Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Int J Pharm ; 665: 124663, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39265854

ABSTRACT

Elucidation of the role of gut microbiota in the metabolism of orally administered drugs may improve therapeutic effectiveness and contribute to the development of personalized medicine. In this study, ten different artificial gut microbiota (AGM), obtained by culturing fecal samples in a continuous fermentation system, were challenged for their metabolizing capacity on a panel of six glucocorticoids selected from either prodrugs or drugs. Data from metabolic stability assays highlighted that, while the hydrolysis-mediated conversion of prodrugs to drugs represented only a minor metabolic pathway, significant differences in the stability of parent compounds and in their conversion rates to multiple reductive metabolites were obtained for the selected drugs. In the latter case, a taxonomic composition-dependent ability to convert parent drugs to metabolites was observed. Indeed, the artificial microbial communities dominated by the genus Bacteroides showed the maximal conversion of parent glucocorticoids to several metabolites. Furthermore, the effect of drugs on AGM was also evaluated through shallow shotgun sequencing and flow cytometry-based total bacterial cell count highlighting that these drugs can affect both the taxonomic composition and growth performances of the human gut microbiota.

2.
Brain Res ; 1491: 78-87, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23122881

ABSTRACT

The implication of cyclooxygenase (COX) type 2 in post-traumatic consequences is so far controversial. In experimental models of traumatic brain injury (TBI), genetic disruption or pharmacological inhibition of COX-2 has been shown to be neuroprotective, deleterious or without effect. Therefore, the aim of our study was to investigate the effect of COX-2 inhibition against neurological deficit and brain oedema after TBI that was induced by mechanical percussion in male Swiss mice. Despite the increased level and activity of COX-2, its inhibition either with nimesulide (12 mg/kg) or meloxicam (2mg/kg) modified neither the neurological score nor the brain water content that were evaluated at 6 and 24h after injury. Interestingly, the non-selective COX inhibition with indomethacin (5mg/kg) significantly promoted neurological recovery at 6 and 24h after trauma, without improving brain oedema. In conclusion, the present study yields considerable evidence that COX-2 may not solely constitute an interesting target for the treatment of TBI consequences. Our data point to a potentially deleterious role of COX-1 in the development of neurological impairment in brain-injured mice. However, the neuroprotective mechanism of indomethacin remains to be clarified.


Subject(s)
Brain Edema/drug therapy , Brain Edema/etiology , Cyclooxygenase Inhibitors/therapeutic use , Head Injuries, Closed/complications , Head Injuries, Closed/drug therapy , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , 6-Ketoprostaglandin F1 alpha/biosynthesis , Animals , Behavior, Animal/physiology , Blotting, Western , Brain/pathology , Brain Edema/pathology , Brain Injuries/complications , Brain Injuries/drug therapy , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/therapeutic use , Exploratory Behavior/physiology , Head Injuries, Closed/enzymology , Immunoenzyme Techniques , Indomethacin/therapeutic use , Male , Mice , Nervous System Diseases/pathology , Psychomotor Performance/physiology , Recovery of Function , Substrate Specificity
3.
J Neurotrauma ; 28(10): 2135-43, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21770756

