Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Cell Death Dis ; 6: e1966, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26539913

ABSTRACT

Cyclopentenone prostaglandins (CyPGs), such as 15-deoxy-Δ(12,14)-prostaglandin J2 (15dPGJ2), are reactive prostaglandin metabolites exerting a variety of biological effects. CyPGs are produced in ischemic brain and disrupt the ubiquitin-proteasome system (UPS). Ubiquitin-C-terminal hydrolase L1 (UCH-L1) is a brain-specific deubiquitinating enzyme that has been linked to neurodegenerative diseases. Using tandem mass spectrometry (MS) analyses, we found that the C152 site of UCH-L1 is adducted by CyPGs. Mutation of C152 to alanine (C152A) inhibited CyPG modification and conserved recombinant UCH-L1 protein hydrolase activity after 15dPGJ2 treatment. A knock-in (KI) mouse expressing the UCH-L1 C152A mutation was constructed with the bacterial artificial chromosome (BAC) technique. Brain expression and distribution of UCH-L1 in the KI mouse was similar to that of wild type (WT) as determined by western blotting. Primary cortical neurons derived from KI mice were resistant to 15dPGJ2 cytotoxicity compared with neurons from WT mice as detected by the WST-1 cell viability assay and caspase-3 and poly ADP ribose polymerase (PARP) cleavage. This protective effect was accompanied with significantly less ubiquitinated protein accumulation and aggregation as well as less UCH-L1 aggregation in C152A KI primary neurons after 15dPGJ2 treatment. Additionally, 15dPGJ2-induced axonal injury was also significantly attenuated in KI neurons as compared with WT. Taken together, these studies indicate that UCH-L1 function is important in hypoxic neuronal death, and the C152 site of UCH-L1 has a significant role in neuronal survival after hypoxic/ischemic injury.


Subject(s)
Brain Ischemia/genetics , Cyclopentanes/toxicity , Neurons/drug effects , Neurons/physiology , Point Mutation , Prostaglandins/toxicity , Ubiquitin Thiolesterase/genetics , Animals , Binding Sites , Brain Ischemia/enzymology , Brain Ischemia/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/enzymology , Neurons/metabolism , Rats , Ubiquitin Thiolesterase/biosynthesis
2.
J Cereb Blood Flow Metab ; 21(12): 1411-21, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11740202

ABSTRACT

Cell death-regulatory genes like caspases and bcl-2 family genes are involved in delayed cell death in the CA1 sector of hippocampus after global cerebral ischemia, but little is known about the mechanisms that trigger their expression. The authors found that expression of Fas and Fas-ligand messenger ribonucleic acid and protein was induced in vulnerable CA1 neurons at 24 and 72 hours after global ischemia. Fas-associating protein with a novel death domain (FADD) also was upregulated and immunoprecipitated and co-localized with Fas. Caspase-10 was activated and interacted with FADD protein to an increasing extent as the duration of ischemia increased. Moreover, caspase-10 co-localized with both FADD and caspase-3. These findings suggest that Fas-mediated death signaling may play an important role in signaling hippocampal neuronal death in CA1 after global cerebral ischemia.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Hippocampus/pathology , fas Receptor/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 10 , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Fas Ligand Protein , Fas-Associated Death Domain Protein , Gene Expression , Hippocampus/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , fas Receptor/genetics
3.
J Cereb Blood Flow Metab ; 21(12): 1422-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11740203

ABSTRACT

Carboxypeptidase E, an exoprotease involved in the processing of bioactive peptides released by a regulated secretory pathway, was identified in a subtractive complementary DNA library derived from an ischemic rat brain by differential screening. In situ hybridization and immunocytochemical analysis showed the presence of carboxypeptidase E messenger RNA and protein in the cerebral cortex, thalamus, striatum, and hippocampus of a healthy rat brain. After 15 minutes of transient global ischemia followed by 8 hours of reperfusion, increased levels of carboxypeptidase E messenger RNA and protein were observed in the hippocampal CA1 and CA3 regions and in the cortex, as detected by Northern and Western blot analyses and in situ hybridization. After extended reperfusion (24 to 72 hours), both carboxypeptidase E messenger RNA and protein levels were decreased. The ischemia-induced changes in carboxypeptidase E expression suggest that this enzyme may play a role in modulating the brain's response to ischemia.


