Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Adv Healthc Mater ; 10(11): e2100059, 2021 06.
Article in English | MEDLINE | ID: mdl-33890419

ABSTRACT

Oxidative stress, which is one of the main harmful mechanisms of pathologies including ischemic stroke, contributes to both neurons and endothelial cell damages, leading to vascular lesions. Although many antioxidants are tested in preclinical studies, no treatment is currently available for stroke patients. Since cerium oxide nanoparticles (CNPs) exhibit remarkable antioxidant capacities, the objective is to develop an innovative coating to enhance CNPs biocompatibility without disrupting their antioxidant capacities or enhance their toxicity. This study reports the synthesis and characterization of functional polymers and their impact on the enzyme-like catalytic activity of CNPs. To study the toxicity and the antioxidant properties of CNPs for stroke and particularly endothelial damages, in vitro studies are conducted on a cerebral endothelial cell line (bEnd.3). Despite their internalization in bEnd.3 cells, coated CNPs are devoid of cytotoxicity. Microscopy studies report an intracellular localization of CNPs, more precisely in endosomes. All CNPs reduces glutamate-induced intracellular production of reactive oxygen species (ROS) in endothelial cells but one CNP significantly reduces both the production of mitochondrial superoxide anion and DNA oxidation. In vivo studies report a lack of toxicity in mice. This study therefore describes and identifies biocompatible CNPs with interesting antioxidant properties for ischemic stroke and related pathologies.


Subject(s)
Cerium , Nanoparticles , Animals , Antioxidants/pharmacology , Cerium/toxicity , Endothelial Cells , Humans , Mice , Polymers
2.
Article in English | MEDLINE | ID: mdl-31405852

ABSTRACT

We report the cases of a 39-year-old woman with chronic lymphocytic leukemia and a 21-year-old man with chronic granulomatous disease treated for cerebral aspergillosis. The patients required radical surgery for infection progression despite adequate isavuconazole plasma concentration or neurological complication. We thus decided to measure the brain isavuconazole concentration. These results suggest that the concentrations of isavuconazole obtained in the infected brain tissue clearly differ from those obtained in the normal brain tissue and the cerebrospinal fluid.


Subject(s)
Brain/microbiology , Granulomatous Disease, Chronic/drug therapy , Nitriles/therapeutic use , Pyridines/therapeutic use , Triazoles/therapeutic use , Adult , Aspergillosis/drug therapy , Aspergillosis/microbiology , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/pathogenicity , Cerebrospinal Fluid , Female , Granulomatous Disease, Chronic/microbiology , Humans , Magnetic Resonance Imaging , Young Adult
3.
Mol Neurobiol ; 55(12): 9156-9168, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29651748

ABSTRACT

Benefits from thrombolysis with recombinant tissue plasminogen activator (rt-PA) after ischemic stroke remain limited due to a narrow therapeutic window, low reperfusion rates, and increased risk of hemorrhagic transformations (HT). Experimental data showed that rt-PA enhances the post-ischemic activation of poly(ADP-ribose)polymerase (PARP) which in turn contributes to blood-brain barrier injury. The aim of the present study was to evaluate whether PJ34, a potent PARP inhibitor, improves poor reperfusion induced by delayed rt-PA administration, exerts vasculoprotective effects, and finally increases the therapeutic window of rt-PA. Stroke was induced by thrombin injection (0.75 UI in 1 µl) in the left middle cerebral artery (MCA) of male Swiss mice. Administration of rt-PA (0.9 mg kg-1) or saline was delayed for 4 h after ischemia onset. Saline or PJ34 (3 mg kg-1) was given intraperitoneally twice, just after thrombin injection and 3 h later, or once, 3 h after ischemia onset. Reperfusion was evaluated by laser Doppler, vascular inflammation by immunohistochemistry of vascular cell adhesion molecule-1 (VCAM-1) expression, and vasospasm by morphometric measurement of the MCA. Edema, cortical lesion, and sensorimotor deficit were evaluated. Treatment with PJ34 improved rt-PA-induced reperfusion and promoted vascular protection including reduction in vascular inflammation (decrease in VCAM-1 expression), HT, and MCA vasospasm. Additionally, the combined treatment significantly reduced brain edema, cortical lesion, and sensorimotor deficit. In conclusion, the combination of the PARP inhibitor PJ34 with rt-PA after cerebral ischemia may be of particular interest in order to improve thrombolysis with an extended therapeutic window.


