Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Neuropathol Appl Neurobiol ; 47(2): 297-315, 2021 02.
Article in English | MEDLINE | ID: mdl-32898926

ABSTRACT

AIMS: Impairment of blood-brain barrier (BBB) is involved in numerous neurological diseases from developmental to aging stages. Reliable imaging of increased BBB permeability is therefore crucial for basic research and preclinical studies. Today, the analysis of extravasation of exogenous dyes is the principal method to study BBB leakage. However, these procedures are challenging to apply in pups and embryos and may appear difficult to interpret. Here we introduce a novel approach based on agonist-induced internalization of a neuronal G protein-coupled receptor widely distributed in the mammalian brain, the somatostatin receptor type 2 (SST2). METHODS: The clinically approved SST2 agonist octreotide (1 kDa), when injected intraperitoneally does not cross an intact BBB. At sites of BBB permeability, however, OCT extravasates and induces SST2 internalization from the neuronal membrane into perinuclear compartments. This allows an unambiguous localization of increased BBB permeability by classical immunohistochemical procedures using specific antibodies against the receptor. RESULTS: We first validated our approach in sensory circumventricular organs which display permissive vascular permeability. Through SST2 internalization, we next monitored BBB opening induced by magnetic resonance imaging-guided focused ultrasound in murine cerebral cortex. Finally, we proved that after intraperitoneal agonist injection in pregnant mice, SST2 receptor internalization permits analysis of BBB integrity in embryos during brain development. CONCLUSIONS: This approach provides an alternative and simple manner to assess BBB dysfunction and development in different physiological and pathological conditions.


Subject(s)
Blood-Brain Barrier/pathology , Capillary Permeability , Immunohistochemistry/methods , Receptors, Somatostatin/analysis , Receptors, Somatostatin/metabolism , Animals , Antibodies, Monoclonal , Mice , Mice, Inbred C57BL , Octreotide/metabolism , Rats , Rats, Wistar
3.
Cerebrovasc Dis ; 41(1-2): 40-9, 2016.
Article in English | MEDLINE | ID: mdl-26599266

ABSTRACT

BACKGROUND: The development of collateral circulation is proposed as an inherent compensatory mechanism to restore impaired blood perfusion after ischemia, at least in the penumbra. We have studied the dynamic macro- and microcirculation after ischemia-reperfusion in the juvenile rat brain and evaluated the impact of neuronal nitric oxide synthase (nNOS) inhibition on the collateral flow. METHODS: Fourteen-day-old (P14) rats were subjected to ischemia-reperfusion and treated with either PBS or 7-nitroindazole (7-NI, an nNOS inhibitor, 25 mg/kg). Arterial blood flow (BF) was measured using 2D-color-coded pulsed ultrasound imaging. Laser speckle contrast (LSC) imaging and sidestream dark-field videomicroscopy were used to measure cortical and microvascular BF, respectively. RESULTS: In basal conditions, 7-NI reduced BF in the internal carotids (by ∼ 25%) and cortical (by ∼ 30%) BF, as compared to PBS. During ischemia, the increased mean BF velocity in the basilar trunk after both PBS and 7-NI demonstrated the establishment of collateral support and patency. Upon re-flow, BF immediately recovered to basal values in the internal carotid arteries under both conditions. The 7-NI group showed increased collateral flow in the penumbral tissue during early re-flow compared to PBS, as shown with both LSC imaging and side-stream dark-field videomicroscopy. The proportion of perfused capillaries was significantly increased under 7-NI as compared to PBS when given before ischemia (67.0 ± 3.9 vs. 46.8 ± 8.8, p < 0.01). Perfused capillaries (63.1 ± 17.7 vs. 81.1 ± 20.7, p < 0.001) and the BF index (2.4 ± 0.6 vs. 1.3 ± 0.1, p < 0.001) significantly increased under 7-NI given at the re-flow onset. CONCLUSIONS: Collateral support in the penumbra is initiated during ischemia, and may be increased during early re-flow by neuronal NOS inhibition (given in pre- and post-treatment), which may preserve brain tissue in juvenile rats.


