1
|
Valero-Hernandez E, Tremoleda JL, Michael-Titus AT. Omega-3 Fatty Acids and Traumatic Injury in the Adult and Immature Brain. Nutrients 2024; 16:4175. [PMID: 39683568 DOI: 10.3390/nu16234175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Traumatic brain injury (TBI) can lead to substantial disability and health loss. Despite its importance and impact worldwide, no treatment options are currently available to help protect or preserve brain structure and function following injury. In this review, we discuss the potential benefits of using omega-3 polyunsaturated fatty acids (O3 PUFAs) as therapeutic agents in the context of TBI in the paediatric and adult populations. Methods: Preclinical and clinical research reports investigating the effects of O3 PUFA-based interventions on the consequences of TBI were retrieved and reviewed, and the evidence presented and discussed. Results: A range of animal models of TBI, types of injury, and O3 PUFA dosing regimens and administration protocols have been used in different strategies to investigate the effects of O3 PUFAs in TBI. Most evidence comes from preclinical studies, with limited clinical data available thus far. Overall, research indicates that high O3 PUFA levels help lessen the harmful effects of TBI by reducing tissue damage and cell loss, decreasing associated neuroinflammation and the immune response, which in turn moderates the severity of the associated neurological dysfunction. Conclusions: Data from the studies reviewed here indicate that O3 PUFAs could substantially alleviate the impact of traumatic injuries in the central nervous system, protect structure and help restore function in both the immature and adult brains.
Collapse
Affiliation(s)
- Ester Valero-Hernandez
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Jordi L Tremoleda
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
2
|
Chen Y, Luan P, Liu J, Wei Y, Wang C, Wu R, Wu Z, Jing M. Spatiotemporally selective astrocytic ATP dynamics encode injury information sensed by microglia following brain injury in mice. Nat Neurosci 2024; 27:1522-1533. [PMID: 38862791 DOI: 10.1038/s41593-024-01680-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 05/13/2024] [Indexed: 06/13/2024]
Abstract
Injuries to the brain result in tunable cell responses paired with stimulus properties, suggesting the existence of intrinsic processes that encode and transmit injury information; however, the molecular mechanism of injury information encoding is unclear. Here, using ATP fluorescent indicators, we identify injury-evoked spatiotemporally selective ATP dynamics, Inflares, in adult mice of both sexes. Inflares are actively released from astrocytes and act as the internal representations of injury. Inflares encode injury intensity and position at their population level through frequency changes and are further decoded by microglia, driving changes in their activation state. Mismatches between Inflares and injury severity lead to microglia dysfunction and worsening of injury outcome. Blocking Inflares in ischemic stroke in mice reduces secondary damage and improves recovery of function. Our results suggest that astrocytic ATP dynamics encode injury information and are sensed by microglia.
Collapse
Affiliation(s)
- Yue Chen
- Chinese Institute for Brain Research, Beijing, China
| | - Pengwei Luan
- Chinese Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Juan Liu
- Chinese Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yelan Wei
- Chinese Institute for Brain Research, Beijing, China
- Department of College of Physical Education and Sport, Beijing Normal University, Beijing, China
| | - Chenyu Wang
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Basic Medical Sciences, Beijing, China
| | - Rui Wu
- Chinese Institute for Brain Research, Beijing, China
- China Agricultural University, Beijing, China
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Miao Jing
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
3
|
Seeger DR, Golovko SA, Golovko MY. Blood-Brain Barrier Is the Major Site for a Rapid and Dramatic Prostanoid Increase upon Brain Global Ischemia. Lipids 2019; 55:79-85. [PMID: 31814137 DOI: 10.1002/lipd.12205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/12/2019] [Accepted: 11/15/2019] [Indexed: 12/17/2022]
Abstract
We and others have demonstrated a rapid and dramatic increase in brain prostanoids upon decapitation-induced brain global ischemia and injury. However, the mechanism for this induction, including the cell types involved, are unknown. In the present study, we have validated and applied a pharmacological approach to inhibit prostanoid synthesis in the blood-brain barrier including endothelial cells. Our results indicate that a nonspecific cyclooxygenase (COX) inhibitor, ketorolac, does not pass the blood-brain barrier and does not enter red blood cells but penetrates endothelial cells. Ketorolac treatment did not affect basal prostanoid levels but completely prevented prostanoid induction upon global ischemia. These data indicate that basal prostanoids are synthesized in brain parenchyma cells, while inducible prostanoids are synthesized in the blood-brain barrier, most likely in endothelial cells. However, future studies with cell and COX isoform-specific gene ablation are needed to further validate this conclusion. These findings identify endothelial cells as a possible target for the development of pharmacological approaches to selectively attenuate inducible prostanoid pools without affecting basal levels under brain ischemia, trauma, surgery, and other related conditions.
Collapse
Affiliation(s)
- Drew R Seeger
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, 1301 N. Columbia Rd., Grand Forks, ND, 58202-9037, USA
| | - Svetlana A Golovko
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, 1301 N. Columbia Rd., Grand Forks, ND, 58202-9037, USA
| | - Mikhail Y Golovko
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, 1301 N. Columbia Rd., Grand Forks, ND, 58202-9037, USA
| |
Collapse
|
4
|
Corrigan F, Mander KA, Leonard AV, Vink R. Neurogenic inflammation after traumatic brain injury and its potentiation of classical inflammation. J Neuroinflammation 2016; 13:264. [PMID: 27724914 PMCID: PMC5057243 DOI: 10.1186/s12974-016-0738-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/28/2016] [Indexed: 01/05/2023] Open
Abstract
Background The neuroinflammatory response following traumatic brain injury (TBI) is known to be a key secondary injury factor that can drive ongoing neuronal injury. Despite this, treatments that have targeted aspects of the inflammatory pathway have not shown significant efficacy in clinical trials. Main body We suggest that this may be because classical inflammation only represents part of the story, with activation of neurogenic inflammation potentially one of the key initiating inflammatory events following TBI. Indeed, evidence suggests that the transient receptor potential cation channels (TRP channels), TRPV1 and TRPA1, are polymodal receptors that are activated by a variety of stimuli associated with TBI, including mechanical shear stress, leading to the release of neuropeptides such as substance P (SP). SP augments many aspects of the classical inflammatory response via activation of microglia and astrocytes, degranulation of mast cells, and promoting leukocyte migration. Furthermore, SP may initiate the earliest changes seen in blood-brain barrier (BBB) permeability, namely the increased transcellular transport of plasma proteins via activation of caveolae. This is in line with reports that alterations in transcellular transport are seen first following TBI, prior to decreases in expression of tight-junction proteins such as claudin-5 and occludin. Indeed, the receptor for SP, the tachykinin NK1 receptor, is found in caveolae and its activation following TBI may allow influx of albumin and other plasma proteins which directly augment the inflammatory response by activating astrocytes and microglia. Conclusions As such, the neurogenic inflammatory response can exacerbate classical inflammation via a positive feedback loop, with classical inflammatory mediators such as bradykinin and prostaglandins then further stimulating TRP receptors. Accordingly, complete inhibition of neuroinflammation following TBI may require the inhibition of both classical and neurogenic inflammatory pathways.
Collapse
Affiliation(s)
- Frances Corrigan
- Adelaide Centre for Neuroscience Research, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Kimberley A Mander
- Adelaide Centre for Neuroscience Research, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anna V Leonard
- Adelaide Centre for Neuroscience Research, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Robert Vink
- Sansom Institute for Health Research, The University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
5
|
Abstract
Posttraumatic epilepsy (PTE) is one of the most common and devastating complications of traumatic brain injury (TBI). Currently, the etiopathology and mechanisms of PTE are poorly understood and as a result, there is no effective treatment or means to prevent it. Antiepileptic drugs remain common preventive strategies in the management of TBI to control acute posttraumatic seizures and to prevent the development of PTE, although their efficacy in the latter case is disputed. Different strategies of PTE prophylaxis have been showing promise in preclinical models, but their translation to the clinic still remains elusive due in part to the variability of these models and the fact they do not recapitulate all complex pathologies associated with human TBI. TBI is a multifaceted disorder reflected in several potentially epileptogenic alterations in the brain, including mechanical neuronal and vascular damage, parenchymal and subarachnoid hemorrhage, subsequent toxicity caused by iron-rich hemoglobin breakdown products, and energy disruption resulting in secondary injuries, including excitotoxicity, gliosis, and neuroinflammation, often coexisting to a different degree. Several in vivo models have been developed to reproduce the acute TBI cascade of events, to reflect its anatomical pathologies, and to replicate neurological deficits. Although acute and chronic recurrent posttraumatic seizures are well-recognized phenomena in these models, there is only a limited number of studies focused on PTE. The most used mechanical TBI models with documented electroencephalographic and behavioral seizures with remote epileptogenesis include fluid percussion, controlled cortical impact, and weight-drop. This chapter describes the most popular models of PTE-induced TBI models, focusing on the controlled cortical impact and the fluid percussion injury models, the methods of behavioral and electroencephalogram seizure assessments, and other approaches to detect epileptogenic properties, and discusses their potential application for translational research.
Collapse
|
6
|
Role of the prostaglandin E2 EP1 receptor in traumatic brain injury. PLoS One 2014; 9:e113689. [PMID: 25426930 PMCID: PMC4245217 DOI: 10.1371/journal.pone.0113689] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/27/2014] [Indexed: 11/19/2022] Open
Abstract
Brain injuries promote upregulation of so-called proinflammatory prostaglandins, notably prostaglandin E2 (PGE2), leading to overactivation of a class of its cognate G-protein-coupled receptors, including EP1, which is considered a promising target for treatment of ischemic stroke. However, the role of the EP1 receptor is complex and depends on the type of brain injury. This study is focused on the investigation of the role of the EP1 receptor in a controlled cortical impact (CCI) model, a preclinical model of traumatic brain injury (TBI). The therapeutic effects of post-treatments with a widely studied EP1 receptor antagonist, SC-51089, were examined in wildtype and EP1 receptor knockout C57BL/6 mice. Neurological deficit scores (NDS) were assessed 24 and 48 h following CCI or sham surgery, and brain immunohistochemical pathology was assessed 48 h after surgery. In wildtype mice, CCI resulted in an obvious cortical lesion and localized hippocampal edema with an associated significant increase in NDS compared to sham-operated animals. Post-treatments with the selective EP1 receptor antagonist SC-51089 or genetic knockout of EP1 receptor had no significant effects on cortical lesions and hippocampal swelling or on the NDS 24 and 48 h after CCI. Immunohistochemistry studies revealed CCI-induced gliosis and microglial activation in selected ipsilateral brain regions that were not affected by SC-51089 or in the EP1 receptor-deleted mice. This study provides further clarification on the respective contribution of the EP1 receptor in TBI and suggests that, under this experimental paradigm, the EP1 receptor would have limited effects in modulating acute neurological and anatomical pathologies following contusive brain trauma. Findings from this protocol, in combination with previous studies demonstrating differential roles of EP1 receptor in ischemic, neurotoxic, and hemorrhagic conditions, provide scientific background and further clarification of potential therapeutic application of prospective prostaglandin G-protein-coupled receptor drugs in the clinic for treatment of TBI and other acute brain injuries.