ABSTRACT

Traumatic brain injury (TBI) induces both focal and diffuse lesions that are concurrently responsible for the ensuing morbidity and mortality and for which no established treatment is available. It has been recently reported that an endogenous neuroprotector, the soluble form α of the amyloid precursor protein (sAPPα), exerts neuroprotective effects following TBI. However, the emergent post-traumatic neuroinflammatory environment compromises sAPPα production and may promote neuronal degeneration and consequent brain atrophy. Hence, the aim of this study was to examine the effects of the anti-inflammatory drug minocycline on sAPPα levels, as well as on long-term histological consequences post-TBI. The weight-drop model was used to induce TBI in mice. Minocycline or its vehicle were administered three times: at 5 min (90 mg/kg, i.p.) and at 3 and 9 h (45 mg/kg, i.p.) post-TBI. The levels of sAPPα, the extent of brain atrophy, and reactive gliosis were evaluated by ELISA, cresyl violet, and immunolabeling of GFAP and CD11b, respectively. Our results revealed a post-TBI sAPPα decrease that was significantly attenuated by minocycline. Additionally, corpus callosum and striatal atrophy, ventriculomegaly, astrogliosis, and microglial activation were observed at 3 months post-TBI. All the above consequences were significantly reduced by minocycline. In conclusion, inhibition of the acute phase of post-TBI neuroinflammation was associated with the sparing of sAPPα and the protection of brain tissue in the long-term, emphasizing the potential role of minocycline as an effective treatment for TBI.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Anti-Bacterial Agents/pharmacology , Brain Chemistry/drug effects , Brain Injuries/drug therapy , Minocycline/pharmacology , Neuroprotective Agents , Animals , Brain Injuries/pathology , CD11b Antigen/metabolism , Cerebral Ventricles/pathology , Corpus Callosum/metabolism , Corpus Callosum/pathology , Corpus Striatum/pathology , Enzyme-Linked Immunosorbent Assay , Gliosis/pathology , Immunohistochemistry , Male , Mice
4.
J Neurotrauma ; 27(5): 911-21, 2010 May.
Article in English | MEDLINE | ID: mdl-20166806

ABSTRACT

Traumatic brain injury (TBI) causes a wide spectrum of consequences, such as microglial activation, cerebral inflammation, and focal and diffuse brain injury, as well as functional impairment. In this study we aimed to investigate the effects of acute treatment with minocycline as an inhibitor of microglial activation on cerebral focal and diffuse lesions, and on the spontaneous locomotor activity following TBI. The weight-drop model was used to induce TBI in mice. Microglial activation and diffuse axonal injury (DAI) were detected by immunohistochemistry using CD11b and ss-amyloid precursor protein (ss-APP) immunolabeling, respectively. Focal injury was determined by the measurement of the brain lesion volume. Horizontal and vertical locomotor activities were measured for up to 12 weeks post-injury by an automated actimeter. Minocycline or vehicle were administered three times post-insult, at 5 min (90 mg/kg i.p.), 3 h, and 9 h post-TBI (45 mg/kg i.p.). Minocycline treatment attenuated microglial activation by 59% and reduced brain lesion volume by 58%, yet it did not affect DAI at 24 h post-TBI. More interestingly, minocycline significantly decreased TBI-induced locomotor hyperactivity at 48 h post-TBI, and its effect lasted for up to 8 weeks. Taken together, the results indicate that microglial activation appears to play an important role in the development of TBI-induced focal injury and the subsequent locomotor hyperactivity, and its short-term inhibition provides long-lasting functional recovery after TBI. These findings emphasize the fact that minocycline could be a promising new therapeutic strategy for head-injured patients.


Subject(s)
Brain Injuries/drug therapy , Gliosis/drug therapy , Head Injuries, Closed/drug therapy , Microglia/drug effects , Microglia/metabolism , Minocycline/pharmacology , Animals , Brain Injuries/pathology , Disease Models, Animal , Follow-Up Studies , Gliosis/pathology , Head Injuries, Closed/pathology , Hyperkinesis/drug therapy , Hyperkinesis/pathology , Hyperkinesis/prevention & control , Male , Mice , Microglia/pathology , Neuroprotective Agents/pharmacology , Time Factors , Treatment Outcome
5.
Brain Res ; 1291: 122-32, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19631631

ABSTRACT

One of the severe complications following traumatic brain injury (TBI) is cerebral edema and its effective treatment is of great interest to prevent further brain damage. This study investigated the effects of minocycline, known for its anti-inflammatory properties, on cerebral edema and its respective inflammatory markers by comparing different dose regimens, on oxidative stress and on neurological dysfunction following TBI. The weight drop model was used to induce TBI in mice. The brain water content was measured to evaluate cerebral edema. Inflammatory markers were detected by ELISA (IL-1beta), zymography and Western blot (MMP-9). The oxidative stress marker (glutathione levels) and neurological function were measured by Griffith technique and string test, respectively. Minocycline was administered i.p. once (5 min), twice (5 min and 3 h) or triple (5 min, 3 h and 9 h) following TBI. The first dose of minocycline only varied (45 or 90 mg/kg), whereas the following doses were all at 45 mg/kg. The single and double administrations of minocycline reduced the increase of inflammatory markers at 6 h post-TBI. Minocycline also reduced cerebral edema at this time point, only after double administration and at the high dose regimen, although with no effect on the TBI-induced oxidized glutathione increase. The anti-edematous effect of minocycline persisted up to 24 h, upon a triple administration, and accompanied by a neurological recovery. In conclusion, we reported an anti-edematous effect of minocycline after TBI in mice according to a specific treatment regimen. These findings emphasize that the beneficial effects of minocycline depend on the treatment regimen following a brain injury.