Subject(s)
Brain Ischemia/metabolism , Carboxypeptidases/genetics , Gene Expression Regulation, Enzymologic , Animals , Apoptosis , Blotting, Western , Brain Ischemia/pathology , Carboxypeptidase H , Carboxypeptidases/analysis , Carboxypeptidases/metabolism , DNA, Complementary/isolation & purification , Gene Library , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Male , Neurons/enzymology , Neurons/pathology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
4.
J Neurotrauma ; 18(7): 675-89, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11497094

ABSTRACT

DNA damage is a common sequela of traumatic brain injury (TBI). Available techniques for the in situ identification of DNA damage include DNA polymerase I-mediated biotin-dATP nick-translation (PANT), the Klenow fragment of DNA polymerase I-mediated biotin-dATP nick-end labeling (Klenow), and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL). While TUNEL has been widely utilized to detect primarily double-strand DNA breaks, the use of PANT to detect primarily single-strand DNA breaks and Klenow to detect both single- and double-strand DNA breaks has not been reported after TBI. Accordingly, coronal brain sections from naive rats and rats at 0, 0.5, 1, 2, 6, 24, and 72 h (n = 3-5/group) after controlled cortical impact with imposed secondary insult were processed using the PANT, Klenow, and TUNEL methods. Cells with DNA breaks were detected by PANT in the ipsilateral hemisphere as early as 0.5 h after injury and were maximal at 6 h (cortex = 66.3+/-15.8, dentate gyrus 58.6+/-12.8, CA1 = 15.8+/-5.9, CA3 = 12.8+/-4.2 cells/x 400 field, mean +/- SEM, all p < 0.05 versus naive). Cells with DNA breaks were detected by Klenow as early as 30 min and were maximal at 24 h (cortex = 56.3+/-14.3, dentate gyrus 78.0+/-16.7, CA1 = 25.8+/-4.7, CA3 = 29.3+/-15.1 cells/x 400 field, all p < 0.05 versus naive). Cells with DNA breaks were not detected by TUNEL until 2 h and were maximal at 24 h (cortex = 47.7+/-21.4, dentate gyrus 63.0+/-11.9, CA1 = 5.6+/-5.4, CA3 = 6.9+/-3.7 cells/x 400 field, cortex and dentate gyrus p < 0.05 versus naive). Dual-label immunofluorescence revealed that PANT-positive cells were predominately neurons. These data demonstrate that TBI results in extensive DNA damage, which includes both single- and double-strand breaks in injured cortex and hippocampus. The presence of multiple types of DNA breaks implicate several pathways in the evolution of DNA damage after TBI.


Subject(s)
Brain Injuries/genetics , DNA Damage/genetics , DNA Nucleotidylexotransferase/genetics , DNA Polymerase I/genetics , DNA, Single-Stranded/genetics , Animals , Brain Injuries/pathology , Disease Models, Animal , In Situ Nick-End Labeling , Male , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley , Time Factors
5.
J Neurochem ; 77(6): 1508-19, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11413234

ABSTRACT

Bax is a pro-apoptotic Bcl-2 family protein that regulates programmed cell death through homodimerization and through heterodimerization with Bcl-2. Bax alpha is encoded by six exons and undergoes alternative splicing. Bax kappa, a splice variant of Bax with conserved BH1, BH2 and BH3 binding domains and a C-terminal transmembrane domain (TM), but with an extra 446-bp insert between exons 1 and 2 leading to loss of an N-terminal ART domain, was identified from an ischemic rat brain cDNA library. Expression of Bax kappa mRNA and protein was up-regulated in hippocampus after cerebral ischemic injury. The increased Bax kappa mRNA was distributed mainly in selectively vulnerable hippocampal CA1 neurons that are destined to die after global ischemia. Overexpression of Bax kappa protein in HN33 mouse hippocampal neuronal cells induced cell death, which was partially abrogated by co-overexpression of Bcl-2. Moreover, co-overexpression of Bax kappa and Bax alpha increased HN33 cell death. The results suggest that the Bax kappa may have a role in ischemic neuronal death.


Subject(s)
Alternative Splicing/physiology , Apoptosis/physiology , Ischemic Attack, Transient/physiopathology , Neurons/cytology , Proto-Oncogene Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Gene Expression/physiology , In Situ Hybridization , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neuroblastoma , Neurons/physiology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/analysis , Rats , Transfection , Tumor Cells, Cultured , bcl-2-Associated X Protein
6.
J Neurosci ; 21(13): 4678-90, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11425895

ABSTRACT

Nuclear changes, including internucleosomal DNA fragmentation, are characteristic features of neuronal apoptosis resulting from transient cerebral ischemia and related brain insults for which the molecular mechanism has not been elucidated. Recent studies suggest that a caspase-3-mediated mechanism may be involved in the process of nuclear degradation in ischemic neurons. In this study, we cloned from rat brain a homolog cDNA encoding caspase-activated deoxyribonuclease (CAD)/DNA fragmentation factor 40 (DFF40), a 40 kDa nuclear enzyme that is activated by caspase-3 and promotes apoptotic DNA degradation. Subsequently, we investigated the role of CAD/DFF40 in the induction of internucleosomal DNA fragmentation in the hippocampus in a rat model of transient global ischemia and in primary neuronal cultures under ischemia-like conditions. At 8-72 hr after ischemia, CAD/DFF40 mRNA and protein were induced in the degenerating hippocampal CA1 neurons. CAD/DFF40 formed a heterodimeric complex in the nucleus with its natural inhibitor CAD (ICAD) and was activated after ischemia in a delayed manner (>24 hr) by caspase-3, which translocated into the nucleus and cleaved ICAD. Furthermore, an induced CAD/DFF40 activity was detected in nuclear extracts in both in vivo and in vitro models, and the DNA degradation activity of CAD/DFF40 was inhibited by purified ICAD protein. These results strongly suggest that CAD/DFF40 is the endogenous endonuclease that mediates caspase-3-dependent internucleosomal DNA degradation and related nuclear alterations in ischemic neurons.