Subject(s)
Brain Ischemia , Brain/drug effects , Cerebrovascular Circulation/drug effects , Collateral Circulation/drug effects , Enzyme Inhibitors/pharmacology , Hemodynamics/drug effects , Indazoles/pharmacology , Microcirculation/drug effects , Nitric Oxide Synthase Type I/antagonists & inhibitors , Animals , Blood Flow Velocity/drug effects , Brain/blood supply , Cerebral Angiography , Rats , Reperfusion
5.
AJNR Am J Neuroradiol ; 34(7): E73-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22555584

ABSTRACT

SUMMARY: In this study, we compared lesion size by using VADC and VT2 at 0, 2, 5, 24, and 48 hours and histologic lesions at 48 hours in a P7 rat stroke model. The best correlation between VHISTO and VADC was at H0, and between VHISTO and VT2, at H2-H5. Early MR imaging signals allowed excluding "no-lesion" and "no-reflow" animals to help standardize this neonatal stroke model and predict lesion size.


Subject(s)
Brain Ischemia/diagnosis , Magnetic Resonance Imaging/methods , Stroke/diagnosis , Animals , Animals, Newborn , Brain Ischemia/pathology , Carotid Artery, Common/pathology , Disease Models, Animal , Echo-Planar Imaging/methods , Forecasting , Hypoxia-Ischemia, Brain/diagnosis , Hypoxia-Ischemia, Brain/pathology , Image Processing, Computer-Assisted/methods , Magnetic Resonance Angiography/methods , Middle Cerebral Artery/pathology , Prognosis , Rats , Rats, Wistar , Reperfusion/methods , Stroke/pathology , Time Factors
6.
Cell Death Dis ; 2: e203, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21881605

ABSTRACT

Brain protection of the newborn remains a challenging priority and represents a totally unmet medical need. Pharmacological inhibition of caspases appears as a promising strategy for neuroprotection. In a translational perspective, we have developed a pentapeptide-based group II caspase inhibitor, TRP601/ORPHA133563, which reaches the brain, and inhibits caspases activation, mitochondrial release of cytochrome c, and apoptosis in vivo. Single administration of TRP601 protects newborn rodent brain against excitotoxicity, hypoxia-ischemia, and perinatal arterial stroke with a 6-h therapeutic time window, and has no adverse effects on physiological parameters. Safety pharmacology investigations, and toxicology studies in rodent and canine neonates, suggest that TRP601 is a lead compound for further drug development to treat ischemic brain damage in human newborns.


Subject(s)
Caspase Inhibitors , Cysteine Proteinase Inhibitors/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Oligopeptides/therapeutic use , Quinolines/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Binding Sites , Caspases/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cytochromes c/metabolism , Disease Models, Animal , Hypoxia-Ischemia, Brain/pathology , Ischemia/pathology , Mice , Neuroprotective Agents/chemistry , Oligopeptides/chemistry , Oligopeptides/pharmacology , Quinolines/chemistry , Rats
7.
Brain Pathol ; 18(1): 1-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17924984

ABSTRACT

Inflammatory processes are a major cause of hypoxic-ischemic brain damage. The present study focuses on both the cerebral histamine system and mast cells in a model of transient focal ischemia induced by permanent left middle cerebral artery, and homolateral transient common carotid artery occlusion (50 minutes) in the P7 newborn rat. Immunohistochemical analysis revealed that ischemia induces histamine (HA) accumulation in the core of the infarct 6-12 h post-ischemia, and in the penumbra at 24-48 h, although in situ hybridization failed to detect any histidine decarboxylase gene transcripts in these regions. Immunohistochemical co-localization of HA with the MAP2 marker revealed that HA accumulates in neuronal cells before they degenerate, and is accompanied by a very significant increase in the number of mast cells at 12 h and 48 h of reperfusion. In mast cells, histamine immunoreactivity is detected at 2, 6 and 12 h after ischemia, whereas it disappears at 24 h, when a concomitant degranulation of mast cells is observed. Taken together, these data suggest that the recruitment of cerebral mast cells releasing histamine may contribute to ischemia-induced neuronal death in the immature brain.