Subject(s)
Brain Edema/drug therapy , Brain Edema/etiology , Brain Injuries/complications , Cerebral Cortex/metabolism , Interleukin-1beta/metabolism , Minocycline/therapeutic use , Oxidative Stress/drug effects , Analysis of Variance , Animals , Anti-Inflammatory Agents/therapeutic use , Blotting, Western , Body Water , Brain Edema/metabolism , Brain Edema/physiopathology , Brain Injuries/drug therapy , Brain Injuries/physiopathology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Male , Matrix Metalloproteinase 9/metabolism , Mice , Neurologic Examination , Oxidative Stress/physiology
6.
BMC Mol Biol ; 9: 62, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18611280

ABSTRACT

BACKGROUND: Traumatic brain injury models are widely studied, especially through gene expression, either to further understand implied biological mechanisms or to assess the efficiency of potential therapies. A large number of biological pathways are affected in brain trauma models, whose elucidation might greatly benefit from transcriptomic studies. However the suitability of reference genes needed for quantitative RT-PCR experiments is missing for these models. RESULTS: We have compared five potential reference genes as well as total cDNA level monitored using Oligreen reagent in order to determine the best normalizing factors for quantitative RT-PCR expression studies in the early phase (0-48 h post-trauma (PT)) of a murine model of diffuse brain injury. The levels of 18S rRNA, and of transcripts of beta-actin, glyceraldehyde-3P-dehydrogenase (GAPDH), beta-microtubulin and S100beta were determined in the injured brain region of traumatized mice sacrificed at 30 min, 3 h, 6 h, 12 h, 24 h and 48 h post-trauma. The stability of the reference genes candidates and of total cDNA was evaluated by three different methods, leading to the following rankings as normalization factors, from the most suitable to the less: by using geNorm VBA applet, we obtained the following sequence: cDNA(Oligreen); GAPDH > 18S rRNA > S100beta > beta-microtubulin > beta-actin; by using NormFinder Excel Spreadsheet, we obtained the following sequence: GAPDH > cDNA(Oligreen) > S100beta > 18S rRNA > beta-actin > beta-microtubulin; by using a Confidence-Interval calculation, we obtained the following sequence: cDNA(Oligreen) > 18S rRNA; GAPDH > S100beta > beta-microtubulin > beta-actin. CONCLUSION: This work suggests that Oligreen cDNA measurements, 18S rRNA and GAPDH or a combination of them may be used to efficiently normalize qRT-PCR gene expression in mouse brain trauma injury, and that beta-actin and beta-microtubulin should be avoided. The potential of total cDNA as measured by Oligreen as a first-intention normalizing factor with a broad field of applications is highlighted. Pros and cons of the three methods of normalization factors selection are discussed. A generic time- and cost-effective procedure for normalization factor validation is proposed.


Subject(s)
Brain Injuries/genetics , Gene Expression Profiling/standards , Reverse Transcriptase Polymerase Chain Reaction/standards , Actins/genetics , Animals , DNA, Complementary/analysis , DNA, Complementary/standards , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Mice , Models, Animal , Nerve Growth Factors/genetics , RNA, Ribosomal, 18S/genetics , Reference Standards , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , Tubulin/genetics , Validation Studies as Topic
7.
J Neurosci ; 25(41): 9367-77, 2005 Oct 12.
Article in English | MEDLINE | ID: mdl-16221845

