively in the apoptotic paradigm. Treatment of cells with the phosphatidyl-inositol 3-kinase (PI3-K) inhibitor, LY294002, inhibited Ang-1-induced phosphorylation of Akt, restored the cleavage of the effector caspase-3, and reduced the protective effect of Ang-1 against SD-induced toxicity. These findings suggest that Ang-1 has a neuroprotective effect against apoptotic stress and that this effect is dependent on the PI3-K/Akt pathway and inhibition of caspase-3 cleavage. This study provides evidence that Ang-1 is not just angiogenic but also neuroprotective. The understanding of neuroprotective mechanisms induced by Ang-1 may promote strategies based on the pleiotropic effects of angiogenic factors. Such approaches could be useful for the treatment of brain diseases in which both neuronal death and angiogenesis are involved.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Apoptosis , Membrane Glycoproteins/pharmacology , Neurons/enzymology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins , Angiopoietin-1 , Animals , Cells, Cultured , Chromones/pharmacology , Culture Media, Serum-Free , Enzyme Activation , Enzyme Inhibitors/pharmacology , Mice , Models, Biological , Morpholines/pharmacology , Neoplasm Proteins/analysis , Neoplasm Proteins/physiology , Neurons/cytology , Neurons/drug effects , Phosphoinositide-3 Kinase Inhibitors , Receptor, TIE-2
3.
J Biol Chem ; 276(49): 46243-50, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11544249

ABSTRACT

Transforming growth factor-beta (TGF-beta) signaling requires a ligand-dependent interaction of TGF-beta receptors Tau beta R-I and Tau beta R-II. It has been previously demonstrated that a soluble TGF-beta type II receptor could be used as a TGF-beta antagonist. Here we have generated and investigated the biochemical and signaling properties of a soluble TGF-beta type I receptor (Tau beta RIs-Fc). As reported for the wild-type receptor, the soluble Tau beta R-I does not bind TGF-beta 1 on its own. Surprisingly, in the absence of TGF-beta1, the Tau beta RIs-Fc mimicked TGF-beta 1-induced transcriptional and growth responses in mink lung epithelial cells (Mv1Lu). Signaling induced by the soluble TGF-beta type I receptor is mediated via the obligatory presence of both TGF-beta type I and type II receptors at the cell surface since no signal was observed in Mv1Lu-derivated mutants for TGF-beta receptors R-1B and DR-26. The comparison between the structures of TGF-betas and a three-dimensional model of the extracellular domain of Tau beta RI has shown that five residues of the supposed binding site of TGF-beta 1 (Lys(31), His(34), Glu(5), Tyr(91), and Lys(94)) were found with equivalent biochemical properties and similar spatial positions.


Subject(s)
Receptors, Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Division/physiology , Cell Line , Cricetinae , DNA Primers , Immunoglobulin G/metabolism , Mink , Molecular Sequence Data , Protein Conformation , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Solubility , Transforming Growth Factor beta/metabolism
4.
J Cereb Blood Flow Metab ; 21(7): 820-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11435794

ABSTRACT

There has been an increasing interest in recent years in the evaluation of the neuronal and glial responses to ischemic insult. Some cytokines, including transforming growth factor-beta (TGF-beta), that are overexpressed after experimental stroke in rodents are thought to be implicated in the neuronal processes that lead to necrosis. Thus, such cytokines could predict tissue fate after stroke in humans, although data are currently sparse for gyrencephalic species. The current study addressed the expression pattern of TGF-beta1 in a nonhuman primate model of middle cerebral artery occlusion. Focal permanent ischemia was induced for 1 or 7 days in 6 baboons and the following investigations were undertaken: cerebral oxygen metabolism (CMRO2) positron emission tomography studies, magnetic resonance imaging, postmortem histology, and reverse transcription-polymerase chain reaction. The aim of the current study was to correlate the expression of TGF-beta1 to the underlying metabolic and histologic state of the threatened cerebral parenchyma. The authors evidenced increased TGF-beta1 mRNA levels (up to 25-fold) in those regions displaying a moderate (20% to 49%) reduction in CMRO2. The current findings suggest that the greatly enhanced expression of TGF-beta1 in the penumbral zones that surround tissue destined to infarction may represent a robust index of potentially salvageable brain. The current investigation, in the nonhuman primate, strengthens the authors' hypothesis, derived from rodent models, that TGF-beta1 may be involved in the physiopathology of human stroke.