Subject(s)
Neuroprotective Agents/therapeutic use , Phenanthrenes/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Reperfusion , Stroke/drug therapy , Thrombosis/drug therapy , Animals , Edema/complications , Edema/drug therapy , Edema/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hemorrhage/complications , Hemorrhage/drug therapy , Hemorrhage/pathology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Inflammation/pathology , Male , Mice , Neuroprotective Agents/pharmacology , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proteolysis/drug effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Stroke/complications , Stroke/pathology , Thrombosis/complications , Thrombosis/pathology , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/therapeutic use , Treatment Outcome , Vasospasm, Intracranial/complications , Vasospasm, Intracranial/drug therapy , Vasospasm, Intracranial/pathology
4.
PLoS One ; 12(9): e0184811, 2017.
Article in English | MEDLINE | ID: mdl-28910378

ABSTRACT

Traumatic brain injury (TBI) results in white matter injury (WMI) that is associated with neurological deficits. Neuroinflammation originating from microglial activation may participate in WMI and associated disorders. To date, there is little information on the time courses of these events after mild TBI. Therefore we investigated (i) neuroinflammation, (ii) WMI and (iii) behavioral disorders between 6 hours and 3 months after mild TBI. For that purpose, we used experimental mild TBI in mice induced by a controlled cortical impact. (i) For neuroinflammation, IL-1b protein as well as microglial phenotypes, by gene expression for 12 microglial activation markers on isolated CD11b+ cells from brains, were studied after TBI. IL-1b protein was increased at 6 hours and 1 day. TBI induced a mixed population of microglial phenotypes with both pro-inflammatory, anti-inflammatory and immunomodulatory markers from 6 hours to 3 days post-injury. At 7 days, microglial activation was completely resolved. (ii) Three myelin proteins were assessed after TBI on ipsi- and contralateral corpus callosum, as this structure is enriched in white matter. TBI led to an increase in 2',3'-cyclic-nucleotide 3'-phosphodiesterase, a marker of immature and mature oligodendrocyte, at 2 days post-injury; a bilateral demyelination, evaluated by myelin basic protein, from 7 days to 3 months post-injury; and an increase in myelin oligodendrocyte glycoprotein at 6 hours and 3 days post-injury. Transmission electron microscopy study revealed various myelin sheath abnormalities within the corpus callosum at 3 months post-TBI. (iii) TBI led to sensorimotor deficits at 3 days post-TBI, and late cognitive flexibility disorder evidenced by the reversal learning task of the Barnes maze 3 months after injury. These data give an overall invaluable overview of time course of neuroinflammation that could be involved in demyelination and late cognitive disorder over a time-scale of 3 months in a model of mild TBI. This model could help to validate a pharmacological strategy to prevent post-traumatic WMI and behavioral disorders following mild TBI.


Subject(s)
Brain Concussion/immunology , Cognition Disorders/etiology , Microglia/immunology , Myelin Sheath/pathology , White Matter/pathology , Animals , Biomarkers/metabolism , Brain Concussion/complications , Brain Concussion/metabolism , Disease Models, Animal , Interleukin-1beta/metabolism , Maze Learning , Mice , Microscopy, Electron, Transmission , Myelin Sheath/metabolism
5.
Stroke ; 47(12): 3048-3052, 2016 12.
Article in English | MEDLINE | ID: mdl-27834752

ABSTRACT

BACKGROUND AND PURPOSE: We previously showed that the selective neuronal nitric oxide synthase inhibitor 7-nitroindazole (7-NI) increases cerebral microcirculation in a juvenile ischemic rat model. We address the roles of cyclooxygenase (COX)-elaborated prostaglandins in collateral recruitment and blood supply. METHODS: Fourteen-day-old rats were subjected to ischemia-reperfusion and treated with either PBS or 7-NI (25 mg/kg) at the reperfusion onset. Six-keto-prostaglandin F1α was measured using ELISA. COX-1 and COX-2 and prostaglandin terminal synthesizing enzymes were evaluated using reverse-transcriptase polymerase chain reaction and immunofluorescence. Microvascular blood flow indexes (artery diameter and capillaries number) were measured using sidestream dark-field videomicroscopy in PBS- and 7-NI-treated ischemic rats in the absence or presence of the COX-2 inhibitor NS-398 (5 mg/kg). Cell death was measured with the TUNEL (terminal transferase dUTP nick end labeling) assay and cleaved-caspase-3 immunostaining. RESULTS: Six-keto-prostaglandin F1α and COX-2, associated with a prostaglandin E synthase, were significantly increased in PBS- and 7-NI-treated animals 15 minutes and 1 hour after ischemia-reperfusion, respectively. In contrast and as compared with PBS, 7-NI significantly decreased prostacyclin synthase and cytosolic prostaglandins E synthase mRNA. Selective COX-2 inhibition significantly decreased blood flow indexes and significantly reversed the effects of 7-NI, including the number of TUNEL+- and cleaved-caspase-3+-nuclei. CONCLUSIONS: These results show that the juvenile rat brains mostly respond to ischemia by a COX-2-dependent prostaglandins production and suggest that the transcriptional responses observed under 7-NI facilitate and reorient COX-2-dependent prostaglandins production.