ABSTRACT

Acute brain injuries have been identified as a risk factor for developing Alzheimer's disease (AD). Because glutamate plays a pivotal role in these pathologies, we studied the influence of glutamate receptor activation on amyloid-beta (Abeta) production in primary cultures of cortical neurons. We found that sublethal NMDA receptor activation increased the production and secretion of Abeta. This effect was preceded by an increased expression of neuronal Kunitz protease inhibitory domain (KPI) containing amyloid-beta precursor protein (KPI-APP) followed by a shift from alpha-secretase to beta-secretase-mediated APP processing. This shift is a result of the inhibition of the alpha-secretase candidate tumor necrosis factor-alpha converting enzyme (TACE) when associated with neuronal KPI-APPs. This KPI-APP/TACE interaction was also present in AD brains. Thus, our findings reveal a cellular mechanism linking NMDA receptor activation to neuronal Abeta secretion. These results suggest that even mild deregulation of the glutamatergic neurotransmission may increase Abeta production and represent a causal risk factor for developing AD.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Endopeptidases/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/genetics , Animals , Aspartic Acid Endopeptidases , Cells, Cultured , Endopeptidases/genetics , Excitatory Amino Acid Agonists/pharmacology , Humans , Mice , N-Methylaspartate/pharmacology , Neurons/drug effects , Protease Inhibitors/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology
8.
J Neurosci Methods ; 144(2): 183-91, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15910976

ABSTRACT

Neuroinflammation is one of the events occurring after acute brain injuries. The aim of the present report was to characterize a rat model to study acute neuroinflammation on the histopathological, biochemical and functional outcomes. Lipopolysaccharide (LPS), known as a strong immunostimulant, was directly injected into the hippocampus. The spatiotemporal evolution of inducible NOS (iNOS) and cell death was studied from 6 h to 7 days. A perfect time course correlation was observed between iNOS immunoreactivity and iNOS activity showing an acute, expansive and transient iNOS induction in the hippocampus with a peak at 24 h. It was associated with a marked increase in NO metabolite (NO(x)) levels, and a high level of myeloperoxidase (MPO) activity. This inflammation precedes a massive cellular loss including at least neurons and astrocytes, and a drop of constitutive NOS activity, restrictive to the ipsilateral hippocampus from 48 h after LPS injection. Moreover, sensorimotor function impairment occurred from 24 h to 7 days with a maximum at 24 h post-LPS injection. Therefore, we characterized an in vivo model of acute neuroinflammation and neurodegeneration, in relation with a neurological deficit, which may be a powerful tool for mechanistic studies and for further evaluation of the potential neuroprotective agents.


Subject(s)
Disease Models, Animal , Encephalitis/physiopathology , Inflammation Mediators/pharmacology , Lipopolysaccharides/pharmacology , Nerve Degeneration/physiopathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Encephalitis/chemically induced , Encephalitis/pathology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Male , Movement Disorders/pathology , Movement Disorders/physiopathology , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Sensation Disorders/chemically induced , Sensation Disorders/pathology , Sensation Disorders/physiopathology , Stroke/pathology , Stroke/physiopathology , Time Factors , Up-Regulation/physiology
9.
Eur J Pharmacol ; 483(2-3): 259-65, 2004 Jan 12.
Article in English | MEDLINE | ID: mdl-14729115

ABSTRACT

The present study examined caspase activation and its modulation by nitric oxide (NO) in a model of oxidative stress induced by injection of malonate (3 micromol), a mitochondrial toxin, into rat striatum. Caspase-3-like enzymatic activity was maximal 6 h after malonate while NO production evaluated by its metabolites nitrites and nitrates was increased at 3 h. The neuronal NO-synthase inhibitor 7-nitroindazole reduced malonate induced-NO production by 50% at 25 mg/kg and enhanced by 32% caspase activation. This result suggests that a moderate production of NO potentiates caspase activation, an effect counterbalanced by NO itself at higher concentrations. Accordingly, complete inhibition of NO production by 7-nitroindazole at 50 mg/kg did not modify malonate-induced caspase activity. Thus NO production by the neuronal isoform of NO-synthase is not the major event leading to caspase activation due to malonate. However, NO seems to have pro- and anti-caspase effects that neutralize each other.