Subject(s)
Biomarkers , Brain Ischemia/metabolism , Gene Expression , Neurons/physiology , Transforming Growth Factor beta/genetics , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/etiology , Brain Ischemia/pathology , Magnetic Resonance Imaging , Male , Middle Cerebral Artery/surgery , Oxygen Consumption , Papio , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tomography, Emission-Computed
5.
Bioorg Med Chem Lett ; 11(9): 1153-6, 2001 May 07.
Article in English | MEDLINE | ID: mdl-11354365

ABSTRACT

The synthesis and pharmacological evaluation of methoxyindazoles as new inhibitors of neuronal nitric oxide synthase are presented. The 7-methoxyindazole, although less potent than 7-NI, is the most active compound of the series in an in vitro enzymatic assay of neuronal nitric oxide synthase activity. This result shows that the nitro-substitution is not indispensable to the biological activity of the indazole ring. 7-Methoxyindazole possesses in vivo NOS inhibitory as well and related antinociceptive properties.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Indazoles/chemical synthesis , Indazoles/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Analgesics/chemical synthesis , Analgesics/pharmacology , Animals , Arginine/pharmacology , Cerebellum/drug effects , Cerebellum/enzymology , Dose-Response Relationship, Drug , Electrons , Mice , Nitric Oxide Synthase Type I , Pain Measurement/drug effects , Rats , Structure-Activity Relationship
6.
J Neurosci ; 21(9): 3024-33, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11312287

ABSTRACT

The glial cell line-derived neurotrophic factor (GDNF) is first characterized for its trophic activity on dopaminergic neurons. Recent data suggested that GDNF could modulate the neuronal death induced by ischemia. The purpose of this study was to characterize the influence of GDNF on cultured cortical neurons subjected to two paradigms of injury (necrosis and apoptosis) that have been identified during cerebral ischemia and to determine the molecular mechanisms involved. First, we demonstrated that both neurons and astrocytes express the mRNA and the protein for GDNF and its receptor complex (GFRalpha-1 and c-Ret). Next, we showed that the application of recombinant human GDNF to cortical neurons and astrocytes induces the activation of the MAP kinase (MAPK) pathway, as visualized by an increase in the phosphorylated forms of extracellular signal-regulated kinases (ERKs). Thereafter, we demonstrated that GDNF fails to prevent apoptotic neuronal death but selectively attenuates slowly triggered NMDA-induced excitotoxic neuronal death via a direct effect on cortical neurons. To further characterize the neuroprotective mechanisms of GDNF against NMDA-mediated neuronal death, we showed that a pretreatment with GDNF reduces NMDA-induced calcium influx. This effect likely results from a reduction of NMDA receptor activity rather than an enhanced buffering or extrusion capacity for calcium. Finally, we also demonstrated that an ERKs activation pathway is necessary for GDNF-mediated reduction of the NMDA-induced calcium response. Together, these results describe a novel mechanism by which the activation of MAPK induced by GDNF modulates NMDA receptor activity, a mechanism that could be responsible for the neuroprotective effect of GDNF in acute brain injury.


Subject(s)
Calcium/metabolism , Drosophila Proteins , MAP Kinase Signaling System/physiology , N-Methylaspartate/metabolism , Nerve Growth Factors , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/metabolism , Animals , Apoptosis/drug effects , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Brain Ischemia/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chelating Agents , Fluorescent Dyes , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Glycosylphosphatidylinositols/metabolism , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Mitogen-Activated Protein Kinases/metabolism , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/toxicity , Necrosis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism
7.
Curr Opin Neurol ; 14(1): 83-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11176222

ABSTRACT

The concept of an ischaemic penumbra, surrounding a focal cerebral lesion, is now widely accepted, although no universal definition of the 'penumbra' exists. In the present review, we consider the penumbra as that volume of brain tissue at the periphery of a focal, irreversibly damaged area that is threatened by recruitment into necrosis. Implicit to such a definition are several secondary concepts. First, the penumbra is both spatial, in that it surrounds the densely ischaemic core, but it is also temporal, in that its evolution toward infarction is a relatively progressive phenomenon. The pertinent literature is summarized. Second, penumbral tissue is potentially salvageable; the most recent animal studies are reviewed. Third, because electrically silent and pathologically damaged tissues have identical functional characteristics, it is evident that most clinical rating scales, be they neurological, behavioural, or psychological, are poorly adapted to address the problem of the penumbra. Finally, the penumbral tissue is remarkably and intensively 'active': multiple processes of cell death and repair occur and involve molecular mechanisms, electrophysiology and the vasculature.