Collapse
|
7
|
Yu GX, Mueller M, Hawkins BE, Mathew BP, Parsley MA, Vergara LA, Hellmich HL, Prough DS, Dewitt DS. Traumatic brain injury in vivo and in vitro contributes to cerebral vascular dysfunction through impaired gap junction communication between vascular smooth muscle cells. J Neurotrauma 2014; 31:739-48. [PMID: 24341563 PMCID: PMC4047850 DOI: 10.1089/neu.2013.3187] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gap junctions (GJs) contribute to cerebral vasodilation, vasoconstriction, and, perhaps, to vascular compensatory mechanisms, such as autoregulation. To explore the effects of traumatic brain injury (TBI) on vascular GJ communication, we assessed GJ coupling in A7r5 vascular smooth muscle (VSM) cells subjected to rapid stretch injury (RSI) in vitro and VSM in middle cerebral arteries (MCAs) harvested from rats subjected to fluid percussion TBI in vivo. Intercellular communication was evaluated by measuring fluorescence recovery after photobleaching (FRAP). In VSM cells in vitro, FRAP increased significantly (p<0.05 vs. sham RSI) after mild RSI, but decreased significantly (p<0.05 vs. sham RSI) after moderate or severe RSI. FRAP decreased significantly (p<0.05 vs. sham RSI) 30 min and 2 h, but increased significantly (p<0.05 vs. sham RSI) 24 h after RSI. In MCAs harvested from rats 30 min after moderate TBI in vivo, FRAP was reduced significantly (p<0.05), compared to MCAs from rats after sham TBI. In VSM cells in vitro, pretreatment with the peroxynitrite (ONOO(-)) scavenger, 5,10,15,20-tetrakis(4-sulfonatophenyl)prophyrinato iron[III], prevented RSI-induced reductions in FRAP. In isolated MCAs from rats treated with the ONOO(-) scavenger, penicillamine, GJ coupling was not impaired by fluid percussion TBI. In addition, penicillamine treatment improved vasodilatory responses to reduced intravascular pressure in MCAs harvested from rats subjected to moderate fluid percussion TBI. These results indicate that TBI reduced GJ coupling in VSM cells in vitro and in vivo through mechanisms related to generation of the potent oxidant, ONOO(-).
Collapse
Affiliation(s)
- Guang-Xiang Yu
- Charles R. Allen Research Laboratories, Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Birnie M, Morrison R, Camara R, Strauss KI. Temporal changes of cytochrome P450 (Cyp) and eicosanoid-related gene expression in the rat brain after traumatic brain injury. BMC Genomics 2013; 14:303. [PMID: 23642095 PMCID: PMC3658912 DOI: 10.1186/1471-2164-14-303] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 04/16/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) induces arachidonic acid (ArA) release from cell membranes. ArA metabolites form a class of over 50 bioactive eicosanoids that can induce both adaptive and/or maladaptive brain responses. The dynamic metabolism of ArA to eicosanoids, and how they affect the injured brain, is poorly understood due to their diverse activities, trace levels, and short half-lives. The eicosanoids produced in the brain postinjury depend upon the enzymes present locally at any given time. Eicosanoids are synthesized by heme-containing enzymes, including cyclooxygenases, lipoxygenases, and arachidonate monoxygenases. The latter comprise a subset of the cytochrome P450 "Cyp" gene family that metabolize fatty acids, steroids, as well as endogenous and exogenous toxicants. However, for many of these genes neither baseline neuroanatomical nor injury-related temporal expression have been studied in the brain.In a rat model of parietal cortex TBI, Cyp and eicosanoid-related mRNA levels were determined at 6 h, 24 h, 3d, and 7d postinjury in parietal cortex and hippocampus, where dynamic changes in eicosanoids have been observed. Quantitative real-time polymerase chain reaction with low density arrays were used to assay 62 rat Cyps, 37 of which metabolize ArA or other unsaturated fatty acids; 16 eicosanoid-related enzymes that metabolize ArA or its metabolites; 8 eicosanoid receptors; 5 other inflammatory- and recovery-related genes, plus 2 mouse Cyps as negative controls and 3 highly expressed "housekeeping" genes. RESULTS Sixteen arachidonate monoxygenases, 17 eicosanoid-related genes, and 12 other Cyps were regulated in the brain postinjury (p < 0.05, Tukey HSD). Discrete tissue levels and distinct postinjury temporal patterns of gene expression were observed in hippocampus and parietal cortex. CONCLUSIONS The results suggest complex regulation of ArA and other lipid metabolism after TBI. Due to the temporal nature of brain injury-induced Cyp gene induction, manipulation of each gene (or its products) at a given time after TBI will be required to assess their contributions to secondary injury and/or recovery. Moreover, a better understanding of brain region localization and cell type-specific expression may be necessary to deduce the role of these eicosanoid-related genes in the healthy and injured brain.
Collapse
Affiliation(s)
- Matthew Birnie
- University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 515, 45267 Cincinnati, OH, USA
| | - Ryan Morrison
- University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 515, 45267 Cincinnati, OH, USA
| | - Ramatoulie Camara
- University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 515, 45267 Cincinnati, OH, USA
| | - Kenneth I Strauss
- University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 515, 45267 Cincinnati, OH, USA
- Present Address: Michigan State University College of Human Medicine, 333 Bostwick Ave NE, 49503 Grand Rapids, MI, USA
| |
Collapse
|
9
|
Wei EP, Hamm RJ, Baranova AI, Povlishock JT. The long-term microvascular and behavioral consequences of experimental traumatic brain injury after hypothermic intervention. J Neurotrauma 2009; 26:527-37. [PMID: 19245307 DOI: 10.1089/neu.2008.0797] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) has been demonstrated to induce cerebral vascular dysfunction that is reflected in altered responses to various vasodilators. While previous reports have focused primarily on the short-term vascular alterations, few have examined these vascular changes for more than 7 days, or have attempted to correlate these alterations with any persisting behavioral changes or potential therapeutic modulation. Accordingly, we evaluated the long-term microvascular and behavioral consequences of experimental TBI and their therapeutic modulation via hypothermia. In this study, one group was injured with no treatment, another group was injured and 1 h later was treated with 120 min of hypothermia followed by slow rewarming, and a third group was non-injured. Animals equipped with cranial windows for visualization of the pial microvasculature were challenged with various vasodilators, including acetylcholine, hypercapnia, adenosine, pinacidil, and sodium nitroprusside, at either 1 or 3 weeks post-TBI. In addition, all animals were tested for vestibulomotor tasks at 1 week post-TBI, and animals surviving for 3 weeks post-TBI were tested in a Morris water maze (MWM). The results of this investigation demonstrated that TBI resulted in long-term vascular dysfunction in terms of altered vascular reactivity to various vasodilators, which was significantly improved with the use of a delayed 120-min hypothermic treatment. In contrast, data from the MWM task indicated that injured animals revealed persistent deficits in the spatial memory test performance, with hypothermia exerting no protective effects. Collectively, these data illustrate that TBI can evoke long-standing brain vascular and spatial memory dysfunction that manifest different responses to hypothermic intervention. These findings further illustrate the complexity of TBI and highlight the fact that the chosen hypothermic intervention may not necessarily exert a global protective response.
Collapse
Affiliation(s)
- Enoch P Wei
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298-0709, USA
| | | | | | | |
Collapse
|
10
|
|
11
|
Strauss KI. Antiinflammatory and neuroprotective actions of COX2 inhibitors in the injured brain. Brain Behav Immun 2008; 22:285-98. [PMID: 17996418 PMCID: PMC2855502 DOI: 10.1016/j.bbi.2007.09.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Revised: 09/14/2007] [Accepted: 09/20/2007] [Indexed: 12/22/2022] Open
Abstract
Overexpression of COX2 appears to be both a marker and an effector of neural damage after a variety of acquired brain injuries, and in natural or pathological aging of the brain. COX2 inhibitors may be neuroprotective in the brain by reducing prostanoid and free radical synthesis, or by directing arachidonic acid down alternate metabolic pathways. The arachidonic acid shunting hypothesis proposes that COX2 inhibitors' neuroprotective effects may be mediated by increased formation of potentially beneficial eicosanoids. Under conditions where COX2 activity is inhibited, arachidonic acid accumulates or is converted to eicosanoids via lipoxygenases and cytochrome P450 (CYP) epoxygenases. Several P450 eicosanoids have been demonstrated to have beneficial effects in the brain and/or periphery. We suspect that arachidonic acid shunting may be as important to functional recovery after brain injuries as altered prostanoid formation per se. Thus, COX2 inhibition and arachidonic acid shunting have therapeutic implications beyond the suppression of prostaglandin synthesis and free radical formation.
Collapse
Affiliation(s)
- Kenneth I. Strauss
- Mayfield Neurotrauma Research Lab, Department of Neurosurgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML515, Cincinnati, OH 45267 ()
| |
Collapse
|
12
|
Cremer OL, Kalkman CJ. Cerebral pathophysiology and clinical neurology of hyperthermia in humans. PROGRESS IN BRAIN RESEARCH 2007; 162:153-69. [PMID: 17645919 DOI: 10.1016/s0079-6123(06)62009-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Deliberate hyperthermia has been used clinically as experimental therapy for neoplastic and infectious diseases. Several case fatalities have occurred with this form of treatment, but most were attributable to systemic complications rather than central nervous system toxicity. Nonetheless, demyelating peripheral neuropathy and neurological symptoms of nausea, delirium, apathy, stupor, and coma have been reported. Temperatures exceeding 40 degrees C cause transient vasoparalysis in humans, resulting in cerebral metabolic uncoupling and loss of pressure-flow autoregulation. These findings may be related to the development of brain edema, intracerebral hemorrhage, and intracranial hypertension observed after prolonged therapeutic hyperthermia. Furthermore, deliberate hyperthermia critically worsens the extent of histopathological damage in animal models of traumatic, ischemic, and hypoxic brain injury. However, it is unknown whether these findings translate to episodes of spontaneous fever in neurologically injured patients. In a clinical setting fever is a strong prognostic marker of a patient's primary degree of neuronal damage, and a causal relation with long-term functional neurological outcome has not been established for most types of brain injury. Furthermore, in the neurosurgical intensive-care unit fever is extremely common whereas antipyretic therapy is only poorly effective. Therefore maintaining strict normothermia may be an impossible goal in many patients. Although there are several physiological arguments for avoiding exogenous hyperthermia in neurologically injured patients, there is no evidence that aggressive attempts at controlling spontaneous fever can improve clinical outcome.
Collapse
Affiliation(s)
- Olaf L Cremer
- Department of Intensive Care Medicine, University Medical Center, Q04.460, PO Box 85500, 3508 GA Utrecht, The Netherlands.
| | | |
Collapse
|
13
|
Phillis JW, Horrocks LA, Farooqui AA. Cyclooxygenases, lipoxygenases, and epoxygenases in CNS: Their role and involvement in neurological disorders. ACTA ACUST UNITED AC 2006; 52:201-43. [PMID: 16647138 DOI: 10.1016/j.brainresrev.2006.02.002] [Citation(s) in RCA: 273] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2005] [Revised: 02/23/2006] [Accepted: 02/24/2006] [Indexed: 01/01/2023]
Abstract
Three enzyme systems, cyclooxygenases that generate prostaglandins, lipoxygenases that form hydroxy derivatives and leukotrienes, and epoxygenases that give rise to epoxyeicosatrienoic products, metabolize arachidonic acid after its release from neural membrane phospholipids by the action of phospholipase A(2). Lysophospholipids, the other products of phospholipase A(2) reactions, are either reacylated or metabolized to platelet-activating factor. Under normal conditions, these metabolites play important roles in synaptic function, cerebral blood flow regulation, apoptosis, angiogenesis, and gene expression. Increased activities of cyclooxygenases, lipoxygenases, and epoxygenases under pathological situations such as ischemia, epilepsy, Alzheimer's disease, Parkinson disease, amyotrophic lateral sclerosis, and Creutzfeldt-Jakob disease produce neuroinflammation involving vasodilation and vasoconstriction, platelet aggregation, leukocyte chemotaxis and release of cytokines, and oxidative stress. These are closely associated with the neural cell injury which occurs in these neurological conditions. The metabolic products of docosahexaenoic acid, through these enzymes, generate a new class of lipid mediators, namely docosatrienes and resolvins. These metabolites antagonize the effect of metabolites derived from arachidonic acid. Recent studies provide insight into how these arachidonic acid metabolites interact with each other and other bioactive mediators such as platelet-activating factor, endocannabinoids, and docosatrienes under normal and pathological conditions. Here, we review present knowledge of the functions of cyclooxygenases, lipoxygenases, and epoxygenases in brain and their association with neurodegenerative diseases.
Collapse
Affiliation(s)
- John W Phillis
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | | | | |
Collapse
|
14
|
Hwang SY, Jung JS, Kim TH, Lim SJ, Oh ES, Kim JY, Ji KA, Joe EH, Cho KH, Han IO. Ionizing radiation induces astrocyte gliosis through microglia activation. Neurobiol Dis 2006; 21:457-67. [PMID: 16202616 DOI: 10.1016/j.nbd.2005.08.006] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 08/03/2005] [Accepted: 08/16/2005] [Indexed: 12/23/2022] Open
Abstract
The aim of this study was to investigate the role of microglia in radiation-induced astrocyte gliosis. We found that a single dose of 15 Gy radiation to a whole rat brain increased immunostaining of glial fibrillary acidic protein in astrocytes 6 h later, and even more so 24 h later, indicating the initiation of gliosis. While irradiation of cultured rat astrocytes had little effect, irradiation of microglia-astrocyte mixed-cultures displayed altered astrocyte phenotype into more processed, which is another characteristic of gliosis. Experiments using microglia-conditioned media indicated this astrocyte change was due to factors released from irradiated microglia. Irradiation of cultured mouse microglial cells induced a dose-dependent increase in mRNA levels for cyclooxygenase-2 (COX-2), interleukin (IL)-1beta, IL-6, IL-18, tumor necrosis factor-alpha and interferon-gamma-inducible protein-10, which are usually associated with microglia activation. Consistent with these findings, irradiation of microglia activated NF-kappaB, a transcription factor that regulates microglial activation. Addition of prostaglandin E2 (PGE2: a metabolic product of the COX-2 enzyme) to primary cultured rat astrocytes resulted in phenotypic changes similar to those observed in mixed-culture experiments. Therefore, it appears that PGE(2) released from irradiated microglia is a key mediator of irradiation-induced gliosis or astrocyte phenotype change. These data suggest that radiation-induced microglial activation and resultant production of PGE2 seems to be associated with an underlying cause of inflammatory complications associated with radiation therapy for malignant gliomas.
Collapse
Affiliation(s)
- So-Young Hwang
- Research Institute, National Cancer Center, Goyang, Gyeonggi, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
The cerebrovascular endothelium exerts a profound influence on cerebral vessels and cerebral blood flow. This review summarizes current knowledge of various dilator and constrictor mechanisms intrinsic to the cerebrovascular endothelium. The endothelium contributes to the resting tone of cerebral arteries and arterioles by tonically releasing nitric oxide (NO•). Dilations can occur by stimulated release of NO•, endothelium-derived hyperpolarization factor, or prostanoids. During pathological conditions, the dilator influence of the endothelium can turn to that of constriction by a variety of mechanisms, including decreased NO• bioavailability and release of endothelin-1. The endothelium may participate in neurovascular coupling by conducting local dilations to upstream arteries. Further study of the cerebrovascular endothelium is critical for understanding the pathogenesis of a number of pathological conditions, including stroke, traumatic brain injury, and subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Jon Andresen
- Department of Anesthesiology, Baylor College of Medicine, One Baylor Plaza, Suite 434D, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
16
|
Moochhala SM, Lu J, Xing MCK, Anuar F, Ng KC, Yang KLS, Whiteman M, Atan S. Mercaptoethylguanidine Inhibition of Inducible Nitric Oxide Synthase and Cyclooxygenase-2 Expressions Induced in Rats After Fluid-Percussion Brain Injury. ACTA ACUST UNITED AC 2005; 59:450-7. [PMID: 16294091 DOI: 10.1097/01.ta.0000174858.79847.6d] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The present study examined the temporal expression of nitric oxide synthase (iNOS) and cyclo-oxygenase (COX)-2 in rat brains after traumatic brain injury (TBI). We studied the effects of mercaptoethylguanidine (MEG), a dual inhibitor of the inducible iNOS and COX with scavenging effect on peroxynitrite, on physiologic variables, brain pathogenesis, and neurologic performance in rats after a lateral fluid percussive-induced TBI. Mean arterial blood pressure and percentage cerebral tissue perfusion in MEG-treated TBI rats showed significant improvement when compared with TBI rats. Immunohistochemical analysis showed a marked number of iNOS and COX-2 immunopositive cells in the cerebral cortex ipsilateral to the injury in TBI rats when compared with MEG-treated TBI rats. MEG also significantly decreased the number of hyperchromatic and shrunken cortical neurons when compared with TBI rats' brain nitrate/nitrite, and prostaglandin E2 levels were attenuated in MEG-treated TBI rats when compared with TBI rats. It is therefore suggested that treatment of MEG via inhibition of iNOS and COX-2 might contribute to improved physiologic variables, neuronal cell survival, and neurologic outcome after TBI.
Collapse
|
17
|
Gopez JJ, Yue H, Vasudevan R, Malik AS, Fogelsanger LN, Lewis S, Panikashvili D, Shohami E, Jansen SA, Narayan RK, Strauss KI. Cyclooxygenase-2-specific inhibitor improves functional outcomes, provides neuroprotection, and reduces inflammation in a rat model of traumatic brain injury. Neurosurgery 2005; 56:590-604. [PMID: 15730585 PMCID: PMC1513642 DOI: 10.1227/01.neu.0000154060.14900.8f] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2004] [Accepted: 12/13/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Increases in brain cyclooxygenase-2 (COX2) are associated with the central inflammatory response and with delayed neuronal death, events that cause secondary insults after traumatic brain injury. A growing literature supports the benefit of COX2-specific inhibitors in treating brain injuries. METHODS DFU [5,5-dimethyl-3(3-fluorophenyl)-4(4-methylsulfonyl)phenyl-2(5)H)-furanone] is a third-generation, highly specific COX2 enzyme inhibitor. DFU treatments (1 or 10 mg/kg intraperitoneally, twice daily for 3 d) were initiated either before or after traumatic brain injury in a lateral cortical contusion rat model. RESULTS DFU treatments initiated 10 minutes before injury or up to 6 hours after injury enhanced functional recovery at 3 days compared with vehicle-treated controls. Significant improvements in neurological reflexes and memory were observed. DFU initiated 10 minutes before injury improved histopathology and altered eicosanoid profiles in the brain. DFU 1 mg/kg reduced the rise in prostaglandin E2 in the brain at 24 hours after injury. DFU 10 mg/kg attenuated injury-induced COX2 immunoreactivity in the cortex (24 and 72 h) and hippocampus (6 and 72 h). This treatment also decreased the total number of activated caspase-3-immunoreactive cells in the injured cortex and hippocampus, significantly reducing the number of activated caspase-3-immunoreactive neurons at 72 hours after injury. DFU 1 mg/kg amplified potentially anti-inflammatory epoxyeicosatrienoic acid levels by more than fourfold in the injured brain. DFU 10 mg/kg protected the levels of 2-arachidonoyl glycerol, a neuroprotective endocannabinoid, in the injured brain. CONCLUSION These improvements, particularly when treatment began up to 6 hours after injury, suggest exciting neuroprotective potential for COX2 inhibitors in the treatment of traumatic brain injury and support the consideration of Phase I/II clinical trials.
Collapse
Affiliation(s)
- Jonas J. Gopez
- Department of Neurosurgery, Temple University, School of Medicine, Philadelphia, Pennsylvania
| | - Hongfei Yue
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania
| | - Ram Vasudevan
- Department of Neurosurgery, Temple University, School of Medicine, Philadelphia, Pennsylvania
| | - Amir S. Malik
- Department of Neurosurgery, University of Texas, Houston Medical Center, Houston, Texas
| | - Lester N. Fogelsanger
- Department of Neurosurgery, Temple University, School of Medicine, Philadelphia, Pennsylvania
| | - Shawn Lewis
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Esther Shohami
- Department of Pharmacology, Hebrew University, Jerusalem, Israel
| | - Susan A. Jansen
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania
| | - Raj K. Narayan
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kenneth I. Strauss
- Reprint requests: Kenneth I. Strauss, Ph.D., Department of Neurosurgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML515, Cincinnati, OH 45267-0515.
| |
Collapse
|
18
|
Ahn MJ, Sherwood ER, Prough DS, Lin CY, DeWitt DS. The effects of traumatic brain injury on cerebral blood flow and brain tissue nitric oxide levels and cytokine expression. J Neurotrauma 2005; 21:1431-42. [PMID: 15672633 DOI: 10.1089/neu.2004.21.1431] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Adult, male, Sprague-Dawley rats were anesthetized, intubated, and mechanically ventilated with 1.5-2.0% isoflurane in oxygen (30%) and air. Rats were prepared for fluid percussion traumatic brain injury (TBI), laser Doppler flowmetry, and measurement of brain tissue nitric oxide (NO) levels using an ISO-NO electrode system. After preparation, isoflurane was reduced to 1.5%, and the rats were randomly assigned to receive sham (n = 6), moderate (1.9 atm, n = 6), or severe (2.8 atm, n = 6) parasagittal fluid percussion TBI. CBF and brain tissue NO levels were measured for 4 h, and then isoflurane levels were increased to 4.0% and the rats were decapitated and the brains were removed. Total RNA was isolated from rat brains and cytokine expression was determined. Laser Doppler flow velocity remained constant in the sham-injured rats but decreased significantly in rats subjected to moderate (p < 0.05) or severe (p < 0.05) TBI. Brain tissue NO levels remained constant in the sham-injured rats but decreased significantly (p < 0.01) after moderate TBI. Severe TBI produced slight, insignificant reductions in NO levels. Cytokine expression was very low in the shaminjured rats. TBI-induced expression of mRNAs for interleukin-1 alpha (IL-1alpha), IL-1beta, IL-6, and tumor necrosis factor-alpha (TNFa). IL-1alpha and IL-1beta mRNA expression increased significantly (p < 0.05 vs. sham-injury) after severe TBI and IL-6 and TNFa mRNA expression increased significant (p < 0.05 vs. sham-injury) after both moderate and severe TBI. Other cytokine mRNA expression was unchanged after TBI.
Collapse
Affiliation(s)
- Myung-Ja Ahn
- Charles R. Allen Research Laboratories, Department of Anesthesiology, The University of Texas Medical Branch, Galveston, Texas 77555-0830, USA
| | | | | | | | | |
Collapse
|
19
|
DeWitt DS, Prough DS. Traumatic Cerebral Vascular Injury: The Effects of Concussive Brain Injury on the Cerebral Vasculature. J Neurotrauma 2003; 20:795-825. [PMID: 14577860 DOI: 10.1089/089771503322385755] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In terms of human suffering, medical expenses, and lost productivity, head injury is one of the major health care problems in the United States, and inadequate cerebral blood flow is an important contributor to mortality and morbidity after traumatic brain injury. Despite the importance of cerebral vascular dysfunction in the pathophysiology of traumatic brain injury, the effects of trauma on the cerebral circulation have been less well studied than the effects of trauma on the brain. Recent research has led to a better understanding of the physiologic, cellular, and molecular components and causes of traumatic cerebral vascular injury. A more thorough understanding of the direct and indirect effects of trauma on the cerebral vasculature will lead to improvements in current treatments of brain trauma as well as to the development of novel and, hopefully, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Douglas S DeWitt
- Charles R. Allen Research Laboratories, Department of Anesthesiology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0830, USA.
| | | |
Collapse
|
20
|
Uzan M, Tanriverdi T, Aydin S, Gumustas K, Tureci E, Kuday C. The Cerebrospinal Fluid Thromboxane A2 and Prostaglandin I2 Levels in Patients With Severe Head Injury. ACTA ACUST UNITED AC 2003. [DOI: 10.1097/00013414-200306000-00001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Kyrkanides S, Moore AH, Olschowka JA, Daeschner JC, Williams JP, Hansen JT, Kerry O'Banion M. Cyclooxygenase-2 modulates brain inflammation-related gene expression in central nervous system radiation injury. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 104:159-69. [PMID: 12225870 DOI: 10.1016/s0169-328x(02)00353-4] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although the contribution of cyclooxygenase-2 (COX-2) to peripheral inflammation is well documented, little is known about its role in brain inflammation. For this purpose we studied COX-2 expression in the mouse brain following ionizing radiation in vivo, as well as in murine glial cell cultures in vitro. The possible role of COX-2 in modulating brain inflammation was examined utilizing NS-398, a COX-2 selective inhibitor. Our results indicate that COX-2 is significantly induced in astrocyte and microglial cultures by radiation injury as well as in brain. Increased levels of prostaglandin E(2) in irradiated brain were reduced by NS-398. Moreover, NS-398 administration significantly attenuated levels of induction for the majority of inflammatory mediators examined, including TNFalpha, IL-1beta, IL-6, iNOS, ICAM-1, and MMP-9. In contrast, the chemokines MIP-2 and MCP-1 showed enhanced levels of induction following NS-398 administration. These results indicate that COX-2 modulates the inflammatory response in brain following radiation injury, and suggest the use of COX-2 selective inhibitors for the management of CNS inflammation.
Collapse
Affiliation(s)
- Stephanos Kyrkanides
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Hurley SD, Olschowka JA, O'Banion MK. Cyclooxygenase inhibition as a strategy to ameliorate brain injury. J Neurotrauma 2002; 19:1-15. [PMID: 11852973 DOI: 10.1089/089771502753460196] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Cyclooxygenase (COX) is the obligate, rate-limiting enzyme for the conversion of arachidonic acid into prostaglandins. Two COX enzymes have been identified: a constitutively expressed COX-1 and an inducible, highly regulated COX-2. Widely used to treat chronic inflammatory disorders, COX inhibitors have shown promise in attenuating inflammation associated with brain injury. However, the use of COX inhibition in the treatment of brain injury has met with mixed success. This review summarizes our current understanding of COX expression in the central nervous system and the effects of COX inhibitors on brain injury. Three major targets for COX inhibition in the treatment brain injury have been identified. These are the cerebrovasculature, COX-2 expression by vulnerable neurons, and the neuroinflammatory response. Evidence suggests that given the right treatment paradigm, COX inhibition can influence each of these three targets. Drug interactions and general considerations for administrative paradigms are also discussed. Although therapies targeted to specific prostaglandin species, such as PGE2, might prove more ameliorative for brain injury, at the present time non-specific COX inhibitors and COX-2 specific inhibitors are readily available to researchers and clinicians. We believe that COX inhibition will be a useful, ameliorative adjunct in the treatment of most forms of brain injury.
Collapse
Affiliation(s)
- Sean D Hurley
- Department of Neurobiology and Anatomy, University of Rochester Medical Center, New York 14642, USA
| | | | | |
Collapse
|
23
|
Dhillon HS, Dong GX, Yurek DM, Estus S, Rangnekar VM, Dendle P, Prasad RM. Regional expression of Par-4 mRNA and protein after fluid percussion brain injury in the rat. Exp Neurol 2001; 170:140-8. [PMID: 11421591 DOI: 10.1006/exnr.2001.7685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regional levels of prostate apoptosis response-4 (Par-4) protein and mRNA were measured after lateral fluid percussion (FP) brain injury in rats. Immunochemical studies indicated that Par-4 immunoreactivity (ir) is present in cortical neurons and hippocampal CA1-CA3 pyramidal neurons in uninjured rats. Increases of Par-4-ir were observed in the CA3 neurons of the ipsilateral hippocampus (IH), but not in injured left cortex (IC) at 48 h after FP brain injury. Levels of the Par-4 mRNA measured by RT-PCR assay and protein measured by Western blot procedure were significantly increased in the injured IC and IH, but not in the contralateral right cortex and hippocampus after brain injury. Levels of both Par-4 protein and mRNA were significantly increased in the IC and IH as early as 2 h and stayed elevated at 24 and 48 h after injury. These data show that the induction of proapoptotic Par-4 mRNA and protein occurs only in the IC and IH that have been observed to undergo apoptosis and neuronal cell loss after lateral FP brain injury. Because increased expression of Par-4 has been observed to contribute to apoptosis and cell death in cultured neurons, the present temporal pattern of Par-4 expression is consistent with a role for Par-4 in apoptosis and neuronal cell death after traumatic brain injury.
Collapse
Affiliation(s)
- H S Dhillon
- Department of Surgery, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Dong GX, Singh DK, Dendle P, Prasad RM. Regional expression of Bcl-2 mRNA and mitochondrial cytochrome c release after experimental brain injury in the rat. Brain Res 2001; 903:45-52. [PMID: 11382386 DOI: 10.1016/s0006-8993(01)02379-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Regional levels of anti-apoptotic Bcl-2 mRNA and the cytosolic cytochrome c protein were measured after lateral fluid percussion (FP) brain injury in rats. Levels of Bcl-2 mRNA were significantly decreased in the injured left cortex (IC) and ipsilateral hippocampus (IH), but not in the contralateral right cortex (CC) and hippocampus (CH) after brain injury. Levels of Bcl-2 mRNA were significantly decreased as early as 2 h and stayed decreased as long as 48 h in the IC and IH after injury. Levels of the cytosolic cytochrome c protein were significantly increased in the IC and IH, but not in the CC and CH after brain injury. Levels of cytosolic cytochrome c were significantly increased in the IC at 30 min, 48 and 72 h, and in the IH at 2 h and as long as 72 h after injury. The increase of cytosolic cytochrome c suggests that the mitochondrial release of cytochrome is increased in the IC and IH after lateral FP brain injury. These data show that the reduction of anti-apoptotic Bcl-2 and increases of mitochondrial release of cytochrome c protein occur only in the IC and IH, regions which have been observed to undergo apoptosis and neuronal cell loss after lateral FP brain injury. Therefore, it is likely that the reduction of Bcl-2 and the increased cytochrome c protein in the cytosol contribute to the observed apoptosis and neuronal cell death in the IC and IH after lateral FP brain injury in rats.
Collapse
Affiliation(s)
- G X Dong
- Division of Neurosurgery, Department of Surgery and Sanders Brown Center on Aging, University of Kentucky Chandler Medical Center, Lexington, KY 40536-0084, USA
| | | | | | | |
Collapse
|
25
|
|
26
|
DeWitt DS, Prough DS. Should pressors be used to augument cerebral blood flow after traumatic brain injury? Crit Care Med 2000; 28:3933-4. [PMID: 11153639 DOI: 10.1097/00003246-200012000-00036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Armstead WM. Role of altered cyclooxygenase metabolism in impaired cerebrovasodilation to nociceptin/orphanin FQ following brain injury. Brain Res Bull 2000; 53:807-12. [PMID: 11179847 DOI: 10.1016/s0361-9230(00)00417-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study was designed to determine the role of altered cyclooxygenase metabolism in impaired pial artery dilation to the newly described opioid, nociceptin orphanin FQ (NOC/oFQ), following fluid percussion brain injury (FPI) in newborn pigs equipped with a closed cranial window. Recent studies show that NOC/oFQ contributes to oxygen free radical generation observed post FPI in a cyclooxygenase dependent manner. FPI was produced by using a pendulum to strike a piston on a saline filled cylinder that was fluid coupled to the brain via a hollow screw inserted through the cranium. NOC/oFQ (10(-8), 10(-6) M) modestly increased cerebrospinal fluid (CSF) 6-keto-PGF(1alpha), and thromboxane B(2) (TXB(2)), the stable breakdown products of PGI(2) and TXA(2), in sham animals (1148 +/- 83 to 1681 +/- 114 and 308 +/- 16 to 424 +/- 21 pg/ml for control and 10(-6) M NOC/oFQ 6-keto-PGF(1alpha), and TXB(2), respectively). In 1-h post FPI animals, basal levels of 6-keto-PGF(1alpha), and TXB(2) were elevated. NOC/oFQ stimulated release of 6-keto-PGF(1alpha), was blocked while such release of TXB(2) was enhanced (720 +/- 63 to 1446 +/- 117 pg/ml for control and 10(-6) M NOC/oFQ CSF TXB(2)). NOC/oFQ (10(-8), 10(-6) M) induced pial artery dilation that was reversed to vasoconstriction by FPI while the cyclooxygenase inhibitor indomethacin (5 mg/kg, intravenous) partially restored such vascular responses (8 +/- 1 and 15 +/- 1 vs. -7 +/- 1 and -12 +/- 1 vs. 7 +/- 1 and 12 +/- 1% for 10(-8), 10(-6) M NOC/oFQ in sham, FPI and FPI-Indo pretreated animals). Similar observations were made in FPI animals pretreated with the thromboxane receptor antagonist SQ 29,548 or the free radical scavenger polyethylene glycol superoxide dismutase and catalase. These data indicate that altered NOC/oFQ induced cyclooxygenase metabolism contributes to impairment of dilation to this opioid following FPI.
Collapse
Affiliation(s)
- W M Armstead
- Department of Anesthesia, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Aibiki M, Maekawa S, Yokono S. Moderate hypothermia improves imbalances of thromboxane A2 and prostaglandin I2 production after traumatic brain injury in humans. Crit Care Med 2000; 28:3902-6. [PMID: 11153633 DOI: 10.1097/00003246-200012000-00029] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To examine the levels of thromboxane B2 (TXB2) and 6-keto prostaglandin F1alpha (6-keto PGF1alpha) production in arterial and internal jugular bulb sera in patients with traumatic brain injury (TBI). TBI is associated with arachidonate release and may be associated with an imbalance of vasoconstricting and vasodilating cyclooxygenase metabolites. DESIGN A prospective, randomized study. SETTING The intensive care unit of a medical university hospital. INTERVENTIONS Twenty-six ventilated TBI patents (Glasgow Coma Scale score on admission, < or = 8 points) were divided randomly into two groups: a hypothermic group (n = 15), in which the patients were cooled to 32 to 33 degrees C after being giving vecuronium, midazolam, and buprenorphine; and a normothermic group (n = 11), in which the patients' body temperature was controlled at 36 to 37 degrees C by surface cooling using the same treatment as the hypothermic group. Body temperature control including normothermia was started 3 to 4 hrs after injury. The duration of hypothermia usually lasted for 3 to 4 days, after which the patients were rewarmed at a rate of approximately 1 C per day. MEASUREMENTS AND MAIN RESULTS Blood sampling for TXB2 and 6-keto PGF1alpha was started shortly after admission in both groups. Arterial TXB2 levels on admission in both groups were elevated remarkably, but not 6-keto PGF1alpha, thereby causing an imbalance of the prostanoids after injury. In the normothermic group, TXB2 decreased transiently, but this prostanoid increased again 3 days after the injury. In the hypothermic group, such prostanoid differences disappeared shortly after therapy, and the condition was sustained for 10 days. Hypothermia attenuated differences in TXB2 levels between arterial and internal jugular bulb sera, which may reflect reduced cerebral prostanoid production. The Glasgow Outcome Scale score 6 months after the insult in the hypothermic group was significantly higher than that in the normothermic group (p = .04). CONCLUSION The current results from a limited number of patients suggest that moderate hypothermia may reduce prostanoid production after TBI, thereby attenuating an imbalance of thromboxane A2 and prostaglandin I2. However, it must be clarified whether the changes in the prostanoid after moderate hypothermia are a secondary effect of other mediator changes or whether they simply represent an epiphenomenon that is mechanistically unrelated to damage in TBI.
Collapse
Affiliation(s)
- M Aibiki
- Intensive Care Unit, Kagawa Medical University Hospital, Japan
| | | | | |
Collapse
|
29
|
Strauss KI, Barbe MF, Marshall RM, Raghupathi R, Mehta S, Narayan RK. Prolonged cyclooxygenase-2 induction in neurons and glia following traumatic brain injury in the rat. J Neurotrauma 2000; 17:695-711. [PMID: 10972245 PMCID: PMC1456323 DOI: 10.1089/089771500415436] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cyclooxygenase-2 (COX2) is a primary inflammatory mediator that converts arachidonic acid into precursors of vasoactive prostaglandins, producing reactive oxygen species in the process. Under normal conditions COX2 is not detectable, except at low abundance in the brain. This study demonstrates a distinctive pattern of COX2 increases in the brain over time following traumatic brain injury (TBI). Quantitative lysate ribonuclease protection assays indicate acute and sustained increases in COX2 mRNA in two rat models of TBI. In the lateral fluid percussion model, COX2 mRNA is significantly elevated (>twofold, p < 0.05, Dunnett) at 1 day postinjury in the injured cortex and bilaterally in the hippocampus, compared to sham-injured controls. In the lateral cortical impact model (LCI), COX2 mRNA peaks around 6 h postinjury in the ipsilateral cerebral cortex (fivefold induction, p < 0.05, Dunnett) and in the ipsilateral and contralateral hippocampus (two- and six-fold induction, respectively, p < 0.05, Dunnett). Increases are sustained out to 3 days postinjury in the injured cortex in both models. Further analyses use the LCI model to evaluate COX2 induction. Immunoblot analyses confirm increased levels of COX2 protein in the cortex and hippocampus. Profound increases in COX2 protein are observed in the cortex at 1-3 days, that return to sham levels by 7 days postinjury (p < 0.05, Dunnett). The cellular pattern of COX2 induction following TBI has been characterized using immunohistochemistry. COX2-immunoreactivity (-ir) rises acutely (cell numbers and intensity) and remains elevated for several days following TBI. Increases in COX2-ir colocalize with neurons (MAP2-ir) and glia (GFAP-ir). Increases in COX2-ir are observed in cerebral cortex and hippocampus, ipsilateral and contralateral to injury as early as 2 h postinjury. Neurons in the ipsilateral parietal, perirhinal and piriform cortex become intensely COX2-ir from 2 h to at least 3 days postinjury. In agreement with the mRNA and immunoblot results, COX2-ir appears greatest in the contralateral hippocampus. Hippocampal COX2-ir progresses from the pyramidal cell layer of the CA1 and CA2 region at 2 h, to the CA3 pyramidal cells and dentate polymorphic and granule cell layers by 24 h postinjury. These increases are distinct from those observed following inflammatory challenge, and correspond to brain areas previously identified with the neurological and cognitive deficits associated with TBI. While COX2 induction following TBI may result in selective beneficial responses, chronic COX2 production may contribute to free radical mediated cellular damage, vascular dysfunction, and alterations in cellular metabolism. These may cause secondary injuries to the brain that promote neuropathology and worsen behavioral outcome.
Collapse
Affiliation(s)
- K I Strauss
- Department of Neurosurgery, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Natale JE, Joseph JG, Helfaer MA, Shaffner DH. Early hyperthermia after traumatic brain injury in children: risk factors, influence on length of stay, and effect on short-term neurologic status. Crit Care Med 2000; 28:2608-15. [PMID: 10921603 DOI: 10.1097/00003246-200007000-00071] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES a) To determine the risk factors for early hyperthermia after traumatic brain injury in children; b) to identify the contribution of early hyperthermia to neurologic status at pediatric intensive care unit (PICU) discharge and to PICU length of stay in head-injured children. STUDY DESIGN Observational cohort study. SETTING PICU at a tertiary care, university medical center. PATIENTS Children (n = 117) admitted to a PICU from July 1995 to May 1997 with traumatic brain injury. These children had a median age of 5.4 yrs (3 wks to 15.2 yrs old), and 33.4% were girls. MEASUREMENTS AND MAIN RESULTS Early hyperthermia (temperature >38.5 degrees C within the first 24 hrs of admission) occurred in 29.9% of patients admitted to the PICU with traumatic brain injury. Risk factors predicting early hyperthermia included Glasgow Coma Scale score in the emergency department < or =8, pediatric trauma score < or =8, cerebral edema or diffuse axonal injury on initial head computed tomography scan, admission blood glucose >150 mg/dL (8.2 mmol/L), admission white cell count >14,300 cells/mm3 (14.3 x 10(9) cells/L), and systolic hypotension. The presence of early hyperthermia significantly increased the risk for Glasgow Coma Scale score <13 at PICU discharge (odds ratio [OR] 9.7, 95% confidence interval [CI] 2.8, 24.4) and PICU stay > or =3 days (OR 13.8, CI 5.1, 37.5). When we used multiple logistic regression models including injury severity and hypotension, early hyperthermia remained an independent predictor of lower Glasgow Coma Scale score at PICU discharge (OR 4.7, CI 1.4, 15.6) and longer PICU length of stay (OR 8.5, CI 2.8, 25.6). CONCLUSIONS Early hyperthermia is independently associated with a measure of early neurologic status and resource utilization in children with traumatic brain injury serious enough to require PICU admission. These results support the prevention of hyperthermia in the management of traumatic brain injury in children. Further research is required to understand the mechanisms of this response and to identify appropriate preventive or therapeutic interventions.
Collapse
Affiliation(s)
- J E Natale
- Pediatric Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
31
|
Dash PK, Mach SA, Moore AN. Regional expression and role of cyclooxygenase-2 following experimental traumatic brain injury. J Neurotrauma 2000; 17:69-81. [PMID: 10674759 DOI: 10.1089/neu.2000.17.69] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Prostaglandins, potent mediators of inflammation, are generated from arachidonic acid (AA) via the action of cyclooxygenase-1 and -2 (COX-1 and COX-2). In this study, we report that lateral cortical impact injury in rats significantly increases COX-2 protein levels both in the cortex surrounding the injury site and the ipsilateral hippocampus. COX-2 protein level was elevated as early as 3 h postinjury and persisted for up to 3 days. Increases in immunoreactivity were detected not only in the adjacent cortex and hippocampus, but were also observed in the contralateral cortex and hippocampus, the ipsilateral piriform cortex and the ipsilateral amygdaloid complex. COX-2 immunoreactive cells appear morphologically normal and do not present any of the characteristic features of apoptosis. Double immunostaining experiments using either a neuron-specific or an astroglial-specific marker show that the expression of COX-2 is localized almost exclusively in neuronal cells. Administration of the COX-2 inhibitor 4-[5-(4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]benzenesulfona mide (celecoxib, marketed as Celebrex) worsens motor, but not cognitive, performance, suggesting that COX-2 induction following traumatic brain injury may play a protective role.
Collapse
Affiliation(s)
- P K Dash
- Department of Neurobiology and Anatomy, University of Texas Medical School, Houston 77225, USA.
| | | | | |
Collapse
|
32
|
Dhillon HS, Carman HM, Prasad RM. Regional activities of phospholipase C after experimental brain injury in the rat. Neurochem Res 1999; 24:751-5. [PMID: 10447458 DOI: 10.1023/a:1020779413122] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Regional activities of phosphoinositide-specific phospholipase C (PLC) were measured after lateral fluid percussion (FP) brain injury in rats. The activity of PLC on phosphatidylinositol 4,5-bisphosphate (PIP2) in the rat cortex required calcium, and at 45 microM concentration it increased PLC activity by about ten-fold. The activity of PLC was significantly increased in the cytosol fraction in the injured (left) cortex (IC) at 5 min, 30 min and 120 min after brain injury. However, in the same site, increases were observed in the membrane fraction only at 5 min after brain injury. In both the contralateral (right) cortex (CC) and ipsilateral hippocampus (IH), the activity of PLC was increased in the cytosol only at 5 min after brain injury. These results suggest that increased activity of PLC may contribute to increases in levels of cellular diacylglycerol and inositol trisphosphate in the IC (the greatest site of injury), and to a smaller extent in the IH and CC, after lateral FP brain injury. It is likely that this increased PLC activity is caused by alteration in either the levels or activities of one or more of its isozymes (PLCbeta, PLCgamma, and PLCdelta) after FP brain injury.
Collapse
Affiliation(s)
- H S Dhillon
- Department of Surgery, University of Kentucky Chandler Medical Center, Lexington 40536-0084, USA
| | | | | |
Collapse
|
33
|
Golding EM, Robertson CS, Bryan RM. The consequences of traumatic brain injury on cerebral blood flow and autoregulation: a review. Clin Exp Hypertens 1999; 21:299-332. [PMID: 10369378 DOI: 10.3109/10641969909068668] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this decade, the brain argueably stands as one of the most exciting and challenging organs to study. Exciting in as far as that it remains an area of research vastly unknown and challenging due to the very nature of its anatomical design: the skull provides a formidable barrier and direct observations of intraparenchymal function in vivo are impractical. Moreover, traumatic brain injury (TBI) brings with it added complexities and nuances. The development of irreversible damage following TBI involves a plethora of biochemical events, including impairment of the cerebral vasculature, which render the brain at risk to secondary insults such as ischemia and intracranial hypertension. The present review will focus on alterations in the cerebrovasculature following TBI, and more specifically on changes in cerebral blood flow (CBF), mediators of CBF including local chemical mediators such as K+, pH and adenosine, endothelial mediators such as nitric oxide and neurogenic mediators such as catecholamines, as well as pressure autoregulation. It is emphasized that further research into these mechanisms may help attenuate the prevalence of secondary insults and therefore improve outcome following TBI.
Collapse
Affiliation(s)
- E M Golding
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
34
|
Kyrkanides S, Olschowka JA, Williams JP, Hansen JT, O'Banion MK. TNF alpha and IL-1beta mediate intercellular adhesion molecule-1 induction via microglia-astrocyte interaction in CNS radiation injury. J Neuroimmunol 1999; 95:95-106. [PMID: 10229119 DOI: 10.1016/s0165-5728(98)00270-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Radiation injury to the central nervous system (CNS) results in glial activation accompanied by expression of pro-inflammatory cytokines and adhesion molecules. In this study we demonstrate intercellular adhesion molecule-1 (ICAM-1) induction in the irradiated mouse brain at the mRNA and protein levels. Immunocytochemical analysis revealed that ICAM-1 protein was primarily expressed in endothelial cells and microglia. In vitro, ionizing radiation significantly induces TNF alpha, IL-1beta and ICAM-1 mRNA in primary microglia cultures. Interestingly, although ionizing radiation activated primary astrocyte cultures, it did not induce ICAM-1 expression. However, exposure of astrocytes to conditioned medium collected from irradiated microglia resulted in ICAM-1 induction, which was abrogated when the conditioned medium was pre-incubated with neutralizing antibodies raised against murine TNF alpha and IL-1beta. These results indicate that pro-inflammatory cytokines may be necessary for ICAM-1 expression in astrocytes in CNS radiation injury.
Collapse
Affiliation(s)
- S Kyrkanides
- Department of Neurobiology and Anatomy, School of Medicine and Dentistry, University of Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
35
|
|
36
|
Hewett SJ. Interferon-gamma reduces cyclooxygenase-2-mediated prostaglandin E2 production from primary mouse astrocytes independent of nitric oxide formation. J Neuroimmunol 1999; 94:134-43. [PMID: 10376946 DOI: 10.1016/s0165-5728(98)00240-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) and prostaglandins (PGs) modulate inflammatory and immune responses in the central nervous system (CNS). Both NO and PG synthesis have been described in appropriately stimulated astrocytes. In other systems, both positive and negative modulation of cyclooxygenase (COX) activity, hence PG synthesis, have been described by NO. Since interferon (IFN)-gamma is known to upregulate the production of NO from astrocytes, the present study was designed to investigate the effect of IFNgamma on PG production from activated astrocytes and to determine whether this effect is mediated by NO. Astrocytic PG production was induced by exposure of murine cortical cultures to lipopolysaccharide (LPS). This induction was time- and concentration-dependent, and prevented by inhibitors of transcription and translation, as well as the selective COX-2 inhibitor, NS-398. LPS-induced expression of COX-2 mRNA and protein was confirmed by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis, respectively. Exposure of LPS-treated astrocytes to IFNgamma resulted in a concentration-dependent decrease in PGE2 accumulation which was accompanied by a striking parallel increase in NO formation. However, the NOS inhibitors, N(G)-nitro-L-arginine or N6-(1-iminoethyl)-lysine, failed to reverse the IFNgamma-mediated diminution of LPS-induced PGE2 production, indicating that the IFN-gamma-mediated reduction in COX-2-dependent PGE2 production occurred independent of NO formation. Additional experiments demonstrated that IFN-gamma acted mainly by downregulating the expression of COX-2 protein. Present results indicate that PG and NO synthesis in mouse cortical astrocytes in vitro are under the direct reciprocal control of IFNgamma.
Collapse
Affiliation(s)
- S J Hewett
- Department of Pharmacology, University of Connecticut Health Center, Farmington 06030-6125, USA.
| |
Collapse
|
37
|
Morrison B, Saatman KE, Meaney DF, McIntosh TK. In vitro central nervous system models of mechanically induced trauma: a review. J Neurotrauma 1998; 15:911-28. [PMID: 9840765 DOI: 10.1089/neu.1998.15.911] [Citation(s) in RCA: 158] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Injury is one of the leading causes of death among all people below the age of 45 years. In the United States, traumatic brain injury (TBI) and spinal cord injury (SCI) together are responsible for an estimated 90,000 disabled persons annually. To improve treatment of the patient and thereby decrease the associated mortality, morbidity, and cost, several in vivo models of central nervous system (CNS) injury have been developed and characterized over the past two decades. To complement the ability of these in vivo models to reproduce the sequelae of human CNS injury, in vitro models of neuronal injury have also been developed. Despite the inherent simplifications of these in vitro systems, many aspects of the posttraumatic sequelae are faithfully reproduced in cultured cells, including ultrastructural changes, ionic derangements, alterations in electrophysiology, and free radical generation. This review presents a number of these in vitro systems, detailing the mechanical stimuli, the types of tissue injured, and the in vivo injury conditions most closely reproduced by the models. The data generated with these systems is then compared and contrasted with data from in vivo models of CNS injury. We believe that in vitro models of mechanical injury will continue to be a valuable tool to study the cellular consequences and evaluate the potential therapeutic strategies for the treatment of traumatic injury of the CNS.
Collapse
Affiliation(s)
- B Morrison
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA
| | | | | | | |
Collapse
|
38
|
Carman HM, Dhillon HS, Zhang D, Geddes JW, Prasad RM. Regional levels of phospholipase Cgamma after fluid percussion brain injury in the rat. Brain Res 1998; 808:116-9. [PMID: 9795178 DOI: 10.1016/s0006-8993(98)00824-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Levels of PLCgamma, a phospholipase C (PLC) isozyme, were significantly increased in the cytosol in the injured left cortex (LC) at 5, 30 and 120 min after brain injury. In the same site, although levels of membrane PLCgamma did not alter at 5 and 30 min, they were found to be decreased at 2 h after brain injury. In general, the levels of both cytosolic and membrane PLCgamma were unaltered in the contralateral right cortex (RC), ipsilateral left hippocampus (LH) and contralateral right hippocampus (RH) between 5 and 120 min after brain injury. These results suggest that, in addition to well-proposed excitatory neurotransmitter-receptor systems, increased levels of PLCgamma may also contribute to alterations in PIP2 signal transduction pathway, particularly in the greatest injury site (LC) after lateral FP brain injury.
Collapse
Affiliation(s)
- H M Carman
- Department of Surgery, University of Kentucky Chandler Medical Center, Lexington, KY 40536-0084, USA
| | | | | | | | | |
Collapse
|
39
|
McIntosh TK, Juhler M, Wieloch T. Novel pharmacologic strategies in the treatment of experimental traumatic brain injury: 1998. J Neurotrauma 1998; 15:731-69. [PMID: 9814632 DOI: 10.1089/neu.1998.15.731] [Citation(s) in RCA: 243] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The mechanisms underlying secondary or delayed cell death following traumatic brain injury are poorly understood. Recent evidence from experimental models suggests that widespread neuronal loss is progressive and continues in selectively vulnerable brain regions for months to years after the initial insult. The mechanisms underlying delayed cell death are believed to result, in part, from the release or activation of endogenous "autodestructive" pathways induced by the traumatic injury. The development of sophisticated neurochemical, histopathological and molecular techniques to study animal models of TBI have enabled researchers to begin to explore the cellular and genomic pathways that mediate cell damage and death. This new knowledge has stimulated the development of novel therapeutic agents designed to modify gene expression, synthesis, release, receptor or functional activity of these pathological factors with subsequent attenuation of cellular damage and improvement in behavioral function. This article represents a compendium of recent studies suggesting that modification of post-traumatic neurochemical and cellular events with targeted pharmacotherapy can promote functional recovery following traumatic injury to the central nervous system.
Collapse
Affiliation(s)
- T K McIntosh
- Department of Neurosurgery, University of Pennsylvania, Philadelphia 19104-6316, USA
| | | | | |
Collapse
|
40
|
Abstract
Previous studies have observed that ATP- and calcium-sensitive K+ (KATP and Kca) channel function is impaired after fluid percussion brain injury (FPI). The present study was designed to characterize the effect of FPI on prostaglandin (PG)E2 and 12 pial artery dilation and the role of activation of these K+ channels in that dilation in newborn pigs equipped with a closed cranial window. FPI of moderate severity (1.9-2.1 atm) was produced by using a pendulum to strike a piston on a saline-filled cylinder that was fluid coupled to the brain via a hollow screw inserted through the cranium. PGE2 vasodilation was blunted by FPI (9+/-1%, 13+/-1%, and 19+/-1% vs. 2+/-1%, 5+/-1%, and 9+/-1%, for 1, 10, and 100 ng/ml PGE2 before and after FPI, respectively). PGE2 dilation was associated with increased CSF cGMP and cAMP concentration and such changes in cyclic nucleotides were blunted by FPI (448+/-10 and 793+/-38 vs. 316+/-11 and 403+/-27 fmol/ml for control and PGE2 induced change in cGMP before and after FPI, respectively). PGI2-induced dilation and associated changes in CSF cyclic nucleotide concentration were similarly blunted by FPI. PGE2 dilation was attenuated by either glibenclamide or iberiotoxin, KATP and K,ca channel antagonists, and coadministration of both K+ channel antagonists further decremented the dilator response (9+/-1%, 14+/-1%, and 21+/-1%; vs. 4+/-1%, 7+/-1%, and 12+/-1%; vs. 2+/-1%, 4+/-1%, and 7+/-1%, for 1, 10, and 100 ng/ml PGE2 during control, after glibenclamide, and after combined glibenclamide and iberiotoxin, respectively). Glibenclamide and iberiotoxin had similar effects on PGI2 dilation. These data show that prostaglandin dilation is attenuated after FPI. These data also show that prostaglandin dilation is dependent on activation of both KATP and Kca channels. Further, these data suggest that attenuated prostaglandin dilation following FPI results from diminished prostaglandin-associated elevation in cyclic nucleotide concentration and impaired KATP and Kca channel function.
Collapse
Affiliation(s)
- W M Armstead
- Department of Anesthesia, University of Pennsylvania and the Children's Hospital of Philadelphia 19104, USA
| |
Collapse
|
41
|
Caggiano AO, Kraig RP. Prostaglandin E2 and 4-aminopyridine prevent the lipopolysaccharide-induced outwardly rectifying potassium current and interleukin-1beta production in cultured rat microglia. J Neurochem 1998; 70:2357-68. [PMID: 9603200 PMCID: PMC2807138 DOI: 10.1046/j.1471-4159.1998.70062357.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Brain inflammation includes microglial activation and enhanced production of diffusible chemical mediators, including prostaglandin E2. Prostaglandin E2 is generally considered a proinflammatory molecule, but it also promotes neuronal survival and down-regulates some aspects of microglial activation. It remains unknown, however, if and how prostaglandin E2 prevents microglial activation. In primary culture, microglial activation is predicted by a characteristic pattern of whole-cell potassium currents and interleukin-1beta production. We investigated if prostaglandin E2 could alter these currents and, if so, whether these currents are necessary for microglial activation. Microglia were isolated from mixed cell cultures prepared from neonatal rat brains and exposed to 0-10 microM prostaglandin E2 and lipopolysaccharide for 24 h. Currents were elicited by using standard patch-clamp technique, and interleukin-1beta production was measured by ELISA. Peak outward current densities in microglia treated with lipopolysaccharide plus prostaglandin E2 (10 nM) were reduced significantly from those of cells treated with lipopolysaccharide alone. Prostaglandin E2 and 4-aminopyridine (a blocker of outward potassium currents) also significantly reduced interleukin-1beta production. Thus, although prostaglandin E2 is classified generally as a proinflammatory chemical, it has complex roles in brain inflammation that include preventing microglial activation, perhaps by reducing the outward potassium current.
Collapse
Affiliation(s)
- A O Caggiano
- Committee on Neurobiology, University of Chicago, Illinois 60637, USA
| | | |
Collapse
|
42
|
Fabian RH, Dewitt DS, Kent TA. The 21-aminosteroid U-74389G reduces cerebral superoxide anion concentration following fluid percussion injury of the brain. J Neurotrauma 1998; 15:433-40. [PMID: 9624628 DOI: 10.1089/neu.1998.15.433] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We examined the effects of the 21-aminosteroid antioxidant U-74389G (16-desmethyl-tirilazad) on the concentration of extracellular superoxide anion following fluid percussion traumatic brain injury (TBI) measured by a cytochrome c-coated electrode and on local cerebral perfusion (CBFld) measured by laser Doppler flowmetry (LDF). U-74389G in a dose of 3 mg/kg reduced superoxide anion concentrations 60 min after TBI significantly but had no significant effect on CBFld. These results indicate that reduction of CBF after TBI can be dissociated from superoxide anion production. Persistent ischemia may limit neuroprotection efficacy and may contribute to divergent outcome results in clinical and animal trials using agents to modify reactive oxygen species.
Collapse
Affiliation(s)
- R H Fabian
- Department of Neurology, University of Texas Medical Branch, Galveston 77555-0539, USA
| | | | | |
Collapse
|
43
|
|
44
|
Al-Turki A, Armstead WM. Altered release of prostaglandins by opioids contributes to impaired cerebral hemodynamics following brain injury. Crit Care Med 1998; 26:917-25. [PMID: 9590323 DOI: 10.1097/00003246-199805000-00029] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES After fluid percussion brain injury (FPI) in the newborn pig, pial arteries constrict and responses to dilator stimuli, including opioids, are blunted. This study was designed to determine if altered release of prostaglandins contributes to blunted opioid dilation of cerebral arteries in newborn piglets following brain injury. DESIGN Prospective, in vivo, cerebral hemodynamic animal study. SETTING University research laboratory. SUBJECTS Newborn (1- to 5-days old) piglets of either gender. INTERVENTIONS In anesthetized, newborn, 1- to 5-day-old pigs, a closed cranial window was used to measure pial artery diameter and to collect cortical periarachnoid cerebrospinal fluid (CSF) for determination of 6-keto-PGF1alpha, the stable metabolite of prostaglandin I2 (PGI2) and thromboxane B2 (TXB2), the stable metabolite of TXA2, via radioimmunoassay. FPI of moderate severity (1.9 to 2.3 atmospheres) was produced by using a pendulum to strike a piston on a saline-filled cylinder that was fluid coupled to the brain via a hollow screw inserted through the cranium. MEASUREMENTS AND MAIN RESULTS Methionine enkephalin (Met) vasodilation was blunted after FPI but was partially restored with indomethacin pretreatment (5 mg/kg i.v.) (8 +/- 1 [SEM] %, 13 +/- 1%, and 20 +/- 1% vs. 1 +/- 1%, 3 +/- 1%, and 5 +/- 1% vs. 7 +/- 1%, 10 +/- 1%, and 15 +/- 1%, respectively, for 10(-10), 10(-8), and 10(-6) M Met during control conditions, after FPI, and after FPI pretreated with indomethacin, n = 6). Similarly, restoration of Met dilation after FPI was observed with SQ 29,548, a TXA2 antagonist. Met-induced 6-keto-PGF1alpha release was blunted following FPI (889 +/- 20, 1130 +/- 33, and 1886 +/- 59 vs. 2630 +/- 36, 2775 +/- 30, and 2825 +/- 36 pg/mL for control, 10(-10), and 10(-6) M Met before and after FPI, respectively, n = 6). In contrast, Met-induced TXB2 release was enhanced after FPI (340 +/- 20, 423 +/- 25, and 473 +/- 30 pg/mL vs. 518 +/- 30, 726 +/- 90, and 901 +/- 35 pg/mL for control, 10(-10), and 10(-6) M Met before and after FPI, respectively, n = 6). Leucine enkephalin- and dynorphin-induced dilation and associated prostaglandin release were similarly altered following FPI. Beta endorphin-induced constriction was enhanced following FPI, and these potentiated responses were blunted after indomethacin or SQ 29,548 pretreatment. CONCLUSIONS These data show that FPI increases CSF 6-keto-PGF1alpha and TXB2 concentrations. These data suggest that altered release of prostaglandins by opioids contribute to impaired cerebral hemodynamics following FPI in piglets.
Collapse
Affiliation(s)
- A Al-Turki
- Department of Anesthesia, University of Pennsylvania and the Children's Hospital of Philadelphia, 19104, USA
| | | |
Collapse
|
45
|
Prasad RM, Laabich A, Dhillon HS, Zhang L, Maki A, Clerici WJ, Hicks R, Butcher J, Barron S. Effects of six weeks of chronic ethanol administration on lactic acid accumulation and high energy phosphate levels after experimental brain injury in rats. J Neurotrauma 1997; 14:919-30. [PMID: 9475373 DOI: 10.1089/neu.1997.14.919] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The effects of 6 weeks of chronic ethanol administration on the lateral fluid percussion (FP) brain injury-induced regional accumulation of lactate and on the levels of total high-energy phosphates were examined in rats. In both the chronic ethanol diet (ethanol diet) and pair-fed isocaloric sucrose control diet (control diet) groups, tissue concentrations of lactate were elevated in the cortices and hippocampi of both the ipsilateral and contralateral hemispheres at 5 min after brain injury. In both diet groups, concentrations of lactate were elevated only in the injured left cortex and the ipsilateral hippocampus at 20 min after FP brain injury. No significant differences were found in the levels of lactate in the cortices and hippocampi of sham animals and brain-injured animals between the ethanol and control diet groups at 5 min and 20 min after injury. In the ethanol and control diet groups, tissue concentrations of total high-energy phosphates (ATP + PCr) were not affected in the cortices and hippocampi at 5 min and 20 min after lateral FP brain injury. No significant differences were found in the levels of total high-energy phosphates in the cortices and hippocampi of the sham and brain-injured animals between the ethanol and control diet groups at 5 min and 20 min after injury. Histologic studies revealed a similar extent of damage in the cortex and in the CA3 region of the ipsilateral hippocampus in both diet groups at 14 days after lateral FP brain injury. These findings suggest that 6 weeks of chronic ethanol administration does not alter brain injury-induced accumulation of lactate, levels of total high energy phosphates, and extent of morphological damage.
Collapse
Affiliation(s)
- R M Prasad
- Department of Surgery, University of Kentucky Chandler Medical Center, University of Kentucky, Lexington 40536-0084, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dewitt DS, Prough DS, Uchida T, Deal DD, Vines SM. Effects of nalmefene, CG3703, tirilazad, or dopamine on cerebral blood flow, oxygen delivery, and electroencephalographic activity after traumatic brain injury and hemorrhage. J Neurotrauma 1997; 14:931-41. [PMID: 9475374 DOI: 10.1089/neu.1997.14.931] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hemorrhage after traumatic brain injury (TBI) in cats produces significant decreases in cerebral oxygen delivery (DcereO2) and electroencephalographic (EEG) activity. To determine whether effective treatments for the separate insults of TBI and hemorrhagic shock would also prove effective after the clinically relevant combination of the two, we measured the effects of a kappa-opiate antagonist (nalmefene), an inhibitor of lipid peroxidation (tirilazad), a thyrotropin-releasing hormone analog (CG3703), a clinically useful pressor agent (dopamine) or a saline placebo on cerebral blood flow (CBF), and EEG activity after TBI and mild hemorrhagic hypotension. Cats (n = 40, 8 per group) were anesthetized with 1.6% isoflurane in N2O:O2 (70:30) and prepared for fluid-percussion TBI and microsphere measurements of CBF. Cats were randomized to receive nalmefene (1 mg/kg), tirilazad (5 mg/kg), CG3703 (2 mg/kg), dopamine (20 microg x kg(-1) x min[-1]) or a saline placebo (2 ml, 0.9% NaCl). Animals were injured (2.2 atm), hemorrhaged to 70% of preinjury blood volume, treated as just described and resuscitated with a volume of 10% hydroxyethyl starch equal to shed blood. CBF was determined and EEG activity recorded before injury, after hemorrhage, and 0, 60, and 120 min after resuscitation (R0, R60, and R120). CBF increased significantly after resuscitation (R0) in the nalmefene- and CG3703-treated groups. CBF did not differ significantly from baseline in any group at R60 or R120. DcereO2 was significantly less than baseline in the saline-, dopamine-, and tirilazad-treated groups at R60 and in the dopamine-, tirilazad-, and CG3703-treated groups at R120. EEG activity remained unchanged in the nalmefene-treated group but deteriorated significantly at R60 or R120 compared to baseline in the other groups. Nalmefene and CG3703 preserved the hyperemic response to hemodilution (otherwise antagonized by TBI), and nalmefene prevented the deterioration in DcereO2 and EEG activity that occurs after TBI and hemorrhage.
Collapse
Affiliation(s)
- D S Dewitt
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston 77555-0591, USA
| | | | | | | | | |
Collapse
|
47
|
Drugs in the Management of Cute Traumatic Brain Injury. Phys Med Rehabil Clin N Am 1997. [DOI: 10.1016/s1047-9651(18)30294-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
48
|
Abstract
Motor vehicle-related accidents account for the largest number of head injuries in all ages. This article reviews types of injury, neurologic assessment, secondary injury, brain swelling, seizures, resuscitation, and intensive care.
Collapse
Affiliation(s)
- R T Mansfield
- Department of Pediatrics, State University of New York at Stony Brook, USA
| |
Collapse
|
49
|
Abstract
Increases in metabolic rate reported in head-injured patients can contribute to increases in respiratory demand, raised body temperature, and host body wasting (cachexia). The objective of the present study was to quantify the metabolic responses to brain trauma in the rat and investigate the underlying mechanisms. Lateral fluid-percussion (FP) injury (applied cortical pressure 1.6-1.8 atm) in the rat resulted in consistent and reproducible cortical brain lesions (44 +/- 6 mm3). Body weight and food intake were reduced significantly 24 h after brain trauma compared to sham-operated (7 and 49%,p < 0.01) and control animals (14 and 65%,p < 0.001), respectively. Resting oxygen consumption (V(O2), measured at 24 degrees C) was increased significantly, by 9-16% above sham-operated, and 14-26% above control animals for 2-7 h after brain trauma (p < 0.05), but V(O2) was not raised thereafter (24-72 h) and colonic temperature was not changed. Raising the ambient temperature from 24 degrees C to 28 degrees C significantly reduced the hypermetabolism of brain-injured rats compared to sham-operated controls. Injection of the beta-adrenoceptor antagonist propranolol (10 mg/kg, i.p.) completely abolished the rise in metabolic rate of brain-injured rats, and reduced significantly the rise in metabolic rate of the sham-operated animals (26%, p < 0.01 and 11%, p < 0.05; respectively). Systemic injection of the cyclo-oxygenase inhibitor indomethacin (1 mg/kg, i.p.) significantly attenuated (by 11%,p < 0.01), but did not completely abolish the hypermetabolism of brain-injured animals. Lateral FP injury in the rat causes a significant cachexia. Weight loss is due to hypophagia, and an increase in energy expenditure, which is mediated by sympathetic activation of thermogenesis and in part by prostaglandins.
Collapse
Affiliation(s)
- S Y Roe
- Biological Sciences, University of Manchester, UK
| | | |
Collapse
|
50
|
Abstract
The recent appreciation that traumatic brain injury is a dynamic process, initiated at the time of injury but not concluded for hours to days afterward, has resulted in the expectation that treatments can be designed to interrupt the processes that result in delayed cellular dysfunction and, thus, can decrease the amount of traumatic brain damage. Thus, for the first time, treatments specific for brain damage are envisioned. These can provide a fundamentally different approach to the treatment of the damaged brain than currently used treatments that deal with epiphenomena of traumatic injury, such as increased intracranial pressure or secondary ischemia. The processes that result in delayed cellular damage may be initiated by transient ionic fluxes induced by traumatic, temporary holes in the cell membrane lipid bilayer (mechanoporation). Resulting changes in intracellular ionic composition, if uncorrected, result in 1) traumatic depolarization with resultant neurotransmitter release, postsynaptic receptor dysfunction, and excitability changes; 2) calcium-mediated activation of proteases and phospholipases, with resultant cytoskeletal protein dissolution and free radical-induced lipid peroxidation; 3) inflammatory processes that elicit tissue-damaging cytokines; and 4) immediate and delayed activation of numerous genes with a resultant production of a panoply of new proteins. The future challenge to neurotrauma investigators is to better understand these processes and to develop interventions that will halt them before permanent brain damage occurs.
Collapse
Affiliation(s)
- Thomas A. Gennarelli
- Department of Neurosurgery and Center for Neurosciences Allegheny University of the Health Sciences Philadelphia, Pennsylvania
| |
Collapse
|