Subject(s)
Caspases/metabolism , Corpus Striatum/drug effects , Malonates/administration & dosage , Nitric Oxide/biosynthesis , Animals , Caspase Inhibitors , Corpus Striatum/metabolism , Indazoles/pharmacology , Male , Nitric Oxide/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
10.
Brain Res ; 989(1): 58-66, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-14519512

ABSTRACT

Traumatic brain injury produces nitric oxide and reactive oxygen species. Peroxynitrite, resulting from the combination of nitric oxide and superoxide anions, triggers DNA strand breaks, leading to the activation of poly(ADP-ribose)polymerase-1. As excessive activation of this enzyme induces cell death, we examined the production of nitrosative stress, the activation of poly(ADP-ribose)polymerase-1, and the role of this enzyme in the outcomes of traumatic brain injury produced by fluid percussion in rats. Immunohistochemistry showed that 3-nitrotyrosine, an indicator of nitrosative stress, and poly(ADP-ribose), a marker of poly(ADP-ribose)polymerase-1 activation, were present as early as 30 min post-injury, and that persisted for 72 h. The poly(ADP-ribose)polymerase inhibitor, 3-aminobenzamide, at 10 and 30 mg/kg, significantly improved the neurological deficit, with a 60% reduction in the brain lesion volume and inhibition of poly(ADP-ribose)polymerase-1 activation. Thus, poly(ADP-ribose)polymerase-1 is involved in the neurological consequences of traumatic brain injury and may be a promising therapeutic target in clinical treatment of acute brain trauma.


Subject(s)
Brain Injuries/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction/physiology , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Brain Injuries/drug therapy , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Male , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/analysis , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
11.
Brain Res ; 987(1): 32-8, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-14499943

ABSTRACT

The present work examined whether polymorphonuclear neutrophil (PMN) infiltration contributes to cortical and striatal brain damage and oxidative stress in a model of transient focal cerebral ischemia. A 2-h occlusion of the left middle cerebral artery and ipsilateral common carotid artery was performed in rats. Administration of the neutropenic agent vinblastine (0.5 mg/kg, i.v.) resulted in a profound decrease in circulating PMNs which was associated with a 80% decrease in myeloperoxidase activity, a marker of PMN infiltration, in both the cortex and the striatum. In the cortex, vinblastine-treated animals exhibited a 44% decrease in the infarct volume and also reduced the oxidative stress (evaluated by the decrease in glutathione concentrations). By contrast, in the striatum, neutropenia modified neither the lesion size nor the oxidative stress. These results indicate that PMN contribution to postischemic injury and oxidative stress is dependent on the brain structure.


Subject(s)
Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Oxidative Stress , Reperfusion Injury/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Brain Ischemia/drug therapy , Brain Ischemia/enzymology , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Corpus Striatum/metabolism , Male , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/enzymology , Vinblastine/pharmacology
12.
J Pharmacol Exp Ther ; 306(2): 588-94, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12730357

ABSTRACT

Nitric oxide (NO) and reactive oxygen species are both implicated in neuronal death due to cerebral ischemia. BN 80933, an original compound associating an inhibitor of neuronal NO synthase with an antioxidant, has been shown to reduce functional and histological damage in rat submitted to cerebral ischemia. The aim of the present study was to confirm these results in mice and to further examine the effects of BN 80933 on inflammatory response, including blood-brain barrier (BBB) disruption, brain edema, and neutrophil infiltration after transient middle cerebral artery occlusion (MCAO). Intravenous administration of BN 80933 at 3 and 10 mg/kg 3 h after MCAO significantly reduced by 26 to 36% the infarct volume evaluated 24 and 48 h after ischemia, and improved the neurological score. Furthermore, BN 80933 at both dosages decreased by 42 to 75% the extravasation of Evans blue in brain parenchyma observed 24 h after ischemia. This reduction in BBB disruption was associated with decreased brain edema as demonstrated by the 37% reduction in brain water content induced by BN 80933 at 3 mg/kg 24 h after MCAO. Neutrophil infiltration in brain parenchyma, evaluated by the myeloperoxidase activity, was also reduced by 45 to 56% in animals treated with BN 80933 at 3 and 10 mg/kg. Together, these results extend the protective capacity of BN 80933 against brain ischemic injury and confirm that BN 80933 represents a promising treatment for stroke.