Subject(s)
6-Ketoprostaglandin F1 alpha/metabolism , Cerebrovascular Circulation , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Microcirculation , Prostaglandin-E Synthases/metabolism , Reperfusion Injury/metabolism , Animals , Disease Models, Animal , Rats
6.
PLoS One ; 9(10): e110776, 2014.
Article in English | MEDLINE | ID: mdl-25329809

ABSTRACT

BACKGROUND: Neuro- and vasoprotective effects of poly(ADP-ribose)polymerase (PARP) inhibition have been largely documented in models of cerebral ischemia, particularly with the potent PARP inhibitor PJ34. Furthermore, after ischemic stroke, physicians are faced with incomplete tissue reperfusion and reocclusion, in which platelet activation/aggregation plays a key role. Data suggest that certain PARP inhibitors could act as antiplatelet agents. In that context, the present in vitro study investigated on human blood the potential antiplatelet effect of PJ34 and two structurally different PARP inhibitors, DPQ and INO-1001. METHODS AND RESULTS: ADP concentrations were chosen to induce a biphasic aggregation curve resulting from the successive activation of both its receptors P2Y(1) and P2Y(12). In these experimental conditions, PJ34 inhibited the second phase of aggregation; this effect was reduced by incremental ADP concentrations. In addition, in line with a P2Y(12) pathway inhibitory effect, PJ34 inhibited the dephosphorylation of the vasodilator stimulated phosphoprotein (VASP) in a concentration-dependent manner. Besides, PJ34 had no effect on platelet aggregation induced by collagen or PAR1 activating peptide, used at concentrations inducing a strong activation independent on secreted ADP. By contrast, DPQ and INO-1001 were devoid of any effect whatever the platelet agonist used. CONCLUSIONS: We showed that, in addition to its already demonstrated beneficial effects in in vivo models of cerebral ischemia, the potent PARP inhibitor PJ34 exerts in vitro an antiplatelet effect. Moreover, this is the first study to report that PJ34 could act via a competitive P2Y(12) antagonism. Thus, this antiplatelet effect could improve post-stroke reperfusion and/or prevent reocclusion, which reinforces the interest of this drug for stroke treatment.


Subject(s)
Blood Platelets/metabolism , Phenanthrenes/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Poly(ADP-ribose) Polymerase Inhibitors , Receptors, Purinergic P2Y12/metabolism , Adenosine Diphosphate/metabolism , Female , Humans , Male , Stroke/drug therapy , Stroke/metabolism
7.
PLoS One ; 9(2): e87367, 2014.
Article in English | MEDLINE | ID: mdl-24586272