Subject(s)
Histamine/metabolism , Hypoxia-Ischemia, Brain/metabolism , Mast Cells/metabolism , Nerve Degeneration/metabolism , Stroke/metabolism , Age Factors , Animals , Animals, Newborn , Biomarkers/analysis , Biomarkers/metabolism , Brain/blood supply , Brain/metabolism , Brain/pathology , Cell Count , Cell Death , Chemotaxis, Leukocyte , Disease Models, Animal , Histamine Release , Hypoxia-Ischemia, Brain/pathology , Infarction, Middle Cerebral Artery/pathology , Microtubule-Associated Proteins/metabolism , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Stroke/pathology , Time Factors
9.
Exp Neurol ; 199(1): 103-12, 2006 May.
Article in English | MEDLINE | ID: mdl-16697370

ABSTRACT

This study examines cell death and proliferation in the white matter after neonatal stroke. In postnatal day 7 injured rat, there was a marked reduction in myelin basic protein (MBP) immunostaining mainly corresponding to numerous pyknotic immature oligodendrocytes and TUNEL-positive astrocytes in the ipsilateral external capsule. In contrast, a substantial restoration of MBP, as indicated by the MBP ratio of left-to-right, occurred in the cingulum at 48 (1.27 +/- 0.12) and 72 (1.30 +/- 0.18, P < 0.05) h of recovery as compared to age-matched controls (1.03 +/- 0.14). Ki-67 immunostaining revealed a first peak of newly generated cells in the dorsolateral hippocampal subventricular zone and cingulum at 72 h after reperfusion. Double immunofluorescence revealed that most of the Ki-67-positive cells were astrocytes at 48 h and NG2 pre-oligodendrocytes at 72 h of recovery. Microglia infiltration occurs over several days in the cingulum, and a huge quantity of macrophages reached the subcortical white matter where they engulfed immature oligodendrocytes. The overall results suggest that the persistent activation of microglia involves a chronic component of immunoinflammation, which overwhelms repair processes and contributes to cystic growth in the developing brain.


Subject(s)
Brain/physiology , Ischemia/pathology , Neuroglia/physiology , Animals , Animals, Newborn , Antigens/metabolism , Brain/growth & development , Brain Infarction/etiology , Brain Infarction/pathology , Cell Count/methods , Female , Fluorescent Antibody Technique/methods , Functional Laterality/physiology , Glial Fibrillary Acidic Protein/metabolism , In Situ Nick-End Labeling/methods , Ischemia/complications , Ki-67 Antigen/metabolism , Male , Myelin Basic Protein/metabolism , Proteoglycans/metabolism , Rats , Statistics, Nonparametric , Time Factors
10.
Neuroscience ; 128(1): 27-38, 2004.
Article in English | MEDLINE | ID: mdl-15450351

ABSTRACT

Aquaporin 9 (AQP9) is a recently cloned water channel that is permeable to monocarboxylate, glycerol and urea. In rat, AQP9 has been found in testis and liver as well as in brain where its expression has been initially shown in glial cells in forebrain. However, the expression of AQP9 has not been investigated in the brainstem. The purpose of this study is to describe the distribution of AQP9-immunoreactive cells throughout the adult rat brain using reverse transcriptase-polymerase chain reaction (RT-PCR), Western blot and immunohistochemistry. We performed immunolabeling on brain from animals perfused with fixative and we show that AQP9 is expressed (i) in astrocytes in the glia limitans, in the white matter and in glial cells of the cerebellum, (ii) in the endothelial cells of pial vessels, and (iii) in specific groups of neurons. The neuronal AQP9 expression was almost exclusively observed in catecholaminergic cells including the adrenergic, noradrenergic and dopaminergic groups, but not in other monoaminergic neurons such as serotonergic or histaminergic cells. A slight labeling was also observed in non-catecholaminergic neurons localized in the paraventricular nucleus of the hypothalamus. These results indicate that AQP9 has a unique brain distribution with a preferential localization in catecholaminergic nuclei known to be involved in many cerebral functions. While the presence of AQP9 in glia limitans and in endothelial cells of the pial vessels could be related to water transport through the blood-brain barrier, its expression in neuronal cells, not directly involved in the osmoregulation, suggests that brain AQP9 could also be used as a metabolite channel since lactate and glycerol can be energy substrates for neurons.


Subject(s)
Aquaporins/biosynthesis , Brain/metabolism , Catecholamines/metabolism , Ion Channels/biosynthesis , Neurons/metabolism , Animals , Blotting, Western , Endothelium, Vascular/metabolism , Immunohistochemistry , Pia Mater/blood supply , Rats , Reverse Transcriptase Polymerase Chain Reaction
11.
Neuropathol Appl Neurobiol ; 29(4): 350-60, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12887595

ABSTRACT

Neuronal apoptosis plays an essential role in early brain development and contributes to secondary neuronal loss after acute ischaemia. Recent studies have provided evidence that caspase-3 is an important downstream event after hypoxia-ischaemia in the immature brain, but a minor event in the adult brain. Our investigations have focused on cell populations that expressed apoptotic effectors in the enzymatic death pathway including cytochrome c, caspase-9 and caspase-3. Expression, activation and cellular localization of these proteins were studied using cleavage of fluorogenic substrate and immunohistochemistry in neonatal rat brain after unilateral focal ischaemia. Caspase-3 enzyme activity was elevated in brain homogenate between 6 and 48 h after reperfusion. This activation was preceded by that of caspase-9, between 3 and 24 h. Apoptotic cell death was finally accomplished by poly-ADP-ribose polymerase cleavage, an endogenous caspase-3 substrate. In addition, immunodetection demonstrated that cytochrome c and activated caspase-9 and caspase-3 were expressed not only in the neurones, the primarily affected cells, but also within the astrocytes, which constituted a dense network delineating the infarct. These results suggested that glial injury may promote the formation of cystic lesions such as those observed clinically in the newborn brain.


Subject(s)
Apoptosis/physiology , Astrocytes/pathology , Hypoxia-Ischemia, Brain/pathology , Mitochondria/metabolism , Neurons/pathology , Animals , Animals, Newborn , Astrocytes/enzymology , Caspase 3 , Caspase 9 , Caspases/metabolism , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Cytochrome c Group/metabolism , Cytosol/metabolism , Female , Hypoxia-Ischemia, Brain/metabolism , Male , Neurons/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Wistar
12.
Bull Acad Natl Med ; 185(3): 555-63; discussion 564-5, 2001.
Article in French | MEDLINE | ID: mdl-11501263

ABSTRACT

Poly(ADP-ribose) polymerase (PARP, EC 2.4.2.30) is known as a nuclear enzyme that is activated by DNA strand breaks to participate in DNA repair. It is also called poly(ADP-ribose) synthase (PARS) or poly(ADP-ribose) transferase (PADRT). In physiological conditions, PARP plays an important role in maintaining genomic stability. However, for several pathological situations, which include massive DNA injury (brain ischemia for example), excessive activation of PARP can deplete stores of nicotinamide adenine dinucleotide (NAD+), the PARP substrate, which, with the subsequent ATP depletion, leads to cell death. PARP activation appears to play a major role in neuronal death induced by cerebral ischemia, traumatic brain injury, Parkinson disease and other pathologies. PARP inhibitors (3-aminobenzamide and other compounds) and PARP gene deletion induced dramatic neuroprotection in experimental animals (rats, mice). Accordingly, these data suggest that PARP inhibitors could provide a novel therapeutic approach in a wide range of neurodegenerative disorders including cerebral ischemia and traumatic brain injury.


Subject(s)
Apoptosis/physiology , Brain Ischemia/enzymology , Neurons/physiology , Poly(ADP-ribose) Polymerases/physiology , Animals , Benzamides/therapeutic use , Brain Ischemia/drug therapy , DNA Damage/drug effects , DNA Damage/physiology , Disease Models, Animal , Gene Deletion , Humans , Mice , Neuroprotective Agents/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/chemistry , Rats
14.
Drug Discov Today ; 5(7): 303-304, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10856915
15.
J Neurochem ; 74(6): 2504-11, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10820212

ABSTRACT

Poly(ADP-ribose) synthase (PARS), an abundant nuclear protein, has been described as an important candidate for mediation of neurotoxicity by nitric oxide. However, in cerebral ischemia, excessive PARS activation may lead to energy depletion and exacerbation of neuronal damage. We examined the effect of inhibiting PARS on the (a) degree of cerebral injury, (b) process of inflammatory responses, and (c) functional outcomes in a neonatal rat model of focal ischemia. We demonstrate that administration of 3-aminobenzamide, a PARS inhibitor, leads to a significant reduction of infarct volume: 63 +/- 2 (untreated) versus 28 +/- 4 mm(3) (treated). The neuroprotective effects currently observed 48 h postischemia hold up at 7 and 17 days of survival time and attenuate neurological dysfunction. Inhibition of PARS activity, demonstrated by a reduction in poly(ADP-ribose) polymer formation, also reduces neutrophil recruitment and levels of nitrotyrosine, an indicator of peroxynitrite generation. Taken together, our results demonstrate that PARS inhibition reduces ischemic damage and local inflammation associated with reperfusion and may be of interest for the treatment of neonatal stroke.


Subject(s)
Benzamides/pharmacology , Brain Ischemia/metabolism , Neuroprotective Agents/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Animals, Newborn , Brain Ischemia/drug therapy , Brain Ischemia/immunology , Cell Death/drug effects , Cerebral Infarction/drug therapy , Cerebral Infarction/immunology , Cerebral Infarction/metabolism , Encephalitis/drug therapy , Encephalitis/immunology , Encephalitis/metabolism , Female , Male , Motor Activity , Neurologic Examination , Neutrophils/immunology , Nitrates/metabolism , Polymers/metabolism , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Signal Transduction/drug effects , Stroke/drug therapy , Stroke/metabolism , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/analysis , Tyrosine/metabolism
16.
Brain Res Dev Brain Res ; 116(2): 133-40, 1999 Sep 06.
Article in English | MEDLINE | ID: mdl-10521557

ABSTRACT

Cerebral ischemia in adult rodents leads to the production of several types of lesions in the genomic DNA, followed by the activation of the damage-response indicator Gadd45. Our purpose was to investigate the structural changes that occur in chromatin DNA and repair processes after ischemic injury in neonatal brain. Neonatal ischemia was induced by the permanent left MCA occlusion in association with 1 h occlusion of the left common carotid artery in 7-day-old Wistar pups. Oligonucleosome fragments that are recognized as the characteristic DNA ladder was observed in a delayed fashion. Double-strand breaks result in high molecular weight fragments of 50- and 300-kbp as demonstrated by pulsed-field gel electrophoresis, and visualized by the TUNEL assay at 24 h of recovery. In contrast, DNA single-strand breaks, shown by the use of DNA polymerase I-mediated biotin-dATP nick translation were not so abundant. Gadd45 immunoreactivity was sequentially increased in vulnerable neurons in the infarct (4 to 24 h) and in sublethally injured neurons in the penumbra (24-48 h). Taken together, these findings suggest that Gadd45 responds to DNA damage following neonatal ischemia. Furthermore, repairing processes seem to be more active in the penumbra and therefore Gadd45 could have also a protective role in cerebral ischemia.


Subject(s)
Brain Ischemia/genetics , DNA Damage , Nerve Tissue Proteins/biosynthesis , Protein Biosynthesis , Proteins , Animals , Animals, Newborn , Brain Ischemia/metabolism , DNA Fragmentation , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Rats , Rats, Wistar , GADD45 Proteins
17.
Ann Pharm Fr ; 57(4): 309-13, 1999 Jul.
Article in French | MEDLINE | ID: mdl-10472703

ABSTRACT

Although physiological cell death has been known for decades, interest in the subject was renewed in 1972 when Kerr, Wyllie and Currie described in detail the ultrastructural changes characteristic of dying cells and coined the term apoptosis to describe the process. When dying during development or following a toxic insult, cells display a wide variety of morphological changes. A binary classification scheme suggests that physiologically appropriate death is due to apoptosis, and that pathological mechanisms involve necrosis. In this report, we will address developments in our understanding of a potential involvement of apoptotic cell death in ischemia which induce selective and delayed neuronal degeneration. Such results may open important steps in therapeutic approaches for the preservation of neurons.


Subject(s)
Apoptosis , Brain Ischemia/pathology , Brain/pathology , Ischemic Attack, Transient/pathology , Animals , Brain/physiopathology , Brain Ischemia/physiopathology , Humans , Ischemic Attack, Transient/physiopathology , Neurons/pathology , Neurons/physiology
18.
Stroke ; 30(9): 1916-23; discussion 1923-4, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10471445

ABSTRACT

BACKGROUND AND PURPOSE: The contribution of inflammatory response to the pathogenesis of ischemic lesions in the neonate is still uncertain. This study described the chronological sequence of inflammatory changes that follow cerebral ischemia with reperfusion in the neonatal P7 rat. METHODS: P7 rats underwent left middle cerebral artery electrocoagulation associated with 1-hour left common carotid artery occlusion. The spatiotemporal pattern of cellular responses was characterized immunocytochemically with the use of antibodies against rat endogenous immunoglobulins to visualize the area of the breakdown of the blood-brain barrier. Infiltration of neutrophils and T lymphocytes was demonstrated by antibodies against myeloperoxidase and a pan-T cell marker, respectively. Antibodies ED1 and OX-42 were applied to identify microglial cells and macrophages. The response of astrocytes was shown with antibodies against glial fibrillary acidic protein. Cell survival was assessed by Bcl-2 expression. Cell death was demonstrated by DNA fragmentation with the use of the terminal deoxynucleotidyl transferase-mediated dUTP biotin nick end labeling (TUNEL) assay and Bax immunodetection. RESULTS: Endogenous immunoglobulin extravasation through the blood-brain barrier occurred at 2 hours of recirculation and persisted until 1 month after ischemia. Neutrophil infiltration began at 24 hours and peaked at 72 to 96 hours (30+/-3.4 neutrophils per 0.3 mm(2); P<0.0001), then disappeared at 14 days after ischemia. T cells were observed between 24 and 96 hours of reperfusion. Resident microglia-macrophages exhibited morphological remnants and expressed the cell death inhibitor Bcl-2 at 24 hours of recirculation. They became numerous within the next 48 hours and peaked at 7 days after ischemia. Phenotypic changes of resident astrocytes were apparent at 24 hours, and they proliferated between 48 hours and 7 days after ischemia. Progressively inflammatory cells showed DNA fragmentation and the cell death activator Bax expression. Cell elimination continued until there was a complete disappearance of the frontoparietal cortex. CONCLUSIONS: These data demonstrate that perinatal ischemia with reperfusion triggers acute inflammatory responses with granulocytic cell infiltration, which may be involved in accelerating the destructive processes.


Subject(s)
Animals, Newborn/physiology , Brain Ischemia/complications , Cerebral Cortex/pathology , Encephalitis/etiology , Reperfusion Injury/complications , Animals , Apoptosis , Astrocytes/pathology , Blood-Brain Barrier , Brain Ischemia/pathology , Encephalitis/pathology , Encephalitis/physiopathology , Female , Lymphocytes/pathology , Macrophages/pathology , Male , Microglia/pathology , Neutrophils/pathology , Rats , Rats, Wistar , Reperfusion Injury/pathology , Time Factors
19.
Brain Res Dev Brain Res ; 112(1): 139-44, 1999 Jan 11.
Article in English | MEDLINE | ID: mdl-9974168

ABSTRACT

To investigate whether long-term functional consequences of status epilepticus (SE) induced by pentylenetetrazol in 10-day-old rats correlated with cell injury and/or death, acid fuchsin and TUNEL staining were performed between 4 to 144 h after SE. Acid fuchsin stained hippocampus, amygdala and cerebral cortex at 24 h but not at 72 and 144 h. No DNA fragmentation was apparent at any time. Thus, immature neurons subjected to sustained seizures suffer transiently but survive probably by activating repair processes.


Subject(s)
Animals, Newborn/physiology , Brain/pathology , Convulsants , Pentylenetetrazole , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Animals , Benzenesulfonates , Cell Death/physiology , Coloring Agents , In Situ Nick-End Labeling , Rats , Rats, Sprague-Dawley , Status Epilepticus/physiopathology , Time Factors
20.
Biomed Pharmacother ; 52(6): 264-9, 1998.
Article in English | MEDLINE | ID: mdl-9755826

ABSTRACT

Hypoxic-ischemic neuronal death has long been considered to represent necrosis, but it now appears that many brain neurons undergo apoptosis after either global or focal ischemic insults. Recent studies demonstrated: 1) DNA cleavage into oligonucleosome-sized fragments demonstrated by a typical ladder pattern; 2) early endonuclease activation, as demonstrated by the presence of high molecular weight DNA fragments (300 to 50 kbp); 3) chromatin condensation and apoptotic bodies formation; 4) activation of apoptosis-associated proteins. These results may indicate that apoptosis contributes to the development of the ischemic infarct and is probably substantially distinct from ischemia-triggered excitotoxicity, which tends to produce necrosis.


Subject(s)
Apoptosis , Brain/pathology , Ischemic Attack, Transient/pathology , Animals , Brain/physiopathology , Cell Survival , Chromatin/pathology , Chromatin/physiology , DNA/isolation & purification , DNA/metabolism , DNA Fragmentation , Electrophoresis, Gel, Pulsed-Field , Immunohistochemistry , Ischemic Attack, Transient/physiopathology , Male , Necrosis , Neurons/pathology , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-bcl-2/analysis , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/analysis , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...