Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Brain Behav Immun ; 45: 15-27, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25218898

ABSTRACT

Bidirectional communication between the immune and neuroendocrine systems is not well understood in the context of traumatic brain injury (TBI). The purpose of this study was to characterize relationships between cerebrospinal fluid (CSF) cortisol and inflammation after TBI, and to determine how these relationships differ by outcome. CSF samples were collected from 91 subjects with severe TBI during days 0-6 post-injury, analyzed for cortisol and inflammatory markers, and compared to healthy controls (n=13 cortisol, n=11 inflammatory markers). Group-based trajectory analysis (TRAJ) delineated subpopulations with similar longitudinal CSF cortisol profiles (high vs. low cortisol). Glasgow Outcome Scale (GOS) scores at 6months served as the primary outcome measure reflecting global outcome. Inflammatory markers that displayed significant bivariate associations with both GOS and cortisol TRAJ (interleukin [IL]-6, IL-10, soluble Fas [sFas], soluble intracellular adhesion molecule [sICAM]-1, and tumor necrosis factor alpha [TNF]-α) were used to generate a cumulative inflammatory load score (ILS). Subsequent analysis revealed that cortisol TRAJ group membership mediated ILS effects on outcome (indirect effect estimate=-0.253, 95% CI (-0.481, -0.025), p=0.03). Correlational analysis between mean cortisol levels and ILS were examined separately within each cortisol TRAJ group and by outcome. Within the low cortisol TRAJ group, subjects with unfavorable 6-month outcome displayed a negative correlation between ILS and mean cortisol (r=-0.562, p=0.045). Conversely, subjects with unfavorable outcome in the high cortisol TRAJ group displayed a positive correlation between ILS and mean cortisol (r=0.391, p=0.006). Our results suggest that unfavorable outcome after TBI may result from dysfunctional neuroendocrine-immune communication wherein an adequate immune response is not mounted or, alternatively, neuroinflammation is prolonged. Importantly, the nature of neuroendocrine-immune dysfunction differs between cortisol TRAJ groups. These results present a novel biomarker-based index from which to discriminate outcome and emphasize the need for evaluating tailored treatments targeting inflammation early after injury.


Subject(s)
Brain Injuries/immunology , Hydrocortisone/immunology , Inflammation/cerebrospinal fluid , Adolescent , Adult , Aged , Brain Injuries/cerebrospinal fluid , Brain Injuries/rehabilitation , Case-Control Studies , Cohort Studies , Cytidine Diphosphate Choline/therapeutic use , Double-Blind Method , Fas Ligand Protein/cerebrospinal fluid , Fas Ligand Protein/immunology , Female , Glasgow Outcome Scale , Humans , Hydrocortisone/cerebrospinal fluid , Hypothermia, Induced/methods , Intercellular Adhesion Molecule-1/cerebrospinal fluid , Intercellular Adhesion Molecule-1/immunology , Interleukin-10/cerebrospinal fluid , Interleukin-10/immunology , Interleukin-1beta/cerebrospinal fluid , Interleukin-1beta/immunology , Interleukin-6/cerebrospinal fluid , Interleukin-6/immunology , Male , Middle Aged , Nootropic Agents/therapeutic use , Prognosis , Prospective Studies , Trauma Severity Indices , Treatment Outcome , Tumor Necrosis Factor-alpha/cerebrospinal fluid , Tumor Necrosis Factor-alpha/immunology , Young Adult
2.
Brain Behav Immun ; 45: 253-62, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25555531

ABSTRACT

Traumatic brain injury (TBI) results in a significant inflammatory burden that perpetuates the production of inflammatory mediators and biomarkers. Interleukin-6 (IL-6) is a pro-inflammatory cytokine known to be elevated after trauma, and a major contributor to the inflammatory response following TBI. Previous studies have investigated associations between IL-6 and outcome following TBI, but to date, studies have been inconsistent in their conclusions. We hypothesized that cohort heterogeneity, temporal inflammatory profiles, and concurrent inflammatory marker associations are critical to characterize when targeting subpopulations for anti-inflammatory therapies. Toward this objective, we used serial cerebrospinal fluid (CSF) samples to generate temporal acute IL-6 trajectory (TRAJ) profiles in a prospective cohort of adults with severe TBI (n=114). We examined the impact of injury type on IL-6 profiles, and how IL-6 profiles impact sub-acute (2weeks-3months) serum inflammatory marker load and long-term global outcome 6-12months post-injury. There were two distinct acute CSF IL-6 profiles, a high and low TRAJ group. Individuals in the high TRAJ had increased odds of unfavorable Glasgow Outcome Scale (GOS) scores at 6months (adjusted OR=3.436, 95% CI: 1.259, 9.380). Individuals in the high TRAJ also had higher mean acute CSF inflammatory load compared to individuals in the low TRAJ (p⩽0.05). The two groups did not differ with respect acute serum profiles; however, individuals in the high CSF IL-6 TRAJ also had higher mean sub-acute serum IL-1ß and IL-6 levels compared with the low TRAJ group (p⩽0.05). Lastly, injury type (isolated TBI vs. TBI+polytrauma) was associated with IL-6 TRAJ group (χ(2)=5.31, p=0.02). Specifically, there was 70% concordance between those with TBI+polytrauma and the low TRAJ; in contrast, isolated TBI was similarly distributed between TRAJ groups. These data provide evidence that sustained, elevated levels of CSF IL-6 are associated with an increased inflammatory load, and these increases are associated with increased odds for unfavorable global outcomes in the first year following TBI. Future studies should explore additional factors contributing to IL-6 elevations, and therapies to mitigate its detrimental effects on outcome.