ABSTRACT

Poly(ADP-ribose)polymerase and sirtuin 1 are both NAD(+)-dependent enzymes. In vitro oxidative stress activates poly(ADP-ribose)polymerase, decreases NAD(+) level, sirtuin 1 activity and finally leads to cell death. Poly(ADP-ribose)polymerase hyperactivation contributes to cell death. In addition, poly(ADP-ribose)polymerase inhibition restores NAD(+) level and sirtuin 1 activity in vitro. In vitro sirtuin 1 induction protects neurons from cell loss induced by oxidative stress. In this context, the role of sirtuin 1 and its involvement in beneficial effects of poly(ADP-ribose)polymerase inhibition were evaluated in vivo in a model of cerebral oxidative stress induced by intrastriatal infusion of malonate in rat. Malonate promoted a NAD(+) decrease that was not prevented by 3-aminobenzamide, a poly(ADP-ribose)polymerase inhibitor, at 4 and 24 hours. However, 3-aminobenzamide increased nuclear SIRT1 activity/expression ratio after oxidative stress. Malonate induced a neurological deficit associated with a striatal lesion. Both were reduced by 3-aminobenzamide and SRT1720, a sirtuin 1 activator, showing beneficial effects of poly(ADP-ribose)polymerase inhibition and sirtuin 1 activation on oxidative stress consequences. EX527, a sirtuin 1 inhibitor, given alone, modified neither the score nor the lesion, suggesting that endogenous sirtuin 1 was not activated during cerebral oxidative stress. However, its association with 3-aminobenzamide suppressed the neurological improvement and the lesion reduction induced by 3-aminobenzamide. The association of 3-aminobenzamide with SRT1720, the sirtuin 1 activator, did not lead to a better protection than 3-aminobenzamide alone. The present data represent the first demonstration that the sirtuin 1 activator SRT1720 is neuroprotective during in vivo cerebral oxidative stress. Furthermore sirtuin 1 activation is involved in the beneficial effects of poly(ADP-ribose)polymerase inhibition after in vivo cerebral oxidative stress.


Subject(s)
Brain/physiopathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/physiology , Poly(ADP-ribose) Polymerase Inhibitors , Sirtuin 1/metabolism , Analysis of Variance , Animals , Benzamides/pharmacology , Blotting, Western , Carbazoles/pharmacology , Immunoblotting , Male , Malonates/administration & dosage , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Sirtuin 1/antagonists & inhibitors , Statistics, Nonparametric
8.
Exp Neurol ; 248: 416-28, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23876515

ABSTRACT

Recombinant tissue plasminogen activator (rt-PA) is the only pharmacological treatment approved for thrombolysis in patients suffering from ischemic stroke, but its administration aggravates the risk of hemorrhagic transformations. Experimental data demonstrated that rt-PA increases the activity of poly(ADP-ribose)polymerase (PARP). The aim of the present study was to investigate whether PJ34, a potent (PARP) inhibitor, protects the blood-brain barrier components from rt-PA toxicity. In our mouse model of cerebral ischemia, administration of rt-PA (10 mg/kg, i.v.) 6h after ischemia aggravated the post-ischemic degradation of ZO-1, claudin-5 and VE-cadherin, increased the hemorrhagic transformations (assessed by brain hemoglobin content and magnetic resonance imaging). Furthermore, rt-PA also aggravated ischemia-induced functional deficits. Combining PJ34 with rt-PA preserved the expression of ZO-1, claudin-5 and VE-cadherin, reduced the hemorrhagic transformations and improved the sensorimotor performances. In vitro studies also demonstrated that PJ34 crosses the blood-brain barrier and may thus exert its protective effect by acting on endothelial and/or parenchymal cells. Thus, co-treatment with a PARP inhibitor seems to be a promising strategy to reduce rt-PA-induced vascular toxicity after stroke.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Phenanthrenes/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Stroke/drug therapy , Tissue Plasminogen Activator/adverse effects , Animals , Blood-Brain Barrier/pathology , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain Ischemia/pathology , Disease Models, Animal , Mice , Phenanthrenes/pharmacology , Stroke/pathology , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/therapeutic use
9.
BMC Res Notes ; 6: 282, 2013 Jul 21.
Article in English | MEDLINE | ID: mdl-23870165

ABSTRACT

BACKGROUND: Long term oxygen therapy improves survival in hypoxemic patients with chronic obstructive pulmonary disease (COPD). Because pressure support ventilation with a home care ventilator is largely unsupervised, there is considerable risk of leakage occurring, which could affect delivered FiO2. We have therefore conducted a bench study in order to measure the effect of different levels of O2 supply and degrees of leakage on delivered FiO2. Ventilator tested: Legendair® (Airox™, Pau, France). Thirty-six measures were performed in each four ventilators with zero, 5 and 10 l.min-1 leakage and 1,2,4 and 8 l O2 flow. FINDINGS: FiO2 decreased significantly with 5 l.min-1 leakage for all O2 flow rates, and with 10 l.min-1 at 4 and 8 l.min-1 O2. CONCLUSION: During application of NIV on home ventilators, leakage can dramatically decrease inspired FiO2 making it less effective. It is important to know the FiO2 dispensed when NIV is used for COPD at home. We would encourage industry to develop methods for FiO2 regulation Chronic use of NIV for COPD with controlled FiO2 or SpO2 requires further studys.