Subject(s)
Ischemic Attack, Transient/prevention & control , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase/antagonists & inhibitors , Pyrazines/therapeutic use , Thiophenes/therapeutic use , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Blood-Brain Barrier/drug effects , Brain Edema/prevention & control , Brain Infarction/prevention & control , Infarction, Middle Cerebral Artery/enzymology , Male , Mice , Nervous System Diseases/prevention & control , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type I , Peroxidase/metabolism , Pyrazines/pharmacology , Thiophenes/pharmacology
13.
Exp Neurol ; 184(2): 973-80, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14769390

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) was shown to be detrimental in cerebral ischemia but the mechanisms whereby PARP is deleterious have yet to be determined. They may include a role in neutrophil infiltration known to aggravate ischemic damage. In this context, we investigated the effect of 3-aminobenzamide (3-AB), a PARP inhibitor, on brain damage and neutrophil infiltration after transient focal cerebral ischemia in mice. Ischemia was induced in male Swiss mice, anaesthetized with chloral hydrate (400 mg/kg, i.p.), by a 15-min-occlusion of the left middle cerebral artery using an intraluminal suture. Treatments with 3-AB were first administered intraperitoneally 15 min before reperfusion and endpoints measured at 24 h. Among the range of dosages studied (20-320 mg/kg), 40 mg/kg gave the maximal neuroprotection with a 30% decrease in the infarct volume and tended to improve the neurological score evaluated by a grip test. The same dosage was, however, devoid of effect when injection was delayed 2 or 6 h after reperfusion. Myeloperoxidase (MPO) activity used as an index of neutrophil infiltration showed that infiltration peaked 48 h after reperfusion in our model. At this time point, 3-AB (40 mg/kg given 15 min before reperfusion) markedly reduced the neutrophil infiltration, as evidenced by a 72%-decrease in MPO activity, and was still neuroprotective. Our results confirm that 3-AB reduces brain damage. Moreover, for the first time, a quantitative study shows that 3-AB decreases neutrophil infiltration elicited by cerebral ischemia.


Subject(s)
Benzamides/pharmacology , Brain Infarction/drug therapy , Neuroprotective Agents/pharmacology , Neutrophil Infiltration/drug effects , Animals , Brain Infarction/etiology , Brain Infarction/pathology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ischemic Attack, Transient/complications , Male , Mice , Peroxidase/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Reperfusion , Time Factors
14.
Eur J Pharmacol ; 457(2-3): 137-46, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12464359

ABSTRACT

The role of nitric oxide (NO) in the development of post-ischemic cerebral infarction has been extensively examined, but fewer studies have investigated its role in other outcomes. In the present study, we first determined the temporal evolution of infarct volume, NO production, neurological deficit and blood-brain barrier disruption in a model of transient focal cerebral ischemia in mice. We then examined the effect of the nonselective NO-synthase inhibitor N(omega)-nitro-L-arginine-methylester (L-NAME). L-NAME given at 3 mg/kg 3 h after ischemia reduced by 20% the infarct volume and abolished the increase in brain NO production evaluated by its metabolites (nitrites/nitrates) 48 h after ischemia. L-NAME with this protocol also reduced the neurological deficit evaluated by the grip test and decreased by 65% the extravasation of Evans blue, an index of blood-brain barrier breakdown. These protective activities of L-NAME suggest that NO has multiple deleterious effects in cerebral ischemia.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Ischemia/complications , Enzyme Inhibitors/pharmacology , Infarction, Middle Cerebral Artery/prevention & control , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Animals , Blood-Brain Barrier/physiology , Brain/enzymology , Brain/metabolism , Brain Chemistry/drug effects , Brain Ischemia/enzymology , Brain Ischemia/physiopathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/etiology , Male , Mice , Nitrates/analysis , Nitric Oxide Synthase/antagonists & inhibitors , Nitrites/analysis
SELECTION OF CITATIONS
SEARCH DETAIL