Subject(s)
Brain Injuries/cerebrospinal fluid , Cytokines/cerebrospinal fluid , Interleukin-6/cerebrospinal fluid , Multiple Trauma/cerebrospinal fluid , Adult , Brain Injuries/immunology , Brain Injuries/rehabilitation , Cohort Studies , Cytokines/immunology , Disease Progression , Female , Glasgow Outcome Scale , Humans , Injury Severity Score , Interleukin-1beta/immunology , Interleukin-6/immunology , Logistic Models , Male , Multiple Trauma/immunology , Prognosis , Prospective Studies
3.
Neurocrit Care ; 19(1): 31-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22535278

ABSTRACT

BACKGROUND: To assess regional brain injury on magnetic resonance imaging (MRI) after pediatric cardiac arrest (CA) and to associate regional injury with patient outcome and effects of hypothermia therapy for neuroprotection. METHODS: We performed a retrospective chart review with prospective imaging analysis. Children between 1 week and 17 years of age who had a brain MRI in the first 2 weeks after CA without other acute brain injury between 2002 and 2008 were included. Brain MRI (1.5 T General Electric, Milwaukee, WI, USA) images were analyzed by 2 blinded neuroradiologists with adjudication; images were visually graded. Brain lobes, basal ganglia, thalamus, brain stem, and cerebellum were analyzed using T1, T2, and diffusion-weighted images (DWI). RESULTS: We examined 28 subjects with median age 1.9 years (IQR 0.4-13.0) and 19 (68 %) males. Increased intensity on T2 in the basal ganglia and restricted diffusion in the brain lobes were associated with unfavorable outcome (all P < 0.05). Therapeutic hypothermia had no effect on regional brain injury. Repeat brain MRI was infrequently performed but demonstrated evolution of lesions. CONCLUSION: Children with lesions in the basal ganglia on conventional MRI and brain lobes on DWI within the first 2 weeks after CA represent a group with increased risk of poor outcome. These findings may be important for developing neuroprotective strategies based on regional brain injury and for evaluating response to therapy in interventional clinical trials.


Subject(s)
Brain Injuries/etiology , Brain Injuries/pathology , Diffusion Magnetic Resonance Imaging/methods , Heart Arrest/complications , Hypothermia, Induced/methods , Adolescent , Brain/pathology , Child , Child, Preschool , Female , Follow-Up Studies , Heart Arrest/pathology , Humans , Infant , Infant, Newborn , Male , Recovery of Function , Retrospective Studies , Treatment Outcome
4.
Neurocrit Care ; 17(3): 401-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22890910

ABSTRACT

BACKGROUND: The objectives of this study were to determine effects of severe traumatic brain injury (TBI) on cerebrospinal fluid (CSF) concentrations of myelin basic protein (MBP) and to assess relationships between clinical variables and CSF MBP concentrations. METHODS: We measured serial CSF MBP concentrations in children enrolled in a randomized controlled trial evaluating therapeutic hypothermia (TH) after severe pediatric TBI. Control CSF was obtained from children evaluated, but found not to be having CNS infection. Generalized estimating equation models and Wilcoxon Rank-Sum test were used for comparisons of MBP concentrations. RESULTS: There were 27 TBI cases and 57 controls. Overall mean (± SEM) TBI case MBP concentrations for 5 days after injury were markedly greater than controls (50.49 ± 6.97 vs. 0.11 ± 0.01 ng/ml, p < 0.01). Mean MBP concentrations were lower in TBI patients <1 year versus >1 year (9.18 ± 1.67 vs. 60.22 ± 8.26 ng/ml, p = 0.03), as well as in cases with abusive head trauma (AHT) versus non-abusive TBI (14.46 ± 3.15 vs. 61.17 ± 8.65 ng/ml, p = 0.03). TH did not affect MBP concentrations. CONCLUSIONS: Mean CSF MBP increases markedly after severe pediatric TBI, but is not affected by TH. Infancy and AHT are associated with low MBP concentrations, suggesting that age-dependent myelination influences MBP concentrations after injury. Given the magnitude of MBP increases, axonal injury likely represents an important therapeutic target in pediatric TBI.


Subject(s)
Brain Injuries/cerebrospinal fluid , Brain Injuries/therapy , Child Abuse , Hypothermia, Induced/methods , Myelin Basic Protein/cerebrospinal fluid , Trauma Severity Indices , Age Factors , Biomarkers/cerebrospinal fluid , Child , Child, Preschool , Diffuse Axonal Injury/cerebrospinal fluid , Diffuse Axonal Injury/therapy , Female , Humans , Infant , Male , Sex Factors
5.
Biochim Biophys Acta ; 1757(5-6): 648-59, 2006.
Article in English | MEDLINE | ID: mdl-16740248

ABSTRACT

Since the (re)discovery of cytochrome c (cyt c) in the early 1920s and subsequent detailed characterization of its structure and function in mitochondrial electron transport, it took over 70 years to realize that cyt c plays a different, not less universal role in programmed cell death, apoptosis, by interacting with several proteins and forming apoptosomes. Recently, two additional essential functions of cyt c in apoptosis have been discovered that are carried out via its interactions with anionic phospholipids: a mitochondria specific phospholipid, cardiolipin (CL), and plasma membrane phosphatidylserine (PS). Execution of apoptotic program in cells is accompanied by substantial and early mitochondrial production of reactive oxygen species (ROS). Because antioxidant enhancements protect cells against apoptosis, ROS production was viewed not as a meaningless side effect of mitochondrial disintegration but rather playing some - as yet unidentified - role in apoptosis. This conundrum has been resolved by establishing that mitochondria contain a pool of cyt c, which interacts with CL and acts as a CL oxygenase. The oxygenase is activated during apoptosis, utilizes generated ROS and causes selective oxidation of CL. The oxidized CL is required for the release of pro-apoptotic factors from mitochondria into the cytosol. This redox mechanism of cyt c is realized earlier than its other well-recognized functions in the formation of apoptosomes and caspase activation. In the cytosol, released cyt c interacts with another anionic phospholipid, PS, and catalyzes its oxidation in a similar oxygenase reaction. Peroxidized PS facilitates its externalization essential for the recognition and clearance of apoptotic cells by macrophages. Redox catalysis of plasma membrane PS oxidation constitutes an important redox-dependent function of cyt c in apoptosis and phagocytosis. Thus, cyt c acts as an anionic phospholipid specific oxygenase activated and required for the execution of essential stages of apoptosis. This review is focused on newly discovered redox mechanisms of complexes of cyt c with anionic phospholipids and their role in apoptotic pathways in health and disease.


Subject(s)
Cytochromes c/metabolism , Mitochondria/metabolism , Phospholipids/metabolism , Amino Acid Sequence , Animals , Antioxidants/metabolism , Apoptosis , Atherosclerosis/metabolism , Cardiolipins/metabolism , Cell Membrane/metabolism , Electron Transport , Humans , Mitochondrial Membranes/metabolism , Molecular Sequence Data , Oxidation-Reduction , Oxygenases/metabolism , Peroxidases/metabolism , Reactive Oxygen Species/metabolism
6.
J Clin Invest ; 104(5): 647-56, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10487779

ABSTRACT

Nitric oxide (NO) derived from the inducible isoform of NO synthase (iNOS) is an inflammatory product implicated both in secondary damage and in recovery from brain injury. To address the role of iNOS in experimental traumatic brain injury (TBI), we used 2 paradigms in 2 species. In a model of controlled cortical impact (CCI) with secondary hypoxemia, rats were treated with vehicle or with 1 of 2 iNOS inhibitors (aminoguanidine and L-N-iminoethyl-lysine), administered by Alzet pump for 5 days and 1. 5 days after injury, respectively. In a model of CCI, knockout mice lacking the iNOS gene (iNOS(-/-)) were compared with wild-type (iNOS(+/+)) mice. Functional outcome (motor and cognitive) during the first 20 days after injury, and histopathology at 21 days, were assessed in both studies. Treatment of rats with either of the iNOS inhibitors after TBI significantly exacerbated deficits in cognitive performance, as assessed by Morris water maze (MWM) and increased neuron loss in vulnerable regions (CA3 and CA1) of hippocampus. Uninjured iNOS(+/+) and iNOS(-/-) mice performed equally well in both motor and cognitive tasks. However, after TBI, iNOS(-/-) mice showed markedly worse performance in the MWM task than iNOS(+/+) mice. A beneficial role for iNOS in TBI is supported.


Subject(s)
Brain Damage, Chronic/prevention & control , Brain Injuries/enzymology , Nerve Tissue Proteins/physiology , Nitric Oxide Synthase/physiology , Nitric Oxide/physiology , Animals , Brain Damage, Chronic/etiology , Brain Damage, Chronic/pathology , Brain Injuries/pathology , Cognition Disorders/etiology , Enzyme Induction , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Hippocampus/radiation effects , Hypoxia/enzymology , Hypoxia/pathology , Lysine/analogs & derivatives , Lysine/pharmacology , Male , Maze Learning/drug effects , Maze Learning/radiation effects , Mice , Mice, Knockout , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/enzymology , Neurons/radiation effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Polymerase Chain Reaction , Radiation Injuries, Experimental/drug therapy , Rats , Rats, Sprague-Dawley , Whole-Body Irradiation , Wounds, Nonpenetrating/enzymology , Wounds, Nonpenetrating/pathology
7.
Biochim Biophys Acta ; 1256(3): 263-74, 1995 Jun 06.
Article in English | MEDLINE | ID: mdl-7786887

ABSTRACT

Epoxyeicosatrienoic acids (EETs), normally present in brain and blood, appear to be released from atherosclerotic vessels in large amounts. Once intravascular, EETs can constrict renal arteries in vivo and dilate cerebral and coronary arteries in vitro. Whether EETs in blood will alter cerebral blood flow (CBF) in vivo is unknown. In the present study, the chemical synthesis of four EET regioisomers was optimized, and their identity and structural integrity established by chromatographic and mass spectral methods. The chemically labile EETs were converted to a sodium salt, complexed with albumin, and infused into anesthetized rats via the common carotid. The objective was to test whether sustained, high levels of intravascular EETs alter CBF. The CBF (cortical H2 clearance) was measured before and 30 min after the continuous infusion of 14,15- (n = 5), 11,12- (n = 5), 8,9- (n = 7) and 5,6-EET (unesterified or as the methyl ester, n = 5 for each). Neither the CBF nor the systemic blood pressure was affected by EETs. Because the infusions elevated the plasma concentrations of EETs about 700-fold above normal levels (1.0 nM), it is unlikely that EETs released from atherosclerotic vessels will alter CBF.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Cerebrovascular Circulation/drug effects , 8,11,14-Eicosatrienoic Acid/administration & dosage , 8,11,14-Eicosatrienoic Acid/chemical synthesis , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Infusions, Intravenous , Male , Rats , Rats, Wistar
8.
J Leukoc Biol ; 61(3): 279-85, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9060450

ABSTRACT

Traumatic brain injury (TBI) is often accompanied by an acute inflammatory reaction mediated initially by neutrophils. Adhesion molecules expressed on vascular endothelium are requisite elements during recruitment of leukocytes at sites of inflammation. In a rat model of TBI the induction and persistent expression of E-selectin (CD62E) on cerebrovascular endothelium ipsilateral, but not contralateral, to the site of contusion was demonstrated (P < 0.05 at 4 and 48 h posttrauma). In addition, these studies confirmed up-regulation and prolonged expression of ICAM-1 (CD54) on endothelium in the traumatized hemisphere (P < 0.05 at 4, 24, 48, and 72 h posttrauma). It is of interest that increased expression of CD54 was noted on blood vessels in the contralateral, non-traumatized hemisphere 48 h posttrauma. Expression of a third endothelial adhesion molecule, PECAM-1 (CD31), was unchanged following trauma. Administration of a murine monoclonal antibody (TM-8) that inhibits the adhesive function of CD54 blocked a significant portion (37.9%) of neutrophil recruitment 24 h posttrauma (P = 0.04). Employing immunocytochemistry and a monoclonal antibody specific for rat neutrophils (RP-3), peak infiltration of neutrophils was shown to occur 48 h after trauma. In contrast to emigration of neutrophils from blood vessels within the contusion, however, entry of neutrophils occurred from the surrounding leptomeninges and choroidal vessels. These studies demonstrate the relevance of CD54 (ICAM-1) in recruitment of neutrophils following TBI. However, the majority of neutrophil influx relies on endothelial adhesion molecules other than CD54. Because emigration of neutrophils was shown to occur predominantly from vessels within the leptomeninges and choroid plexus, intrathecal delivery of agents that inhibit the adhesive interactions between neutrophils, endothelial CD54, and other endothelial adhesion molecules to be defined may offer a novel form of therapy to prevent the acute inflammatory response that follows TBI.


Subject(s)
Brain Injuries/metabolism , E-Selectin/metabolism , Intercellular Adhesion Molecule-1/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers , Cell Movement , Endothelium, Vascular/metabolism , Male , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/physiology , Rats , Rats, Sprague-Dawley
9.
J Leukoc Biol ; 67(2): 160-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10670575

ABSTRACT

Platelet (P-) selectin and intercellular adhesion molecule-1 (ICAM-1) mediate accumulation of neutrophils in brain. However, the mechanisms regulating neutrophil accumulation and damage after traumatic brain injury (TBI) are poorly defined. We hypothesized that mice deficient in both P-selectin and ICAM-1 (-/-) would have decreased brain neutrophil accumulation and edema, and improved functional and histopathological outcome after TBI compared with wild-type (+/+). In Protocol I, neutrophils and brain water content were quantified at 24 h after TBI. No difference in brain neutrophil accumulation was observed between groups; however, brain edema was decreased in dual P-selectin and ICAM-1 -/- (P < 0.05 vs. +/+ mice). In Protocol II, after TBI, tests of motor and memory function and histopathology were assessed over 21 days. No difference in motor or memory function or histopathological damage was observed between +/+ and -/- mice. A role for adhesion molecules in the pathogenesis of brain edema independent of leukocyte accumulation in brain is suggested.


Subject(s)
Brain Edema/prevention & control , Brain Injuries/complications , Intercellular Adhesion Molecule-1/physiology , P-Selectin/physiology , Animals , Blood-Brain Barrier , Brain Edema/etiology , Brain Edema/metabolism , Brain Injuries/metabolism , Chemotaxis, Leukocyte , Contusions/complications , Contusions/metabolism , Intercellular Adhesion Molecule-1/genetics , Male , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Movement Disorders/etiology , P-Selectin/genetics , Peritonitis/immunology , Wounds, Nonpenetrating/complications , Wounds, Nonpenetrating/metabolism
10.
Neuroscience ; 305: 268-78, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26265550

ABSTRACT

Extremely mild hypothermia to 36.0 °C is not thought to appreciably differ clinically from 37.0 °C. However, it is possible that 36.0 °C stimulates highly sensitive hypothermic signaling mechanism(s) and alters biochemistry. To the best of our knowledge, no such ultra-sensitive pathway/mechanisms have been described. Here we show that cold stress protein RNA binding motif 3 (RBM3) increases in neuron and astrocyte cultures maintained at 33 °C or 36 °C for 24 or 48 h, compared to 37 °C controls. Neurons cultured at 36 °C also had increased global protein synthesis (GPS). Finally, we found that melatonin or fibroblast growth factor 21 (FGF21) augmented RBM3 upregulation in young neurons cooled to 36 °C. Our results show that a 1 °C reduction in temperature can induce pleiotropic biochemical changes by upregulating GPS in neurons which may be mediated by RBM3 and that this process can be pharmacologically mimicked and enhanced with melatonin or FGF21.


Subject(s)
Neurons/metabolism , RNA-Binding Proteins/metabolism , Temperature , Analysis of Variance , Animals , Antioxidants/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Female , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hypothermia, Induced/methods , Melatonin/pharmacology , Neurons/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Time Factors
11.
J Cereb Blood Flow Metab ; 8(4): 546-51, 1988 Aug.
Article in English | MEDLINE | ID: mdl-3392115

ABSTRACT

Platelet-activating factor has been implicated in a variety of disease processes including ischemic brain injury and endotoxic shock, but its effects on cerebral blood flow (CBF) and metabolism in normal brain have not been described. The effects of platelet-activating factor on global CBF (hydrogen clearance) and the global cerebral metabolic rate for oxygen (CMRO2) were studied in halothane-N2O anesthetized Wistar rats. Hexadecyl-platelet-activating factor infused into the right carotid artery (67 pmol/min) for 60 min decreased mean arterial pressure (MAP) from 122 +/- 4 (x +/- SEM) to 77 +/- 6 mm Hg and CBF from 159 +/- 12 to 116 +/- 14 ml/100 g/min (p less than 0.002). In contrast, CMRO2 increased from 9.7 +/- 0.9 to 11.7 +/- 1.1 ml/100 g/min after 15 min (p less than 0.05). In controls rendered similarly hypotensive by blood withdrawal and infused with the platelet-activating factor vehicle, CMRO2 was unchanged, whereas CBF transiently decreased then returned to baseline at 60 min. These cerebrovascular and cerebrometabolic effects of PAF are reminiscent of and may be relevant to hypoperfusion and hypermetabolism observed after global brain ischemia and in endotoxic shock.


Subject(s)
Brain/metabolism , Cerebrovascular Circulation/drug effects , Platelet Activating Factor/pharmacology , Animals , Blood Pressure/drug effects , Carotid Arteries , Cerebral Hemorrhage/complications , Hypotension/etiology , Hypotension/physiopathology , Injections, Intra-Arterial , Male , Oxygen Consumption , Rats , Rats, Inbred Strains
12.
J Cereb Blood Flow Metab ; 15(5): 860-4, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7673379

ABSTRACT

As part of the acute inflammatory response, neutrophils accumulate in the central nervous system after injury. Recently, a soluble human recombinant complement receptor (sCR1; BRL 55730; T Cell Sciences, Inc., Cambridge, MA, U.S.A.) has been developed that inhibits the activation of both the classical and the alternative pathways of complement. sCR1 attenuates the effects of the acute inflammatory response in several models of injury outside the central nervous system. The role of complement in traumatic brain injury, however, remains undefined. We hypothesized that treatment with sCR1 would attenuate neutrophil accumulation in the brain after cerebral trauma. Using a randomized, blinded protocol, 18 anesthetized Sprague-Dawley rats were pre-treated with sCR1 or saline (control) at both 2 h and 2 min before trauma (weight drop) to the exposed right parietal cortex. A third dose of sCR1 (or saline) was given 6 h after trauma. Coronal brain sections centered on the site of trauma were obtained at 24 h after trauma and analyzed for myeloperoxidase (MPO) activity as a marker of neutrophil accumulation. Complete blood counts with differential were obtained before treatment with sCR1 and at 24 h after trauma. At 24 h after trauma, brain MPO activity was reduced by 41% in sCR1-treated rats compared with control rats [0.1599 +/- 0.102 versus 0.2712 +/- 0.178 U/g (mean +/- SD); p = 0.02]. The neutrophil count in peripheral blood increased approximately twofold in both groups. Neutrophil accumulation occurring in the brain after trauma is inhibited by sCR1 treatment. This suggests that complement activation is involved in the local inflammatory response to traumatic brain injury and plays an important role in neutrophil accumulation in the injured brain.


Subject(s)
Brain Injuries/pathology , Complement C1/metabolism , Neutrophils/pathology , Receptors, Complement/physiology , Animals , Blood/metabolism , Brain/metabolism , Leukocyte Count , Male , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Solubility
13.
J Cereb Blood Flow Metab ; 12(5): 802-8, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1506444

ABSTRACT

In a rat model of complete global brain ischemia (neck tourniquet) lasting either 3 min or 20 min, we monitored global CBF (sagittal sinus H2 clearance) and CMRO2 for 6 h to test the hypothesis that delayed postischemic hyperemia and uncoupling of CBF and CMRO2 occur depending on the severity of the insult. Early postischemic hyperemia occurred in both the 3-min and 20-min groups (p less than 0.05 vs. baseline values) and resolved by 15 min. Hypoperfusion occurred in the 3-min group between 15 and 60 min postischemia (approximately 23% reduction), and in the 20-min group from 15 to 120 min postischemia (approximately 50% reduction) (p less than 0.05), and then resolved. CMRO2 was not significantly different from baseline at any time after ischemia in the 3-min group. After 20 min of ischemia, however, CMRO2 was decreased (approximately 60%) throughout the postischemic period (p less than 0.05). At 5 min after ischemia, CBF/CMRO2 was increased in both groups but returned to baseline from 60 to 120 min postischemia. In the 3-min group, CBF/CMRO2 remained at baseline throughout the rest of the experiment. However, in the 20-min group, CBF/CMRO2 once again increased (approximately 100%), reaching a significant level at 180 min and remaining so for the rest of the 6-h period (p less than 0.05). These data demonstrate biphasic uncoupling of CBF and CMRO2 after severe (20 min) global ischemia in rats. This relatively early reemergence of CBF/CMRO2 uncoupling after 180 min of reperfusion is similar to that observed after prolonged cardiac arrest and resuscitation in humans.


Subject(s)
Brain Ischemia/physiopathology , Brain/metabolism , Cerebrovascular Circulation/physiology , Animals , Brain/physiopathology , Brain Ischemia/complications , Brain Ischemia/metabolism , Hyperemia/etiology , Hyperemia/metabolism , Male , Rats , Rats, Inbred Strains , Time Factors
14.
J Cereb Blood Flow Metab ; 16(2): 244-52, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8594056

ABSTRACT

Hypothermia is beneficial in adult models of traumatic brain injury (TBI), but it has not been evaluated in an immature animal model. We hypothesized that brief hypothermia applied after TBI would reduce cerebral edema and lesion volume in immature rats. Male Wistar rats (3-4 weeks of age, 90-140 g) were anesthetized, intubated, mechanically ventilated, and subjected to TBI by weight drop onto the exposed right parietal cortex. Hypothermic rats were then cooled to a brain temperature of 32.0 +/- 0.5 degrees C for 4 h, and control rats were maintained at a brain temperature of 37.0 +/- 0.5 degrees C. Cerebral edema (wet - dry weight method) was assessed at 5 days. At 4 h, a reduction of percent brain water in the traumatized hemisphere was observed in hypothermic versus normothermic rats (81.75 +/- 0.60 vs. 82.53 +/- 0.67%; p<0.05), but by 24 h posttrauma, the groups were similar (p = 0.82). Total lesion volume (47.2 +/- 8.5 vs. 44.4 +/- 10.0 mm3; p = 0.51) and necrotic volume (20.2 +/- 6.3 vs. 20.0 +/- 7.9 mm3; p = 0.95) were similar in the hypothermic and normothermic groups. We conclude that in this model, a transient (4-h) application of moderate (32 degrees C) hypothermia reduces the cerebral edema characteristically seen in immature rats at 4 h, but this reduction is not sustained at 24 h. Attenuating or delaying the development of cerebral edema could have important therapeutic relevance after TBI. Transient hypothermia, however, did not reduce lesion volume at 5 days posttrauma.


Subject(s)
Aging , Brain Injuries/therapy , Hypothermia, Induced , Animals , Body Temperature , Brain/pathology , Brain Edema/therapy , Brain Injuries/pathology , Male , Necrosis , Rats , Rats, Wistar
15.
J Cereb Blood Flow Metab ; 16(2): 253-61, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8594057

ABSTRACT

The effect of posttraumatic hypothermia (brain temperature controlled at 32 degrees C for 4 h) on mortality after severe controlled cortical impact (CCI) was studied in rats. Four posttraumatic brain temperatures were compared: 37 degrees C (n = 10), 36 degrees C (n = 4), 32 degrees C (n = 10), and uncontrolled (UC; n = 6). Rats were anesthetized and subjected to severe CCI (4.0-m/s velocity, 3.0-mm depth) to the exposed left parietal cortex. At 10 min posttrauma the rats were cooled or maintained at their target brain temperature, using external cooling or warming. Brain temperature in the UC group was recorded but not regulated, and rectal temperature was maintained at 37 +/- 0.5 degrees C. After 4 h, rats were rewarmed over a 1-h period to 37 degrees C, extubated, and observed for 24 h. In the 37 and 36 degree C groups, 24-h mortality was 50% (37 degrees C = 5/10, 36 degrees C = 2/4). In the 32 degree C group, 24-h mortality was 10% (1/10). In the UC group, brain temperature was 35.4 +/- 0.6 degrees C during the 4-h treatment period and 24-h mortality was 0% (0/6). Mortality was higher in groups with brain temperatures > or = 36 degrees C versus those with brain temperatures < 36 degrees C (50 vs. 6%, respectively; p < 0.05). Additionally, electroencephalograms (EEG) were recorded in subsets of each temperature group and the percentage of time that the EEG was suppressed (isoelectric) was determined. Percentage of EEG suppression was greater in the hypothermic (32 degrees C, n = 6; UC, n = 4) groups than in the normothermic (36 degrees C, n = 3; 37 degrees C, n = 6) groups (23.3 +/- 14.3 vs. 1.2 +/- 3.1%, respectively; p < 0.05). Posttraumatic hypothermia suppressed EEG during treatment and reduced mortality after severe CCI. The threshold for this protective effect appears to be a brain temperature < 36 degrees C. Thus, even mild hypothermia may be beneficial after severe brain trauma.


Subject(s)
Brain Injuries/therapy , Hypothermia, Induced , Parietal Lobe/injuries , Animals , Body Temperature , Brain Injuries/physiopathology , Electroencephalography , Male , Rats , Rats, Sprague-Dawley , Seizures/etiology , Seizures/physiopathology
16.
J Cereb Blood Flow Metab ; 17(8): 865-74, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9290584

ABSTRACT

We measured CBF and CO2 reactivity after traumatic brain injury (TBI) produced by controlled cortical impact (CCI) using magnetic resonance imaging (MRI) and spin-labeled carotid artery water protons as an endogenous tracer. Fourteen Sprague-Dawley rats divided into TBI (CCI; 4.02 +/- 0.14 m/s velocity; 2.5 mm deformation), sham, and control groups were studied 24 hours after TBI or surgery. Perfusion maps were generated during normocarbia (Paco2 30 to 40 mm Hg) and hypocarbia (PaCO2 15 to 25 mm Hg). During normocarbia, CBF was reduced within a cortical region of interest (ROI, injured versus contralateral) after TBI (200 +/- 82 versus 296 +/- 65 mL.100 g-1.min-1, P < 0.05). Within a contusion-enriched ROI, CBF was reduced after TBI (142 +/- 73 versus 280 +/- 64 mL.100 g-1.min-1, P < 0.05). Cerebral blood flow in the sham group was modestly reduced (212 +/- 112 versus 262 +/- 118 mL.100 g-1.min-1, P < 0.05). Also, TBI widened the distribution of CBF in injured and contralateral cortex. Hypocarbia reduced cortical CBF in control (48%), sham (45%), and TBI rats (48%) versus normocarbia, P < 0.05. In the contusion-enriched ROI, only controls showed a significant reduction in CBF, suggesting blunted CO2 reactivity in the sham and TBI group. CO2 reactivity was reduced in the sham (13%) and TBI (30%) groups within the cortical ROI (versus contralateral cortex). These values were increased twofold within the contusion-enriched ROI but were not statistically significant. After TBI, hypocarbia narrowed the CBF distribution in the injured cortex. We conclude that perfusion MRI using arterial spin-labeling is feasible for the serial, noninvasive measurement of CBF and CO2 reactivity in rats.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/physiopathology , Carbon Dioxide/metabolism , Cerebrovascular Circulation , Animals , Brain Injuries/pathology , Magnetic Resonance Imaging , Male , Perfusion , Rats , Rats, Sprague-Dawley , Spin Labels
17.
J Cereb Blood Flow Metab ; 19(8): 835-42, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10458590

ABSTRACT

Poly(ADP-ribose) polymerase (PARP), or poly-(ADP-ribose) synthetase, is a nuclear enzyme that consumes NAD when activated by DNA damage. The role of PARP in the pathogenesis of traumatic brain injury (TBI) is unknown. Using a controlled cortical impact (CCI) model of TBI and mice deficient in PARP, the authors studied the effect of PARP on functional and histologic outcome after CCI using two protocols. In protocol 1, naive mice (n = 7 +/+, n = 6 -/-) were evaluated for motor and memory acquisition before CCI. Mice were then subjected to severe CCI and killed at 24 hours for immunohistochemical detection of nitrated tyrosine, an indicator of peroxynitrite formation. Motor and memory performance did not differ between naive PARP +/+ and -/- mice. Both groups showed nitrotyrosine staining in the contusion, suggest ing that peroxynitrite is produced in contused brain. In protoco 2, mice (PARP +/+, n = 8; PARP -/-, n = 10) subjected to CCI were tested for motor and memory function, and contusion volume was determined by image analysis. PARP -/- mice demonstrated improved motor and memory function after CC versus PARP +/+ mice (P < 0.05). However, contusion volume was not different between groups. The results suggest a detri mental effect of PARP on functional outcome after TBI.


Subject(s)
Brain Injuries/physiopathology , Brain Injuries/psychology , Brain/physiopathology , Cognition/physiology , Maze Learning , Memory/physiology , Motor Activity/physiology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Animals , Brain/enzymology , Brain/pathology , Brain Injuries/genetics , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Cues , Immunohistochemistry , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/deficiency , Space Perception
18.
J Cereb Blood Flow Metab ; 18(6): 610-5, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626184

ABSTRACT

We tested the hypothesis that quinolinic acid, a tryptophan-derived N-methyl-D-aspartate agonist produced by macrophages and microglia, would be increased in CSF after severe traumatic brain injury (TBI) in humans, and that this increase would be associated with outcome. We also sought to determine whether therapeutic hypothermia reduced CSF quinolinic acid after injury. Samples of CSF (n = 230) were collected from ventricular catheters in 39 patients (16 to 73 years old) during the first week after TBI, (Glasgow Coma Scale [GCS] < 8). As part of an ongoing study, patients were randomized within 6 hours after injury to either hypothermia (32 degrees C) or normothermia (37 degrees C) treatments for 24 hours. Otherwise, patients received standard neurointensive care. Quinolinic acid was measured by mass spectrometry. Univariate and multivariate analyses were used to compare CSF quinolinic acid concentrations with age, gender, GCS, time after injury, mortality, and treatment (hypothermia versus normothermia). Quinolinic acid concentration in CSF increased maximally to 463 +/- 128 nmol/L (mean +/- SEM) at 72 to 83 hours after TBI. Normal values for quinolinic acid concentration in CSF are less than 50 nmol/L. Quinolinic acid concentration was increased 5- to 50-fold in many patients. There was a powerful association between time after TBI and increased quinolinic acid (P < 0.00001), and quinolinic acid was higher in patients who died than in survivors (P = 0.003). Age, gender, GCS, and treatment (32 degrees C versus 37 degrees C) did not correlate with CSF quinolinic acid. These data reveal a large increase in quinolinic acid concentration in CSF after TBI in humans and raise the possibility that this macrophage-derived excitotoxin may contribute to secondary damage.


Subject(s)
Brain Injuries/cerebrospinal fluid , Brain Injuries/mortality , Quinolinic Acid/cerebrospinal fluid , Adolescent , Adult , Aged , Brain Injuries/therapy , Humans , Hypothermia, Induced , Kinetics , Middle Aged , Prognosis , Regression Analysis
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.
Keio J Med ; 45(3): 248-52; discussion 252-3, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8897768

ABSTRACT

In order to determine the effect of depleting circulating polymorphonuclear neutrophils (PMN's) on brain microcirculation and lesion size in an acute stroke model, Spontaneously Hypertensive Rats (SHR) were injected intraperitoneally with either 2 ml RP-3 antineutrophil antibody followed in 4 hours by MCAO (n = 5), 2 ml saline followed in 4 hours by middle cerebral artery occlusion (MCAO) (n = 6), or 2 ml saline followed in 4 hours by sham operation (n = 3). After 4 hours of ischemia or a 4 hour interval (sham-operated animals), microvascular perfusion was assessed by means of an intravascular fluorescent tracer technique: FITC-dextran and Evans blue were injected intravenously 10 seconds and 5 seconds, respectively, before decapitation. Lesion volume was calculated by interpolation from histologic sections cut from 8 predefined stereotactic levels. MCAO with the normal complement of neutrophils led to significant impairment of perfusion in nutrient vessels and a maximal ischemic lesion volume. Depletion of circulating leukocytes by RP-3 significantly attenuated the microvessel perfusion impairment and reduced the volume of ischemic brain injury.


Subject(s)
Cerebrovascular Disorders/physiopathology , Hypertension/physiopathology , Microcirculation/physiology , Neutrophils/cytology , Animals , Cerebrovascular Disorders/blood , Cerebrovascular Disorders/complications , Hypertension/blood , Hypertension/complications , Leukocyte Count , Male , Rats , Rats, Inbred SHR
SELECTION OF CITATIONS
SEARCH DETAIL