Subject(s)
Oxygen Inhalation Therapy , Pulmonary Disease, Chronic Obstructive/therapy , Home Care Services , Humans , Pulmonary Disease, Chronic Obstructive/physiopathology
10.
Mil Med ; 178(4): e498-502, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23707839

ABSTRACT

Proctologic emergency are very common and are a true challenge for a general practitioner (GP) in a sea-based environment. Performing simple surgical procedures could be essential for the management of these patients. Thrombosed external hemorrhoids are very painful and necessitate the extraction of the blood clot under local anesthesia. The perianal abscess and the pilonidal abscess are also painful entities and represent a significant septic risk. The surgical management of the latter two is simple but requires general anesthesia. Using ketamine and midazolam with these procedures offers a very high level of anesthetic safety. This short article describes the mentioned procedures that are richly illustrated.


Subject(s)
Anesthesia/methods , Anus Diseases/surgery , Colorectal Surgery/statistics & numerical data , Digestive System Surgical Procedures/methods , Emergencies , Military Personnel , Acute Disease , Anus Diseases/epidemiology , Digestive System Surgical Procedures/statistics & numerical data , France/epidemiology , Humans , Incidence
11.
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
12.
Bull Acad Natl Med ; 196(4-5): 893-907, 2012.
Article in French | MEDLINE | ID: mdl-23550451

ABSTRACT

Recent conflicts have led the French Army Health Service to specify the setting condition for the survival of combat casualties in overseas operations. The majority of them are victims of explosion injuries, and an early and effective control of bleeding is the primary means of improving survival. A procedure called "Combat Rescue" is taught. This chronological procedure favours external haemostasis and led to specific equipment, in particular a tourniquet and a haemostatic bandage of high efficiency. It is applied in recent years on the Afghan out-of-hospital theatre. A very front medical presence, which is systematic during evacuations, is a feature of the French Army Health Service operations support.


Subject(s)
Afghan Campaign 2001- , Military Personnel , Warfare , Wounds and Injuries/mortality , Wounds and Injuries/therapy , Critical Care/methods , Critical Care/organization & administration , Emergency Treatment/instrumentation , Emergency Treatment/methods , Emergency Treatment/standards , France , Hospitals, Military , Humans , Military Personnel/statistics & numerical data , Outpatients/statistics & numerical data
13.
Crit Care Med ; 39(10): 2300-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21666443

ABSTRACT

OBJECTIVES: Traumatic brain injury causes deleterious brain edema, leading to high mortality and morbidity. Brain edema exacerbates neurologic deficits and may be attributable to the breakdown of endothelial cell junction protein, leukocyte infiltration, and matrix metalloproteinase activation. These all contribute to loss of blood-brain barrier integrity. The pleiotropic effects of statins, hydroxymethylglutaryl-coenzyme A reductase inhibitors, may inhibit posttraumatic brain edema. We therefore investigated the effect of acute simvastatin on neurologic deficits, cerebral edema, and its origins. DESIGN: Randomized laboratory animal study. SETTINGS: University-affiliated research laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: Rats were subjected to lateral fluid percussion traumatic brain injury. Our preliminary dose-effect study indicated that 37.5 mg/kg simvastatin, administered orally 1 hr and 6 hrs after traumatic brain injury, has the greatest anti-edematous effect. This dose was used to study its effects on brain edema and on its mechanisms. MEASUREMENTS AND MAIN RESULTS: We first assessed the effects of simvastatin 24 hrs after traumatic brain injury on brain edema, brain claudin-5 expression, and the vascular endothelial-cadherin (pTyr731)/total vascular endothelial-cadherin ratio, matrix metalloproteinase-9 activity, intercellular adhesion molecule-1 expression, and polymorphonuclear neutrophil infiltration. We also evaluated blood-brain barrier permeability by measuring Evans blue and fluorescein sodium salt extravasation into the cerebral parenchyma. We then investigated whether simvastatin reduces neurologic deficits, edema, and blood-brain barrier permeability earlier than 24 hrs; these effects were evaluated 6 hrs after traumatic brain injury. The anti-edematous effect of simvastatin 24 hrs after traumatic brain injury was associated with increased claudin-5 and decreased intercellular adhesion molecule-1, polymorphonuclear neutrophil infiltration, and blood-brain barrier permeability, with no effect on matrix metalloproteinase-9 activity or vascular endothelial-cadherin phosphorylation. Earlier, 6-hrs after traumatic brain injury, simvastatin reduced neurologic deficits, cerebral edema, and blood-brain barrier permeability. CONCLUSIONS: Simvastatin could be a new therapy for reducing posttraumatic edema by preventing damage to tight junctions and neutrophil infiltration into the parenchyma, thus preserving blood-brain barrier integrity.