Subject(s)
Apoptosis , Caspases/metabolism , DNA Fragmentation/physiology , Deoxyribonucleases/metabolism , Ischemic Attack, Transient/metabolism , Neurons/metabolism , Animals , Apoptosis Regulatory Proteins , Brain/blood supply , Brain/metabolism , Caspase 3 , Cells, Cultured , Cloning, Molecular , DNA Fragmentation/drug effects , Deoxyribonucleases/genetics , Gene Expression Regulation , Gene Expression Regulation, Developmental , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Molecular Sequence Data , Neurons/cytology , Neurons/drug effects , Organ Specificity , Poly-ADP-Ribose Binding Proteins , Proteins/metabolism , Proteins/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
7.
J Cereb Blood Flow Metab ; 21(3): 233-43, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11295878

ABSTRACT

In the rat, 60 minutes of transient ischemia to the middle cerebral artery results in infarction of the caudate putamen. Ischemic preconditioning with 20 minutes of transient focal ischemia produced tolerance (attenuated infarction volume) to 60 minutes of subsequent focal ischemia administered three days, five days, or seven days later. Western blots from tolerant caudate putamen demonstrated increased bcl-2 expression, maximum at 3 days and persisting through 7 days. Immunocytochemical examination found that bcl-2 was expressed in cells with both neuronal and nonneuronal morphology in striatum after preconditioning ischemia. bcl-2 antisense oligodeoxynucleotides (ODNs), bcl-2 sense ODNs, or artificial cerebrospinal fluid (CSF, vehicle) was infused into the lateral ventricle for the 72 hours between the 20-minute ischemic preconditioning and the 60-minute period of ischemia. Antisense ODN treatment reduced expression of bcl-2 in the striatum and blocked the induction of tolerance by preconditioning ischemia. Sense and CSF treatments had no effect on either bcl-2 expression or tolerance. In this model of induced tolerance to focal ischemia, bcl-2 appears to be a major determinant.


Subject(s)
Brain Ischemia/physiopathology , Cerebral Infarction/physiopathology , Corpus Striatum/physiology , Ischemic Preconditioning , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Brain Ischemia/pathology , Cerebral Infarction/pathology , Corpus Striatum/blood supply , In Situ Nick-End Labeling , Male , Oligonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley
8.
J Cereb Blood Flow Metab ; 21(4): 321-33, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11323518

ABSTRACT

Activation of terminal caspases such as caspase-3 plays an important role in the execution of neuronal cell death after transient cerebral ischemia. Although the precise mechanism by which terminal caspases are activated in ischemic neurons remains elusive, recent studies have postulated that the mitochondrial cell death-signaling pathway may participate in this process. The bcl-2 family member protein Bax is a potent proapoptotic molecule that, on translocation from cytosol to mitochondria, triggers the activation of terminal caspases by increasing mitochondrial membrane permeability and resulting in the release of apoptosis-promoting factors, including cytochrome c. In the present study, the role of intracellular Bax translocation in ischemic brain injury was investigated in a rat model of transient focal ischemia (30 minutes) and reperfusion (1 to 72 hours). Immunochemical studies revealed that transient ischemia induced a rapid translocation of Bax from cytosol to mitochondria in caudate neurons, with a temporal profile and regional distribution coinciding with the mitochondrial release of cytochrome c and caspase-9. Further, in postischemic caudate putamen in vivo and in isolated brain mitochondria in vitro, the authors found enhanced heterodimerization between Bax and the mitochondrial membrane permeabilization-related proteins adenine nucleotide translocator (ANT) and voltage-dependent anion channel. The ANT inhibitor bongkrekic acid prevented Bax and ANT interactions and inhibited Bax-triggered caspase-9 release from isolated brain mitochondria in vitro. Bongkrekic acid also offered significant neuroprotection against ischemia-induced caspase-3 and caspase-9 activation and cell death in the brain. These results strongly suggest that the Bax-mediated mitochondrial apoptotic signaling pathway may play an important role in ischemic neuronal injury.


Subject(s)
Apoptosis/physiology , Ischemic Attack, Transient/metabolism , Mitochondria/metabolism , Neurons/cytology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Animals , Blotting, Western , Bongkrekic Acid/pharmacology , Caspase 3 , Caspase 9 , Caspases/metabolism , Cytochrome c Group/metabolism , Dimerization , Male , Mitochondria/chemistry , Mitochondrial ADP, ATP Translocases/antagonists & inhibitors , Mitochondrial ADP, ATP Translocases/metabolism , Neurons/enzymology , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/chemistry , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , bcl-2-Associated X Protein
9.
J Cereb Blood Flow Metab ; 21(2): 99-109, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11176275

ABSTRACT

Programmed cell death (PCD) is an ordered and tightly controlled set of changes in gene expression and protein activity that results in neuronal cell death during brain development. This article reviews the molecular pathways by which PCD is executed in mammalian cells and the potential relation of these pathways to pathologic neuronal cell death. Whereas the classical patterns of apoptotic morphologic change often do not appear in the brain after ischemia, there is emerging biochemical and pharmacologic evidence suggesting a role for PCD in ischemic brain injury. The most convincing evidence for the induction of PCD after ischemia includes the altered expression and activity in the ischemic brain of deduced key death-regulatory genes. Furthermore, studies have shown that alterations in the activity of these gene products by peptide inhibitors, viral vector-mediated gene transfer, antisense oligonucleotides, or transgenic mouse techniques determine, at least in part, whether ischemic neurons live or die after stroke. These studies provide strong support for the hypothesis that PCD contributes to neuronal cell death caused by ischemic injury. However, many questions remain regarding the precise pathways that initiate, sense, and transmit cell death signals in ischemic neurons and the molecular mechanisms by which neuronal cell death is executed at different stages of ischemic injury. Elucidation of these pathways and mechanisms may lead to the development of novel therapeutic strategies for brain injury after stroke and related neurologic disorders.


Subject(s)
Apoptosis , Brain Ischemia/pathology , Animals , Apoptosis/genetics , Brain Ischemia/prevention & control , Caspase Inhibitors , DNA Fragmentation , Enzyme Inhibitors/therapeutic use , Gene Expression , Humans , Mitochondria/physiology , Neurons/pathology , Neurons/ultrastructure , Proto-Oncogene Proteins c-bcl-2/physiology
10.
J Pediatr ; 138(1): 18-25, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11148507

ABSTRACT

BACKGROUND: Excitotoxicity is an important mechanism in secondary neuronal injury after traumatic brain injury (TBI). Excitatory amino acids (EAAs) are increased in cerebrospinal fluid (CSF) in adults after TBI; however, studies in pediatric head trauma are lacking. We hypothesized that CSF glutamate, aspartate, and glycine would be increased after TBI in children and that these increases would be associated with age, child abuse, poor outcome, and cerebral ischemia. METHODS: EAAs were measured in 66 CSF samples from 18 children after severe TBI. Control samples were obtained from 19 children who received lumbar punctures to rule out meningitis. RESULTS: Peak and mean CSF glycine and peak CSF glutamate levels were increased versus control values. Subgroups of patients with TBI were compared by using univariate regression analysis. Massive increases in CSF glutamate were found in children <4 years old and in child abuse victims. Increased CSF glutamate and glycine were associated with poor outcome. A trend toward an association between high glutamate concentration and ischemic blood flow was observed. CONCLUSIONS: CSF EAAs are increased in infants and children with severe TBI. Young age and child abuse were associated with extremely high CSF glutamate concentrations after TBI. A possible role for excitotoxicity after pediatric TBI is supported.


Subject(s)
Aspartic Acid/cerebrospinal fluid , Brain Injuries/cerebrospinal fluid , Brain Injuries/etiology , Cerebral Ventricles , Child Abuse , Excitatory Amino Acids/cerebrospinal fluid , Glutamic Acid/cerebrospinal fluid , Glycine/cerebrospinal fluid , Adolescent , Age Factors , Brain Injuries/diagnostic imaging , Brain Injuries/mortality , Brain Ischemia/etiology , Case-Control Studies , Child , Child Abuse/statistics & numerical data , Child, Preschool , Disabled Persons/statistics & numerical data , Glasgow Coma Scale , Glasgow Outcome Scale , Humans , Infant , Prognosis , Survival Analysis , Time Factors , Tomography, X-Ray Computed
11.
Crit Care Med ; 29(12): 2287-93, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11801827

ABSTRACT

OBJECTIVES: To measure adenosine concentration in the cerebrospinal fluid of infants and children after severe traumatic brain injury and to evaluate the contribution of patient age, Glasgow Coma Scale score, mechanism of injury, Glasgow Outcome Score, and time after injury to cerebrospinal fluid adenosine concentrations. To evaluate the relationship between cerebrospinal fluid adenosine and glutamate concentrations in this population. DESIGN: Prospective survey. SETTING: Pediatric intensive care unit in a university-based children's hospital. PATIENTS: Twenty-seven critically ill infants and children who had severe traumatic brain injury (Glasgow Coma Scale < 8), who required placement of an intraventricular catheter and drainage of cerebrospinal fluid as part of their neurointensive care. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Patients ranged in age from 2 months to 14 yrs. Cerebrospinal fluid samples (n = 304) were collected from 27 patients during the first 7 days after traumatic brain injury. Control cerebrospinal fluid samples were obtained from lumbar puncture on 21 infants and children without traumatic brain injury or meningitis. Adenosine concentration was measured by using high-pressure liquid chromatography. Adenosine concentration was increased markedly in cerebrospinal fluid of children after traumatic brain injury vs. controls (p < .001). The increase in cerebrospinal fluid adenosine was independently associated with Glasgow Coma Scale < or = 4 vs. > 4 and time after injury (both p < .005). Cerebrospinal fluid adenosine concentration was not independently associated with either age (< or = 4 vs. > 4 yrs), mechanism of injury (abuse vs. other), or Glasgow Outcome Score (good/moderately disabled vs. severely disabled, vegetative, or dead). Of the 27 patients studied, 18 had cerebrospinal fluid glutamate concentration previously quantified by high-pressure liquid chromatography. There was a strong association between increases in cerebrospinal fluid adenosine and glutamate concentrations (p < .005) after injury. CONCLUSIONS: Cerebrospinal fluid adenosine concentration is increased in a time- and severity-dependent manner in infants and children after severe head injury. The association between cerebrospinal fluid adenosine and glutamate concentrations may reflect an endogenous attempt at neuroprotection against excitotoxicity after severe traumatic brain injury.


Subject(s)
Adenosine/cerebrospinal fluid , Brain Injuries/cerebrospinal fluid , Brain Injuries/physiopathology , Adolescent , Brain Injuries/etiology , Case-Control Studies , Child , Child Abuse , Child, Preschool , Excitatory Amino Acids/cerebrospinal fluid , Glasgow Coma Scale , Glasgow Outcome Scale , Glutamic Acid/cerebrospinal fluid , Humans , Infant , Linear Models , Multivariate Analysis , Pennsylvania , Prospective Studies , Time Factors
12.
Crit Care Med ; 28(10): 3511-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11057809

ABSTRACT

OBJECTIVE: Core temperature is reduced spontaneously after asphyxial cardiac arrest in rats. To determine whether spontaneous hypothermia influences neurologic damage after asphyxial arrest, we compared neurologic outcome in rats permitted to develop spontaneous hypothermia vs. rats managed with controlled normothermia. INTERVENTIONS: Male Sprague-Dawley rats were asphyxiated for 8 mins and resuscitated. After extubation, a cohort of rats was managed with controlled normothermia (CN) by placement in a servo-controlled incubator set to maintain rectal temperature at 37.4 degrees C for 48 hrs. CN rats were compared with permissive hypothermia (PH) rats that were returned to an ambient temperature environment after extubation. Rats were killed at either 72 hrs (PH72hr, n = 14; CN72hr, n = 9) or 6 wks (PH6wk, n = 6, CN6wk, n = 6) after resuscitation. PH72 rats were historic controls for the CN72 rats, whereas PH6 and CN6 rats were randomized and studied contemporaneously. MEASUREMENTS: A clinical neurodeficit score (NDS) was determined daily. A pathologist blinded to group scored 40 hematoxylin and eosin -stained brain regions for damage by using a 5-point scale (0 = none, 5 = severe). Quantitative analysis of CA1 hippocampus injury was performed by counting normal-appearing neurons in a defined subsection of CA1. MAIN RESULTS: Mean rectal temperatures measured in the PH6wk rats (n = 6) were 36.9, 34.8, 35.5, 36.7, and 37.4 degrees C at 2, 8, 12, 24, and 36 hrs, respectively. Mortality rate (before termination) was lower in PH compared with CN (0/20 vs. 7/15; p < .005). PH demonstrated a more favorable progression of NDS (p = .04) and less weight loss (p < .005) compared with CN. Median histopathology scores were lower (less damage) in PH72hr vs. CN72hr for temporal cortex (0 vs. 2.5), parietal cortex (0 vs. 2), thalamus (0 vs. 3), CA1 hippocampus (1.5 vs. 4.5), CA2 hippocampus (0 vs. 3.5), subiculum (0 vs. 4), and cerebellar Purkinje cell layer (2 vs. 4) (all p < .05). There was almost complete loss of normal-appearing CA1 neurons in CN72hr rats (6 +/- 2 [mean +/- SD] normal neurons compared with 109 +/- 12 in naïve controls). In contrast, PH72hr rats demonstrated marked protection (97 +/- 23 normal-appearing neurons) that was still evident, although attenuated, at 6 wks (42 +/- 24 normal-appearing neurons, PH6wk). CONCLUSION: Rats resuscitated from asphyxial cardiac arrest develop delayed, mild to moderate, prolonged hypothermia that is neuroprotective.


Subject(s)
Asphyxia/complications , Heart Arrest/complications , Hypothermia/etiology , Hypoxia, Brain/etiology , Hypoxia, Brain/prevention & control , Animals , Body Temperature , Disease Models, Animal , Hypothermia/metabolism , Hypothermia/physiopathology , Hypoxia, Brain/mortality , Hypoxia, Brain/pathology , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Resuscitation , Single-Blind Method , Time Factors
13.
J Neurotrauma ; 17(10): 831-41, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11063051

ABSTRACT

The proto-oncogene bcl-2 plays a key role in regulating programmed cell death in neurons. The present review discusses the mechanisms by which bcl-2 family genes regulate programmed cell death, and their role in controlling cell death in cerebral ischemia and traumatic brain. Expression of several bcl-2 family members is altered in brain tissues after ischemia and trauma, suggesting that bcl-2 family genes could play a role in determining the fate of injured neurons. Furthermore, alteration of expression of bcl-2 family genes using transgenic approaches, viral vectors, or anti-sense oligonucleotides modifies neuronal cell death and neurological outcome after injury. These data suggest that the activity of bcl-2 family gene products participates in determining cellular and neurologic outcomes in ischemia and trauma. Strategies that either mimic the death-suppressor effects or inhibit the death-promoter effects of bcl-2 family gene products may improve outcome after ischemia and trauma.


Subject(s)
Apoptosis/genetics , Brain Injuries/genetics , Brain Ischemia/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Brain Injuries/metabolism , Brain Injuries/physiopathology , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Genes, bcl-2/physiology , Humans , Proto-Oncogene Mas
14.
J Neurochem ; 75(5): 2178-89, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11032908

ABSTRACT

Oxidative stress may contribute to many pathophysiologic changes that occur after traumatic brain injury. In the current study, contemporary methods of detecting oxidative stress were used in a rodent model of traumatic brain injury. The level of the stable product derived from peroxidation of arachidonyl residues in phospholipids, 8-epi-prostaglandin F(2alpha), was increased at 6 and 24 h after traumatic brain injury. Furthermore, relative amounts of fluorescent end products of lipid peroxidation in brain extracts were increased at 6 and 24 h after trauma compared with sham-operated controls. The total antioxidant reserves of brain homogenates and water-soluble antioxidant reserves as well as tissue concentrations of ascorbate, GSH, and protein sulfhydryls were reduced after traumatic brain injury. A selective inhibitor of cyclooxygenase-2, SC 58125, prevented depletion of ascorbate and thiols, the two major water-soluble antioxidants in traumatized brain. Electron paramagnetic resonance (EPR) spectroscopy of rat cortex homogenates failed to detect any radical adducts with a spin trap, 5,5-dimethyl-1-pyrroline N:-oxide, but did detect ascorbate radical signals. The ascorbate radical EPR signals increased in brain homogenates derived from traumatized brain samples compared with sham-operated controls. These results along with detailed model experiments in vitro indicate that ascorbate is a major antioxidant in brain and that the EPR assay of ascorbate radicals may be used to monitor production of free radicals in brain tissue after traumatic brain injury.


Subject(s)
Brain Chemistry , Brain Injuries/metabolism , Dinoprost/analogs & derivatives , Free Radicals/metabolism , Oxidative Stress , Animals , Antioxidants/metabolism , Ascorbic Acid/metabolism , Biomarkers/analysis , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chromatography, High Pressure Liquid , Cyclooxygenase 2 , Dinoprost/metabolism , Disease Models, Animal , Electron Spin Resonance Spectroscopy , F2-Isoprostanes , Free Radicals/analysis , Hippocampus/drug effects , Hippocampus/metabolism , Isoenzymes/antagonists & inhibitors , Male , Oxidation-Reduction , Prostaglandin-Endoperoxide Synthases , Rats , Rats, Sprague-Dawley , Wounds, Nonpenetrating
15.
Crit Care Med ; 28(9): 3218-23, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11008985

ABSTRACT

OBJECTIVES: To evaluate the effect of application of transient, moderate hypothermia on outcome after experimental traumatic brain injury (TBI) with a secondary hypoxemic insult. DESIGN: Prospective, randomized study. SETTING: University-based animal research facility. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: All rats were subjected to severe TBI followed by 30 mins of moderate hypoxemia, associated with mild hypotension. Rats were randomized to three groups: a) normothermia (37 degrees C + 0.5 degrees C); b) immediate hypothermia (32 degrees C +/- 0.5 degrees C initiated after trauma, before hypoxemia); and c) delayed hypothermia (32 degrees C +/- 0.5 degrees C after hypoxemia). The brain temperature was controlled for 4 hrs after TBI and hypoxemia. MEASUREMENTS AND MAIN RESULTS: Animals were evaluated after TBI for motor and cognitive performance using beam balance (days 1-5 after TBI), beam walking (days 1-5 after TBI), and Morris Water Maze (days 14-18 after TBI) assessments. On day 21 after TBI, rats were perfused with paraformaldehyde and brains were histologically evaluated for lesion volume and hippocampal neuron counts. All three groups showed marked deficits in beam balance, beam walking, and Morris Water Maze performance. However, these deficits did not differ between groups. There was no difference in lesion volume between groups. All animals had significant hippocampal neuronal loss on the side ipsilateral to injury, but this loss was similar between groups. CONCLUSIONS: In this rat model of severe TBI with secondary insult, moderate hypothermia for 4 hrs posttrauma failed to improve motor function, cognitive function, lesion volume or hippocampal neuronal survival. Combination therapies may be necessary in this difficult setting.


Subject(s)
Brain Concussion/physiopathology , Hypothermia, Induced , Hypoxia, Brain/physiopathology , Maze Learning/physiology , Motor Skills/physiology , Postural Balance/physiology , Animals , Body Temperature/physiology , Brain Concussion/pathology , Cell Count , Hippocampus/injuries , Hippocampus/pathology , Hippocampus/physiopathology , Hypoxia, Brain/pathology , Male , Neurons/pathology , Neurons/physiology , Rats , Rats, Sprague-Dawley
16.
J Pediatr ; 137(2): 197-204, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10931412

ABSTRACT

OBJECTIVES: To determine whether bcl-2, a protein that inhibits apoptosis, would be increased in cerebrospinal fluid (CSF) in infants and children after traumatic brain injury (TBI) and to examine the association of bcl-2 concentration with clinical variables. STUDY DESIGN: Bcl-2 was measured in CSF from 23 children (aged 2 months-16 years) with severe TBI and from 19 children without TBI or meningitis (control subjects) by enzyme-linked immunosorbent assay. CSF oligonucleosome concentration was also determined as a marker of DNA degradation. Brain samples from 2 patients undergoing emergent decompressive craniectomies were analyzed for bcl-2 with Western blot and for DNA fragmentation with TUNEL (terminal deoxynucleotidyl-transferase mediated biotin-dUTP nick-end labeling). RESULTS: CSF bcl-2 concentrations were increased in patients with TBI versus control subjects (P =.01). Bcl-2 was increased in patients with TBI who survived versus those who died (P =.02). CSF oligonucleosome concentration tended to be increased after TBI (P =.07) and was not associated with bcl-2. Brain tissue samples showed an increase in bcl-2 in patients with TBI versus adult brain bank control samples and evidence of DNA fragmentation within cells with apoptotic morphology. CONCLUSIONS: Bcl-2 may participate in the regulation of cell death after TBI in infants and children. The increase in bcl-2 seen in patients who survived is consistent with a protective role for this anti-apoptotic protein after TBI.


Subject(s)
Apoptosis , Brain Injuries/physiopathology , Proto-Oncogene Proteins c-bcl-2/cerebrospinal fluid , Adolescent , Age Factors , Analysis of Variance , Brain Injuries/cerebrospinal fluid , Brain Injuries/etiology , Brain Injuries/mortality , Case-Control Studies , Child , Child, Preschool , Female , Glasgow Coma Scale , Humans , Infant , Linear Models , Male , Multivariate Analysis , Nucleosomes/metabolism , Pennsylvania/epidemiology , Survival Analysis , Temporal Lobe/metabolism
17.
Brain Res Mol Brain Res ; 79(1-2): 169-73, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10925156

ABSTRACT

Heat shock proteins (HSP's) are a family of highly conserved proteins whose expression is increased by stress. The expression of many HSP's is induced in neurons by ischemia; however, the response of the 10 kDa mitochondrial matrix HSP (HSP10) is less well characterized. To address this issue, asphyxial cardiac arrest was induced in 28 male Sprague-Dawley rats. Northern blot analysis revealed that hsp10 mRNA was increased 2.7-fold in asphyxiated rats compared to sham-operated controls. In situ hybridization demonstrated increased mRNA in the cortex, septal nuclei, hippocampus, thalamic nuclei, purkinje cell layer of the cerebellum, and isolated brainstem nuclei of asphyxiated rats. The increase of mRNA was most robust 8 h after the injury but remained increased for 72 h. These results show that hsp10 mRNA is increased following asphyxial cardiac arrest in rats and suggest that hsp10 could be another determinate of neuronal survival after ischemia.


Subject(s)
Brain/metabolism , Chaperonin 10/genetics , Ischemic Attack, Transient/genetics , Neurons/metabolism , RNA, Messenger/genetics , Transcription, Genetic , Animals , Asphyxia , Gene Expression Regulation , Heart Arrest , Hippocampus/metabolism , Ischemic Attack, Transient/metabolism , Male , Mitochondria/metabolism , Organ Specificity , Purkinje Cells/metabolism , Rats , Rats, Sprague-Dawley
18.
J Cereb Blood Flow Metab ; 20(7): 1033-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10908036

ABSTRACT

Previous studies have shown that overexpression of bcl-2 in transgenic mice or by viral vectors protects the brain against cerebral ischemia. However, it is not known whether bcl-2, which is endogenously expressed in response to ischemia, exerts a protective effect. To address this question, the authors blocked the endogenous expression of bcl-2 after ischemia using antisense oligodeoxynucleotides (ODN). Antisense, sense, scrambled ODN, or vehicles were infused in the lateral ventricle of the rat for 24 hours after 30 minutes of temporary middle cerebral artery occlusion. Twenty-four hours later the brains were removed and bcl-2 protein expression was assayed by Western blot. Antisense ODN, but not sense or scrambled ODN treatment, significantly inhibited bcl-2 protein expression after ischemia. Bcl-2 protein expression was also studied 24 hours after 60 minutes of temporary middle cerebral artery occlusion in vehicle and antisense ODN-treated rats. After 60 minutes of ischemia and vehicle treatment, bcl-2 was expressed in many neurons in the ventral cortical mantle and the medial striatum. After antisense ODN treatment there were few neurons in this region expressing bcl-2, instead most neurons TUNEL labeled. Treatment with the antisense ODN, but not sense ODN, increased infarction volume as determined by cresyl violet staining 72 hours after ischemia compared with vehicle controls. These results suggested that endogenously expressed bcl-2 promoted survival in ischemic neurons and was not simply an epiphenomenon in neurons already destined to live or die.


Subject(s)
Brain Ischemia/physiopathology , Neurons/physiology , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Death , Male , Neurons/drug effects , Rats , Rats, Sprague-Dawley
19.
Pediatrics ; 106(1 Pt 1): 118-22, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10878160

ABSTRACT

OBJECTIVE: In experimental models of ischemic-anoxic brain injury, changes in body temperature after the insult have a profound influence on neurologic outcome. Specifically, hypothermia ameliorates whereas hyperthermia exacerbates neurologic injury. Accordingly, we sought to determine the temperature changes occurring in children after resuscitation from cardiac arrest. STUDY DESIGN: The clinical records of 13 children resuscitated from cardiac arrest were analyzed. Patients were identified through the emergency department and pediatric intensive care unit arrest logs. Only patients surviving for > or =12 hours after resuscitation were considered for analysis. Charts were reviewed for body temperatures, warming or cooling interventions, antipyretic and antimicrobial administration, and evidence of infection. RESULTS: Seven patients had a minimum temperature (T min) of < or =35 degrees C and 11 had a maximum temperature (T max) of > or =38.1 degrees C. Hypothermia often preceded hyperthermia. All 7 patients with T min < or =35 degrees C were actively warmed with heating lamps and 5 of 7 responded to warming with a rebound of body temperatures > or =38.1 degrees C. None of the 6 patients with T min >35 degrees C were actively warmed but all developed T max > or =38.1 degrees C. Six patients received antipyretics and 11 received antibiotics. Fever was not associated with a positive culture in any case. Conclusion. Spontaneous hypothermia followed by hyperthermia is common after resuscitation from cardiac arrest. Temperature should be closely monitored after cardiac arrest and fever should be managed expectantly.


Subject(s)
Fever/etiology , Heart Arrest/complications , Hypothermia/etiology , Body Temperature , Child , Child, Preschool , Heart Arrest/mortality , Heart Arrest/physiopathology , Heart Arrest/therapy , Humans , Infant , Resuscitation , Retrospective Studies
20.
J Neurosci Res ; 59(6): 767-74, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10700014

ABSTRACT

T-cell restricted intracellular antigen-related protein (TIAR) is an RNA recognition motif-type RNA-binding protein that has been implicated in the apoptotic death of T-lymphocytes and retinal pigment epithelial cells. Western blots prepared with a monoclonal antibody against TIAR showed expression in normal rat hippocampus, and induction by 15 min of global cerebral ischemia. This increased expression was evident at 8 hr after ischemia and maximal at 24 hr, whereas expression at 72 hr was reduced below basal levels. Expression of TIAR protein was also increased in parietal cortex 6 and 24 hr after 90 min of focal cerebral ischemia induced by middle cerebral artery (MCA) occlusion, as well as in cultured cortical neurons and astroglia after exposure to hypoxia in vitro. Immunocytochemistry showed that increased expression of TIAR occurred mainly in the CA1 sector of hippocampus 24 hr after global ischemia, and in cortical and striatal neurons 24 hr after 20 or 90 min of focal ischemia. Double-labeling studies showed that TIAR protein expression was co-localized with DNA damage in neuronal cells. The findings suggest that TIAR may be involved in neuronal cell death after cerebral ischemic injury.


Subject(s)
Brain Ischemia/physiopathology , Cerebral Cortex/physiopathology , Neurons/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Reperfusion Injury/physiopathology , Animals , Cell Culture Techniques , Cell Death/physiology , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Hypoxia/physiopathology , Male , Neurons/pathology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...