Subject(s)
Brain Ischemia/diagnostic imaging , Brain Ischemia/physiopathology , Magnetic Resonance Imaging , Tomography, Emission-Computed , Animals , Brain Ischemia/therapy , Cerebrovascular Circulation , Electrophysiology , Humans , Neuroprotective Agents/therapeutic use
8.
Neuroreport ; 12(2): 289-93, 2001 Feb 12.
Article in English | MEDLINE | ID: mdl-11209937

ABSTRACT

The anaphylatoxin C3a is a potent inflammatory polypeptide released at sites of complement activation. To test whether C3a might alter neuronal outcome following an ischemic insult, we determined the effects of purified human C3a on murine primary cortical cell cultures exposed to apoptotic or excitotoxic paradigms. C3a prevented neither serum deprivation-induced apoptotic neuronal death, nor AMPA/kainate-mediated excitotoxicity. However, in mixed cultures of neurons and astrocytes, C3a dose-dependently protected neurons against NMDA toxicity (47% neuroprotection using 100 nM C3a, p < 0.01, n = 12). The neuroprotective effect of C3a was observable only in the presence of astrocytes. These observations suggest that C3a is involved in excitotoxicity-mediated neuronal death through astrocyte stimulation and extend its role beyond immune functions.


Subject(s)
Apoptosis/physiology , Complement C3a/genetics , Neurons/cytology , Anaphylatoxins/analysis , Anaphylatoxins/genetics , Animals , Apoptosis/drug effects , Astrocytes/chemistry , Astrocytes/cytology , Astrocytes/physiology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Complement C3a/analysis , Excitatory Amino Acid Agonists/toxicity , Gene Expression/physiology , Guinea Pigs , Mice , Mice, Inbred Strains , N-Methylaspartate/toxicity , Neurons/chemistry , Neurons/physiology , Neurotoxins/pharmacology , RNA, Messenger/analysis
9.
Nat Med ; 7(1): 59-64, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135617

ABSTRACT

Tissue-plasminogen activator (t-PA) is now available for the treatment of thrombo-embolic stroke but adverse effects have been reported in some patients, particularly hemorrhaging. In contrast, the results of animal studies have indicated that t-PA could increase neuronal damage after focal cerebral ischemia. Here we report for the first time that t-PA potentiates signaling mediated by glutamatergic receptors by modifying the properties of the N-methyl-D-aspartate (NMDA) receptor. When depolarized, cortical neurons release bio-active t-PA that interacts with and cleaves the NR1 subunit of the NMDA receptor. Moreover, the treatment with recombinant t-PA leads to a 37% increase in NMDA-stimulated fura-2 fluorescence, which may reflect an increased NMDA-receptor function. These results were confirmed in vivo by the intrastriatal injection of recombinant-PA, which potentiated the excitotoxic lesions induced by NMDA. These data provide insight into the regulation of NMDA-receptor-mediated signaling and could initiate therapeutic strategies to improve the efficacy of t-PA treatment in man.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Tissue Plasminogen Activator/metabolism , Animals , Calcium/metabolism , Cell Death , Hydrolysis , Ion Transport , Membrane Potentials , Neurons/metabolism , Neurons/physiology
10.
Brain Res ; 879(1-2): 122-9, 2000 Oct 06.
Article in English | MEDLINE | ID: mdl-11011013

ABSTRACT

Transient ischaemic depolarizations (IDs) are thought to play a key role in the pathogenesis of focal cerebral ischaemia. Most transient IDs are akin to spreading depression (SD), although a negative DC shift is not observed in half the cases. The other IDs may represent transient anoxic depolarizations. Using cortical DC and blood flow recordings, following middle cerebral artery occlusion in rats, we show here that: (i) these later depolarizations do indeed represent transient anoxic depolarizations; (ii) SD-like IDs, DC and haemodynamic parameters are similar to those of SDs when blood flow remains close to base line and; (iii) when blood flow decreases, the hyperaemia associated with SD-like IDs is largely reduced and there is an increasing proportion of cortical sites which fail to display a DC shift. These data demonstrate the coexistence of two mechanisms of IDs, and yield new information as to the flow-dependence of DC and haemodynamic correlates of SD-like IDs, the pathophysiological significance of which remains to be determined.