Subject(s)
Brain Edema/drug therapy , Brain Injuries/complications , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Animals , Antigens, CD/biosynthesis , Blood-Brain Barrier/metabolism , Brain Edema/etiology , Brain Edema/pathology , Cadherins/biosynthesis , Claudin-5 , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/biosynthesis , Male , Matrix Metalloproteinase 9/biosynthesis , Membrane Proteins/biosynthesis , Neutrophils/drug effects , Neutrophils/pathology , Rats , Rats, Sprague-Dawley
14.
J Neurotrauma ; 27(6): 1069-79, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20380552

ABSTRACT

Recent evidence supports a crucial role for matrix metalloproteinase-9 (MMP-9) in blood-brain barrier (BBB) disruption and vasogenic edema formation after traumatic brain injury (TBI). Although the exact causes of MMP-9 upregulation after TBI are not fully understood, several arguments suggest a contribution of the enzyme poly(ADP-ribose)polymerase (PARP) in the neuroinflammatory response leading to MMP-9 activation. The objectives of this study were to evaluate the effect of PARP inhibition by 3-aminobenzamide (3-AB) (1) on MMP-9 upregulation and BBB integrity, (2) on edema formation as assessed by magnetic resonance imaging (MRI), (3) on neuron survival as assessed by (1)H magnetic resonance spectroscopy ((1)H-MRS), and (4) on neurological deficits at the acute phase of TBI. Western blots and zymograms showed blunting of MMP-9 upregulation 6 h after TBI. BBB permeability was decreased at the same time point in 3-AB-treated rats compared to vehicle-treated rats. Cerebral MRI showed less "free" water in 3-AB-treated than in vehicle-treated rats 6 h after TBI. MRI findings 24 h after TBI indicated predominant cytotoxic edema, and at this time point no significant differences were found between 3-AB- and vehicle-treated rats with regard to MMP-9 upregulation, BBB permeability, or MRI changes. At both 6 and 24 h, neurological function was better in the 3-AB-treated than in the vehicle-treated rats. These data suggest that PARP inhibition by 3-AB protected the BBB against hyperpermeability induced by MMP-9 upregulation, thereby decreasing vasogenic edema formation 6 h after TBI. Furthermore, our data confirm the neuroprotective effect of 3-AB at the very acute phase of TBI.


Subject(s)
Benzamides/pharmacology , Blood-Brain Barrier/metabolism , Brain Injuries/metabolism , Brain/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Blotting, Western , Brain/drug effects , Brain/pathology , Brain Edema/metabolism , Brain Edema/pathology , Brain Injuries/pathology , Enzyme Inhibitors/pharmacology , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Matrix Metalloproteinase 9/metabolism , Permeability , Poly(ADP-ribose) Polymerase Inhibitors , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric
15.
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
16.
Crit Care ; 13(3): R76, 2009.
Article in English | MEDLINE | ID: mdl-19454040

ABSTRACT

INTRODUCTION: Serum B-type natriuretic peptide (BNP) is frequently elevated after subarachnoid hemorrhage (SAH), but whether this high BNP level is related to transient elevation of left ventricular filling pressure (LVFP) is unknown. However, in patients with preexistent cardiac pathologies, it is impossible to differentiate between BNP elevation caused by chronic cardiac abnormalities and BNP related to acute neurocardiac injury. METHODS: All adult patients with SAH admitted to our intensive care unit were eligible. Patients were excluded for the following reasons: admission >48 hours after aneurysm rupture, pre-existing hypertension, or cardiac disease. Levels of BNP and cardiac troponin Ic were measured daily for 7 days. Echocardiography was performed by a blinded cardiologist on days 1, 2, and 7. Doppler signals from the mitral inflow, tissue Doppler, and the color M-mode-derived flow propagation velocity (FPV) were obtained to assess echo-estimated LVFP. RESULTS: During a 3-year period, sixty-six consecutive patients with SAH were admitted. Thirty one patients were studied. The BNP level was >100 ng/L in 25 patients (80%) during the first 3 days, with a peak on day 2 (median, 126 ng/L) followed by a gradual decrease (median variation days 1 to 7, 70%). All patients had an ejection fraction >50%. Early transmitral velocity/tissue Doppler mitral annular early diastolic velocity was low: 5.4 (+/- 1.5) on day 1, 5.8 (+/- 1.2) on day 2, and 5.1 (+/- 0.9) on day 7. Early transmitral velocity/FPV was also low: 1.27 (+/- 0.4), 1.25 (+/- 0.3), and 1.1 (+/- 0.2) on days 1, 2, and 7, respectively. Cardiac troponin Ic levels ranged from 0 to 3.67 microg/L and were correlated with BNP (r = 0.63, P < 0.01). CONCLUSIONS: BNP rises gradually over two days and return to normal within a week after SAH. Its release is associated with myocardial necrosis, but is unrelated to elevated LVFP assessed by echocardiography.


Subject(s)
Natriuretic Peptide, Brain/blood , Subarachnoid Hemorrhage/blood , Ventricular Dysfunction, Left/blood , Acute Disease , Adult , Cardiomyopathies/blood , Cardiomyopathies/pathology , Echocardiography, Doppler , Female , Hemodynamics , Humans , Male , Necrosis/blood , Prospective Studies , Troponin I/blood , Ventricular Dysfunction, Left/diagnostic imaging
17.
Eur J Pharmacol ; 588(1): 52-7, 2008 Jun 24.
Article in English | MEDLINE | ID: mdl-18468597

ABSTRACT

Hemorrhagic transformation is an aggravating event that occurs in 15 to 43% of patients suffering from ischemic stroke. This phenomenon due to blood-brain barrier breakdown appears to be mediated in part by matrix metalloproteinases (MMPs) among which MMP-2 and MMP-9 could be particularly involved. Recent experimental studies demonstrated that post-ischemic MMP-9 overexpression is regulated by poly(ADP-ribose)polymerase (PARP). In this context, our study aimed to evaluate the effect of PJ34 (N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-2-(N,N-dimethylamino)acetamide), a potent PARP inhibitor, on MMP-2 and MMP-9 levels and on hemorrhagic transformations in a model of permanent focal cerebral ischemia in mice. PJ34 (6.25-12.5 mg/kg, i.p.) was given at the time of ischemia onset and 4 h later. Hemorrhagic transformations, divided into microscopic and macroscopic hemorrhages, were counted 48 h after ischemia on 12 coronal brain slices. Microscopic and macroscopic hemorrhages were respectively reduced by 38% and 69% with 6.25 mg/kg PJ34. The anti-hemorrhagic effect of PJ34 was associated with a 57% decrease in MMP-9 overexpression assessed by gelatin zymography. No increase in MMP-2 activity was observed after ischemia in our model. The vascular protection achieved by PJ34 was associated with a reduction in the motor deficit (P<0.05) and in infarct volume (-31%, P<0.01). In conclusion, our study demonstrates for the first time that PJ34 reduces hemorrhagic transformations after cerebral ischemia. Thus this PARP inhibitor exhibits both anti-hemorrhagic and neuroprotective effects that may be of valuable interest for the treatment of stroke.


Subject(s)
Brain Ischemia/pathology , Cerebral Hemorrhage/prevention & control , Enzyme Inhibitors/pharmacology , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Brain Ischemia/complications , Brain Ischemia/psychology , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/pathology , Cerebral Infarction/pathology , Male , Metalloproteases/metabolism , Mice , Motor Activity/drug effects , Psychomotor Performance/drug effects
18.
Ann Pharmacother ; 42(4): 584-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18420530