Subject(s)
Cerebral Cortex/blood supply , Cerebrovascular Circulation/physiology , Hemodynamics/physiology , Ischemic Attack, Transient/physiopathology , Analysis of Variance , Animals , Cerebral Cortex/physiopathology , Cortical Spreading Depression , Electric Stimulation , Male , Middle Cerebral Artery , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Reperfusion , Software , Time Factors
11.
Exp Neurol ; 164(2): 314-21, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915570

ABSTRACT

In previous studies of our group, we have reported differential alterations in opioidergic receptor subtypes densities in infarcted and periinfarcted brain tissue following middle cerebral artery occlusion (MCAO) in mice. Other studies have also described subcortical alterations consecutive to focal cortical ischemia. For a better understanding of ischemic processes in exofocal areas, we have investigated the evolution of opioidergic receptors following focal cortical ischemia through the quantification of relative binding densities, B(max) and K(d) values for the mu, delta, and kappa subtypes. Our results demonstrate that opioid receptor subtypes exhibit adaptations at distance from the ischemic core, mainly in the striatum, the thalamus, and the substantia nigra. Indeed, mu and delta B(max) values were increased in ventral thalamic nuclei, while kappa relative binding densities were transiently increased in nucleus medialis dorsalis and nucleus lateralis, pars posterior. Moreover, the B(max) of mu and delta receptors were transiently decreased at 6 h post-MCAO in ipsi- and contralateral patches and matrices of the striatum. Conversely, the mu B(max) values were increased in ipsi- and contralateral substantia nigra, pars compacta, and pars reticulata, 24 h following MCAO. In contralateral substantia nigra, pars compacta, kappa B(max) was found to be decreased at 24 h post-MCAO. These alterations could reflect neuronal dysfunction in exofocal brain structures, consecutively to the degeneration of defined neuroanatomical pathways. Our study indicates that opioidergic receptors could be used as markers of the neuronal reorganization that take place in subcortical areas following an ischemic insult of the brain cortex.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Analysis of Variance , Animals , Autoradiography , Binding Sites , Brain/cytology , Brain/pathology , Brain Ischemia/pathology , Corpus Striatum/cytology , Corpus Striatum/metabolism , Infarction, Middle Cerebral Artery , Ligands , Mice , Substantia Nigra/cytology , Substantia Nigra/metabolism , Thalamic Nuclei/cytology , Thalamic Nuclei/metabolism
12.
J Cereb Blood Flow Metab ; 20(6): 956-66, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10894179

ABSTRACT

In the brain, the expression of the pleiotropic cytokine interleukin-6 (IL-6) is enhanced in various chronic or acute central nervous system disorders. However, the significance of IL-6 production in such neuropathologic states remains controversial. The present study investigated the role of IL-6 after cerebral ischemia. First, the authors showed that focal cerebral ischemia in rats early up-regulated the expression of IL-6 mRNA, without affecting the transcription of its receptors (IL-6Ralpha and gp130). Similarly, the striatal injection of N-methyl-D-aspartate (NMDA) in rats, a paradigm of excitotoxic injury, activated the expression of IL-6 mRNA. The involvement of glutamatergic receptor activation was further investigated by incubating cortical neurons with NMDA or alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA). NMDA and ionomycin (a calcium ionophore) up-regulated IL-6 mRNA, suggesting that neurons may produce IL-6 in response to the calcium influx mediated through NMDA receptors. The potential role of IL-6 during ischemic/excitotoxic insults was then studied by testing the effect of IL-6 against apoptotic or excitotoxic challenges in cortical cultures. IL-6 did not prevent serum deprivation- or staurosporine-induced apoptotic neuronal death, or AMPA/kainate-mediated excitotoxicity. However, in both mixed and pure neuronal cultures, IL-6 dose-dependently protected neurons against NMDA toxicity. This effect was blocked by a competitive inhibitor of IL-6. Overall, the results suggest that the up-regulation of IL-6 induced by cerebral ischemia could represent an endogenous neuroprotective mechanism against NMDA receptor-mediated injury.


Subject(s)
Interleukin-6/immunology , Ischemic Attack, Transient/immunology , Neuroprotective Agents/immunology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Astrocytes/cytology , Brain Chemistry/drug effects , Brain Chemistry/immunology , Cells, Cultured , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Excitatory Amino Acid Agonists/pharmacology , Gene Expression/drug effects , Gene Expression/immunology , Infarction, Middle Cerebral Artery/immunology , Interleukin-6/genetics , Ionomycin/pharmacology , Ionophores/pharmacology , Male , N-Methylaspartate/pharmacology , Neurons/chemistry , Neurons/cytology , Neurons/immunology , Neurotoxins/pharmacology , Polymerase Chain Reaction , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, AMPA/physiology , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , Receptors, Kainic Acid/physiology , Transcription, Genetic/immunology
13.
Glia ; 31(1): 39-50, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10816605

ABSTRACT

There is considerable evidence that complement activation occurs within the CNS in inflammatory and degenerative disorders, but little is known about its involvement in the pathophysiology of cerebral ischemia. Our study sought to characterize the glial response and the expression of complement factors after permanent focal cerebral ischemia in the mouse, using semiquantitative reverse transcription-polymerase chain reaction (RT-PCR), in situ hybridization, and immunohistochemistry. mRNA expression of glial fibrillary acidic protein (GFAP) increased at day 1 and peaked 3 days after middle cerebral artery (MCA) occlusion in the perifocal area. Immunohistochemical staining for GFAP indicated that astroglia were activated the day after MCA occlusion. Microglial activation, as assessed by lectin-binding experiments, increased by 1 day after MCA occlusion in the perifocal area and peaked at 3 days postocclusion. RT-PCR experiments demonstrated an increased expression of clusterin, C1qB, and C4 mRNA in the ischemic cortex, with a peak level at 3 days after MCA occlusion. Clusterin, C1qB, and C4 mRNA were located in the perifocal area, as assessed by in situ hybridization. Reactive astrocytes within the cortex medial to the ischemic lesion were found to be strongly immunoreactive for clusterin. In addition, we observed C1q-positive macrophage-like cells within the infarcted core at 3 days postocclusion. At 7 days after the onset of ischemia, increased C4 immunostaining was restricted to perifocal neurons. We conclude that local expression of complement components may contribute to the inflammation observed in this model, thereby representing an important process in secondary injury mechanisms after focal cerebral ischemia.


Subject(s)
Brain Ischemia/physiopathology , Complement System Proteins/analysis , Glycoproteins/metabolism , Molecular Chaperones , Neuroglia/physiology , Animals , Astrocytes/physiology , Brain Ischemia/blood , Brain Ischemia/metabolism , Clusterin , Complement C1q/analysis , Complement C4/analysis , Glial Fibrillary Acidic Protein/metabolism , Male , Mice , Neuroglia/metabolism
14.
Glia ; 30(3): 271-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10756076

ABSTRACT

Erythropoietin (Epo), the major hormone controlling the hypoxia-induced increase in the number of erythrocytes, has also a functional role in the brain. However, few data exist as to the cellular source of brain-derived Epo as well as to the molecular mechanisms that control Epo expression in the central nervous system. Using patch-clamp and RT-PCR methods, we provide direct evidence that, besides astrocytes, neurons are a source of Epo in the brain. Both the astrocytic and neuronal expression of Epo mRNA are induced not only by hypoxia, but also by desferrioxamine (DFX) and cobalt chloride (CoCl(2)), two agents known to mimic the hypoxic induction of Epo in hepatoma cells. This induction is blocked by cycloheximide suggesting that de novo protein synthesis is required. Furthermore, the addition of H(2)O(2) decreases the hypoxia-induced Epo mRNA levels. These data indicate that, following hypoxia, a common oxygen sensing and signaling pathway leads to increased Epo gene expression in both nervous and hepatoma cells; this pathway would be dependent on the redox-state of the brain. Furthermore, we show that the in vivo administration of CoCl(2) and DFX to mice induces an increased Epo mRNA level in the neocortex. As Epo protects the brain against ischemia, our in vivo experiments suggest that the use of molecules such as CoCl(2) or DFX, that provoke an increased Epo gene expression in the brain, could be useful in the development of potential therapeutic strategies for the treatment of hypoxic or ischemic brain injury.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Erythropoietin/genetics , Hypoxia/metabolism , Neurons/metabolism , Oxidation-Reduction , Oxygen/metabolism , RNA, Messenger/metabolism , Animals , Cells, Cultured , Hypoxia/genetics , Male , Mice , Mice, Inbred Strains , Polymerase Chain Reaction , Time Factors
15.
Exp Neurol ; 161(1): 373-82, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10683302

ABSTRACT

In the present study, we have examined the expression of anaphylatoxin C3a and C5a receptors (C3aR and C5aR) at the mRNA and protein levels in ischemic brain tissues following permanent middle cerebral artery (MCA) occlusion in the mouse. C3aR and C5aR mRNAs were both detected by semiquantitative reverse transcription and polymerase chain reaction (RT-PCR) and the cellular distribution of each receptor was analyzed by immunohistochemistry. Significant increases in the expression of C3aR and C5aR mRNAs in the ischemic cortex were observed; the expression of both reached a peak at 2 days after MCA occlusion (4.3- and 3.4-fold increases, respectively, compared with nonoperated control cortical samples; P < 0.00625 with Bonferroni's correction, n = 3). C3aR and C5aR stainings were found constitutively on neurons and astrocytes. In ischemic tissues, we observed that C3aR and C5aR were expressed de novo on endothelial cells of blood vessels, at 6 h and 2 days after MCA occlusion, respectively. C3aR and C5aR immunostaining was increased in macrophage-like cells and reactive astrocytes 7 days postocclusion. C3a and C5a may play an important role in promoting inflammatory and/or repair processes in the ischemic brain by regulating glial cell activation and chemotaxis.


Subject(s)
Antigens, CD/genetics , Brain Chemistry/immunology , Brain Ischemia/metabolism , Membrane Proteins , Receptors, Complement/genetics , Animals , Antigens, CD/analysis , Arterial Occlusive Diseases/immunology , Arterial Occlusive Diseases/metabolism , Brain Ischemia/immunology , Complement C3a/metabolism , Complement C5a/metabolism , DNA Primers , Gene Expression/immunology , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/metabolism , Male , Mice , Mice, Inbred Strains , RNA, Messenger/analysis , Receptor, Anaphylatoxin C5a , Receptors, Complement/analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/genetics
16.
J Chem Neuroanat ; 20(3-4): 215-24, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11207420

ABSTRACT

Over recent years, activation studies that have been undertaken using brain imaging techniques, such as functional magnetic resonance imaging, positron emission tomography or near infrared spectroscopy, have greatly improved our knowledge of the functional anatomy of the brain. Nevertheless, activation studies do not directly quantify the variations of synaptic transmission (neuronal activity) but detect it indirectly either through the visualisation of changes in cerebral blood flow, oxidative or glycolytic metabolism (for positron emission tomography), or through the measurement of a global index that is dependent on both cerebral blood flow and oxidative metabolism (for functional magnetic resonance imaging and near infrared spectroscopy). Such approaches are based on the concept of a tight parallelism--termed coupling--between variations in neuronal activity, metabolism and cerebral blood flow. However, several "uncoupled" situations between these parameters have been reported over the last decade through experimental, pharmacological and pathophysiological studies. The aim of this review is to focus on these data that have to be taken into account for the interpretation of the results obtained in activation paradigms.


Subject(s)
Alzheimer Disease/diagnostic imaging , Alzheimer Disease/physiopathology , Cerebrovascular Circulation , Neurons/physiology , Tomography, Emission-Computed , Animals , Humans , Magnetic Resonance Imaging
17.
J Cereb Blood Flow Metab ; 19(12): 1345-53, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10598939

ABSTRACT

Various studies describe increased concentrations of transforming growth factor-beta (TGF-beta) in brain tissue after acute brain injury. However, the role of endogenously produced TGF-beta after brain damage to the CNS remains to be clearly established. Here, the authors examine the influence of TGF-beta produced after an episode of cerebral ischemia by injecting a soluble TGF-beta type II receptor fused with the Fc region of a human immunoglobulin (TbetaRIIs-Fc). First, this molecular construct was characterized as a selective antagonist of TGF-beta. Then, the authors tested its ability to reverse the effect of TGF-beta1 on excitotoxic cell death in murine cortical cell cultures. The addition of 1 microg/mL of TbetaRIIs-Fc to the exposure medium antagonized the neuroprotective activity of TGF-beta1 in N-methyl-D-aspartate (NMDA)-induced excitotoxic cell death. These results are consistent with the hypothesis that TGF-beta1 exerts a negative modulatory action on NMDA receptor-mediated excitotoxicity. To determine the role of TGF-beta1 produced in response to brain damage, the authors used a model of an excitotoxic lesion induced by the intrastriatal injection of 75 nmol of NMDA in the presence of 1.5 microg of TbetaRIIs-Fc. The intrastriatal injection of NMDA was demonstrated to induce an early upregulation of the expression of TGF-beta1 mRNA. Furthermore, when added to the excitotoxin, TbetaRIIs-Fc increased (by 2.2-fold, P < 0.05) the lesion size. These observations were strengthened by the fact that an intracortical injection of TbetaRIIs-Fc in rats subjected to a 30-minute reversible cerebral focal ischemia aggravated the volume of infarction. In the group injected with the TGF-beta1 antagonist, a 3.5-fold increase was measured in the infarction size (43.3 +/- 9.5 versus 152.8 +/- 46.3 mm3; P < 0.05). In conclusion, by antagonizing the influence of TGF-beta in brain tissue subjected to excitotoxic or ischemic lesion, the authors markedly exacerbated the resulting extent of necrosis. These results suggest that, in response to such insults, brain tissue responds by the synthesis of a neuroprotective cytokine, TGF-beta1, which is involved in the limitation of the extent of the injury. The pharmacologic potentiation of this endogenous defensive mechanism might represent an alternative and novel strategy for the therapy of hypoxic-ischemic cerebral injury.


Subject(s)
Ischemic Attack, Transient/physiopathology , Neurons/cytology , Neuroprotective Agents , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/physiology , Animals , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Cerebral Infarction/prevention & control , Fetus , Gene Expression Regulation/drug effects , Humans , Immunoglobulin Fc Fragments , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/prevention & control , Male , Mice , Middle Cerebral Artery , N-Methylaspartate/toxicity , Neurons/drug effects , Protein Serine-Threonine Kinases , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/physiology , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics
18.
Brain Res ; 842(1): 148-58, 1999 Sep 18.
Article in English | MEDLINE | ID: mdl-10526105

ABSTRACT

It has been previously suggested that the transient ischemic depolarizations (IDs), thought involved in the gradual expansion of ischemic injury in the first hours following middle cerebral artery occlusion (MCAo), are akin to spreading depression (SD). However, previous studies indicate that the characteristics of these events are heterogeneous (unlike those of SDs). We therefore sought to determine whether different types of IDs exist or not. Using four cortical microelectrodes, we compared the spatial and the temporal characteristics of IDs that occur following intraluminal MCAo in halothane-anesthetized rats to those of electrically induced SDs. An average 4.6+/-3.2 series of events, sequentially affecting the four electrodes, were recorded in 5 h following the induction of ischemia. The distribution of ID duration disclosed two types: short IDs (<7 min, 53% of all events) and long IDs (>7 min; 9% of all events). Most long IDs occurred within the first 30 min and as the initial electrophysiological event. Later on and often restricted to a single or reduced number of recording sites, intermittent IDs were of reduced amplitude or even replaced entirely by suppressed electrocorticographic activity (38% of all events). While the amplitude, duration and spreading characteristics were similar between short IDs and SDs provoked in the cortex of non-ischemic rats, those of long IDs were markedly different. Our results indicate that two types of IDs exist and confirm that most IDs (short ones) are similar in nature to SDs. Long IDs may represent a penumbral anoxic depolarization (AD), reversed by an improvement of perfusion, in the early stages of ischemia. Furthermore, we show that intermittent blockade of depolarization waves occurs and that its incidence increases with time. This blockade may reflect adaptive mechanisms which take place to prevent further depolarizations, the nature of which remains to be determined. The present description of electrophysiological abnormalities might have implications for anti-depolarization therapy in focal cerebral ischemia and to interpret the results of non-invasive techniques which enable the imaging of depolarized areas following stroke.


Subject(s)
Brain Ischemia/physiopathology , Animals , Blood Pressure/physiology , Brain Ischemia/pathology , Cortical Spreading Depression , Electric Stimulation , Electrodes, Implanted , Electroencephalography , Electrophysiology , Infarction, Middle Cerebral Artery/physiopathology , Male , Middle Cerebral Artery/physiology , Rats , Rats, Sprague-Dawley , Stereotaxic Techniques
19.
Brain Res ; 840(1-2): 115-24, 1999 Sep 04.
Article in English | MEDLINE | ID: mdl-10517959

ABSTRACT

Neurochemical activation of the substantia innominata (SI) in the rat, through the direct injection of the cholinergic agonist carbachol, has been reported to induce large increases in cerebral blood flow (CBF) throughout cortical and subcortical projection regions. The present study aimed to determine whether the vasomotor responses to cholinergic stimulation of the SI were, or were not, the consequence of an increase in metabolic activity. To this end, coupled measurements of CBF and cerebral glucose use (CGU) were undertaken during carbachol-elicited stimulation of the SI. Infusion of carbachol into the basal forebrain induced significant CBF increases in several ipsilateral cortical and subcortical areas including the amygdala. In contrast, CGU increased only in the ipsilateral amygdala and SI. Thus, we tested the hypothesis of a direct neurogenic, rather than metabolic, contribution of the basalocortical system. In this respect, carbachol-elicited stimulation resulted in significant increases in extracellular acetylcholine concentrations in the ipsilateral parietal cortex; systemic pretreatment with the muscarinic receptor antagonist scopolamine completely abolished the increase in cortical CBF elicited by cholinergic stimulation of the SI in the ipsilateral frontoparietal motor cortex while it failed to affect the increase observed in the ipsilateral temporal cortex. Several conclusions can be drawn from the present study. The stimulation of the SI by carbachol induces an increase in CBF that can be dissociated from changes in underlying glucose metabolism. Secondly, these induced changes in cortical CBF are paralleled by an increase in acetylcholine release. Lastly, the failure of scopolamine to block the flow response in all cortical regions would suggest that SI stimulation will evoke the release of vasodilatatory neurotransmitter(s) as well as acetylcholine itself.


Subject(s)
Carbachol/pharmacology , Cerebrovascular Circulation/physiology , Cholinergic Agonists/pharmacology , Cholinergic Fibers/physiology , Substantia Innominata/physiology , Acetylcholine/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebrovascular Circulation/drug effects , Glucose/metabolism , Male , Muscarinic Antagonists/pharmacology , Neural Pathways/physiology , Rats , Rats, Sprague-Dawley , Scopolamine/pharmacology , Substantia Innominata/drug effects
20.
FASEB J ; 13(11): 1315-24, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10428756

ABSTRACT

The tissue type plasminogen activator (t-PA) is a serine protease that is involved in neuronal plasticity and cell death induced by excitotoxins and ischemia in the brain. t-PA activity in the central nervous system is regulated through the activation of serine protease inhibitors (serpins) such as the plasminogen activator inhibitor (PAI-1), the protease nexin-1 (PN-1), and neuroserpin (NSP). Recently we demonstrated in vitro that PAI-1 produced by astrocytes mediates the neuroprotective effect of the transforming growth factor-beta1 (TGF-beta1) in NMDA-induced neuronal cell death. To investigate whether serpins may be involved in neuronal cell death after cerebral ischemia, we determined, by using semiquantitative RT-PCR and in situ hybridization, that focal cerebral ischemia in mice induced a dramatic overexpression of PAI-1 without any effect on PN-1, NSP, or t-PA. Then we showed that although the expression of PAI-1 is restricted to astrocytes, PN-1, NSP, and t-PA are expressed in both neurons and astrocytes. Moreover, by using semiquantitative RT-PCR and Western blotting, we observed that only the expression of PAI-1 was modulated by TGF-beta1 treatment via a TGF-beta-inducible element contained in the PAI-1 promoter (CAGA box). Finally, we compared the specificity of TGF-beta1 action with other members of the TGF-beta family by using luciferase reporter genes. These data show that TGF-beta and activin were able to induce the overexpression of PAI-1 in astrocytes, but that bone morphogenetic proteins, glial cell line-derived neutrophic factor, and neurturin did not. These results provide new insights into the regulation of the serpins/t-PA axis and the mechanism by which TGF-beta may be neuroprotective.


Subject(s)
Brain Ischemia/metabolism , Carrier Proteins/metabolism , Neuropeptides/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Serpins/metabolism , Tissue Plasminogen Activator/metabolism , Transforming Growth Factor beta/metabolism , Amyloid beta-Protein Precursor , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain Ischemia/genetics , Brain Ischemia/pathology , Carrier Proteins/genetics , Cell Death/genetics , Cells, Cultured , Gene Expression Regulation , Mice , Neurons/pathology , Neuropeptides/genetics , Plasminogen Activator Inhibitor 1/genetics , Protease Nexins , Receptors, Cell Surface , Serpins/genetics , Tissue Plasminogen Activator/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Neuroserpin
SELECTION OF CITATIONS
SEARCH DETAIL
...