ABSTRACT

OBJECTIVE: To report a case of hyperventilation caused by topiramate therapy and propose a pathophysiologic mechanism for this disorder. CASE SUMMARY: A 52-year-old woman with refractory seizure disorder was admitted to the burn care unit with burns over 10% of her body. Her seizure medications, unchanged and well tolerated for several months, included carbamazepine 1200 mg, lamotrigine 500 mg, phenobarbital 80 mg, and topiramate 150 mg per day. During hospitalization, despite a relatively normal arterial pH, the woman developed persistent hyperventilation, with respiratory rates up to 50 breaths/min. Alkalinization did not reduce the hyperventilation. Thoracic contrast-enhanced computed tomographic scan ruled out pulmonary embolism and persistent pneumonia. Salicylate and biguanide screening were negative; results of repeated thyroid and liver function tests were normal. Cerebral magnetic resonance imaging excluded a cerebral pathology. After cerebrospinal fluid (CSF) analysis showed acidosis (pH 7.14), topiramate was withdrawn and the patient's general condition rapidly improved. Forty-eight hours later, the CSF pH had increased to 7.26. The woman was discharged from the burn care unit on the 42nd hospital day. DISCUSSION: Hyperchloremic normal anion gap metabolic acidosis, which can lead to hyperventilation, has been reported as an adverse effect of topiramate treatment. However, our patient had respiratory alkalosis. Concurrent etiologies of peripheral hyperventilation were excluded, leaving central neurogenic hyperventilation as the remaining etiology. Such central neurogenic hyperventilation associated with topiramate has previously been reported in intensive care. Our case report demonstrates CSF acidosis. Withdrawing topiramate reduced both CSF acidosis and hyperventilation. The mechanism of topiramate-induced CSF acidosis remains unclear. According to the Naranjo probability scale, the relationship of hyperventilation to administration of topiramate in our patient was probable. CONCLUSIONS: Normal doses of topiramate may provoke central neurogenic hyperventilation, as a result of CSF acidosis. The acid-base status of critically ill patients receiving topiramate should be monitored carefully.


Subject(s)
Acidosis/cerebrospinal fluid , Acidosis/chemically induced , Anticonvulsants/adverse effects , Fructose/analogs & derivatives , Hyperventilation/chemically induced , Drug Therapy, Combination , Female , Fructose/adverse effects , Fructose/therapeutic use , Humans , Middle Aged , Seizures/drug therapy , Topiramate
19.
Mil Med ; 172(7): 708-12, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17691682

ABSTRACT

The French army is often engaged in stability and support operations in Africa, and its military health service has gained much experience. The goal of this article is to present our military medical management strategies during the two main phases of military action. These situations most often begin with an initial combat phase, with combat casualty care. This consists of first aid, i.e., treatment of bleeding points, followed by battlefield forward medical care, damage control surgery, and resuscitation in forward surgical units. The quieter second phase of peacekeeping operations is dominated by the management of tropical diseases and their prevention, essential for the preservation of the military strength.


Subject(s)
Military Medicine , Military Personnel , Travel , Tropical Climate , Tropical Medicine , Warfare , Wounds and Injuries/surgery , Africa , France , Humans , Risk Factors , Triage
20.
Neurosci Lett ; 424(2): 106-10, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17716817

ABSTRACT

The aim of this study was to investigate how the brain is affected during systemic inflammation. For this purpose, Swiss mice were challenged with a single intraperitoneal dose of lipopolysaccharide (LPS; 250microg/mouse) to mimic aspects of systemic infection. Spatial learning in Y-maze test demonstrated a differential learning profile during the training test between control and LPS-treated mice, with an alteration in the latter group. We show that systemic LPS-induced inflammation and oxidative injury as assessed by reactive oxygen species (ROS) and nitrites/nitrates (NOx) production associated with reduced glutathione (GSH) depletion, cyclooxygenase-2 (COX-2) expression, and lipid peroxidation. LPS also induced a loss in mitochondrial integrity as shown by a significant decrease in membrane potential and impairment in mitochondrial redox activity. Thus, peripheral inflammation by producing brain inflammation and oxidative injury causes mnesic deficits. It remains to determine whether such events can induce neuronal dysfunction/degeneration and, with time, lead to cholinergic deficiency, amyloid deposits and cognitive impairments as they occur in Alzheimer's disease.


Subject(s)
Cognition Disorders/etiology , Mitochondria/pathology , Mitochondrial Diseases/etiology , Systemic Inflammatory Response Syndrome/complications , Animals , Behavior, Animal , Cyclooxygenase 2 , Disease Models, Animal , Glutathione , Lipid Peroxidation , Lipopolysaccharides , Male , Maze Learning/drug effects , Maze Learning/physiology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/physiology , Nitrates , Nitrites , Reactive Oxygen Species/metabolism , Space Perception/drug effects , Space Perception/physiology , Systemic Inflammatory Response Syndrome/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL