51
|
Cerebral blood volume alterations in the perilesional areas in the rat brain after traumatic brain injury--comparison with behavioral outcome. J Cereb Blood Flow Metab 2010; 30:1318-28. [PMID: 20145657 PMCID: PMC2949222 DOI: 10.1038/jcbfm.2010.15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the traumatic brain injury (TBI) the initial impact causes both primary injury, and launches secondary injury cascades. One consequence, and a factor that may contribute to these secondary changes and functional outcome, is altered hemodynamics. The relative cerebral blood volume (CBV) changes in rat brain after severe controlled cortical impact injury were characterized to assess their interrelations with motor function impairment. Magnetic resonance imaging (MRI) was performed 1, 2, 4 h, and 1, 2, 3, 4, 7, and 14 days after TBI to quantify CBV and water diffusion. Neuroscore test was conducted before, and 2, 7, and 14 days after the TBI. We found distinct temporal profile of CBV in the perilesional area, hippocampus, and in the primary lesion. In all regions, the first response was drop of CBV. Perifocal CBV was reduced for over 4 days thereafter gradually recovering. After the initial drop, the hippocampal CBV was increased for 2 weeks. Neuroscore demonstrated severely impaired motor functions 2 days after injury (33% decrease), which then slowly recovered in 2 weeks. This recovery parallelled the recovery of perifocal CBV. CBV MRI can detect cerebrovascular pathophysiology after TBI in the vulnerable perilesional area, which seems to potentially associate with time course of sensory-motor deficit.
Collapse
|
52
|
Ge Y, Patel MB, Chen Q, Grossman EJ, Zhang K, Miles L, Babb JS, Reaume J, Grossman RI. Assessment of thalamic perfusion in patients with mild traumatic brain injury by true FISP arterial spin labelling MR imaging at 3T. Brain Inj 2010; 23:666-74. [PMID: 19557570 DOI: 10.1080/02699050903014899] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To assess cerebral blood flow (CBF) changes in patients with mild traumatic brain injury (MTBI) using an arterial spin labelling (ASL) perfusion MRI and to investigate the severity of neuropsychological functional impairment with respect to haemodynamic changes. MATERIALS AND METHODS Twenty-one patients with MTBI and 20 healthy controls were studied at 3T MR. The median time since the onset of brain injury in patients was 24.6 months. Both patients and controls underwent a traditional consensus battery of neurocognitive tests. ASL was performed using true fast imaging with steady state precession and a flow-sensitive alternating inversion recovery preparation. Regional CBF were measured in both deep and cortical gray matter as well as white matter at the level of basal ganglia. RESULTS The mean regional CBF was significantly lower in patients with MTBI (45.9 +/- 9.8 ml/100 g min(-1)) as compared to normal controls (57.1 +/- 8.1 ml/100 g min(-1); p = 0.002) in both sides of thalamus. The decrease of thalamic CBF was significantly correlated with several neurocognitive measures including processing and response speed, memory/learning, verbal fluency and executive function in patients. CONCLUSIONS Haemodynamic impairment can occur and persist in patients with MTBI, the extent of which is more severe in thalamic regions and correlate with neurocognitive dysfunction during the extended course of disease.
Collapse
Affiliation(s)
- Yulin Ge
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York 10016, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Schwarzmaier SM, Kim SW, Trabold R, Plesnila N. Temporal profile of thrombogenesis in the cerebral microcirculation after traumatic brain injury in mice. J Neurotrauma 2010; 27:121-30. [PMID: 19803784 DOI: 10.1089/neu.2009.1114] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with an almost immediate reduction in cerebral blood flow (CBF). Because cerebral perfusion pressure is often normal under these circumstances it was hypothesized that the reduction of post-traumatic CBF has to occur at the level of the microcirculation. The aim of the current study was to investigate whether cerebral microvessels are involved in the development of blood flow disturbances following experimental TBI. C57/BL6 mice (n = 12) were intubated and ventilated under control of end-tidal Pco(2) ((ET)P(CO2)). After preparation of a cranial window and baseline recordings, the animals were subjected to experimental TBI by controlled cortical impact (CCI; 6 m/sec, 0.5 mm). Vessel lumina and intravascular cells were visualized by in vivo fluorescence microscopy (IVM) using the fluorescent dyes FITC-dextran and rhodamine 6G, respectively. Vessel diameter, cell-endothelial interactions, and thrombus formation were quantified within the traumatic penumbra by IVM up to 2 h after CCI. Arteriolar diameters increased after CCI by 26.2 +/- 2.5% (mean +/- SEM, p < 0.01 versus baseline), and remained at this level until the end of the observation period. Rolling of leukocytes on the cerebrovascular endothelium was observed both in arterioles and venules, while leukocyte-platelet aggregates were found only in venules. Microthrombi occluded up to 70% of venules and 33% of arterioles. The current data suggest that the immediate post-traumatic decrease in peri-contusional blood flow is not caused by arteriolar vasoconstriction, but by platelet activation and the subsequent formation of thrombi in the cerebral microcirculation.
Collapse
Affiliation(s)
- Susanne M Schwarzmaier
- Institute for Surgical Research in the Walter Brendel Center for Experimental Medicine, Department of Neurosurgery, University of Munich Medical Center-Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | | | | | | |
Collapse
|
54
|
Mao H, Yang KH, King AI, Yang K. Computational neurotrauma--design, simulation, and analysis of controlled cortical impact model. Biomech Model Mechanobiol 2010; 9:763-72. [PMID: 20372960 DOI: 10.1007/s10237-010-0212-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 03/19/2010] [Indexed: 11/28/2022]
Abstract
The controlled cortical impact (CCI) model is widely used in many laboratories to study traumatic brain injury (TBI). Although external impact parameters during CCI tests could be clearly defined, little is known about the internal tissue-level mechanical responses of the rat brain. Furthermore, the external impact parameters tend to vary considerably among different labs making the comparison of research findings difficult if not impossible. In this study, a design of computer experiments was performed with typical external impact parameters commonly found in the literature. An anatomically detailed finite element (FE) rat brain model was used to simulate the CCI experiments to correlate external mechanical parameters (impact depth, impact velocity, impactor shape, impactor size, and craniotomy pattern) with rat brain internal responses, as predicted by the FE model. Systematic analysis of the results revealed that impact depth was the leading factor affecting the predicted brain internal responses. Interestingly, impactor shape ranked as the second most important factor, surpassing impactor diameter and velocity which were commonly reported in the literature as indicators of injury severity along with impact depth. The differences in whole brain response due to a unilateral or a bilateral craniotomy were small, but those of regional intracranial tissue stretches were large. The interaction effects of any two external parameters were not significant. This study demonstrates the potential of using numerical FE modeling to engineer better experimental TBI models in the future.
Collapse
Affiliation(s)
- Haojie Mao
- Bioengineering Center, Wayne State University, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
55
|
Zweckberger K, Plesnila N. Anatibant®, a selective non-peptide bradykinin B2 receptor antagonist, reduces intracranial hypertension and histopathological damage after experimental traumatic brain injury. Neurosci Lett 2009; 454:115-7. [DOI: 10.1016/j.neulet.2009.02.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 02/06/2009] [Accepted: 02/06/2009] [Indexed: 11/28/2022]
|
56
|
Yao C, Williams AJ, Ottens AK, May Lu XC, Chen R, Wang KK, Hayes RL, Tortella FC, Dave JR. Detection of protein biomarkers using high-throughput immunoblotting following focal ischemic or penetrating ballistic-like brain injuries in rats. Brain Inj 2009; 22:723-32. [PMID: 18720098 DOI: 10.1080/02699050802304706] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PRIMARY OBJECTIVE Recent efforts have been aimed at developing a panel of protein biomarkers for the diagnosis/prognosis of the neurological damage associated with acute brain injury. METHODS AND PROCEDURES This study utilized high-throughput immunoblotting (HTPI) technology to compare changes between two animal models of acute brain injury: penetrating ballistic-like brain injury (PBBI) which mimics the injury created by a gunshot wound and transient middle cerebral artery occlusion (MCAo) which is a model of stroke. Brain and blood were collected at 24-hours post-injury. MAIN OUTCOMES AND RESULTS This study identified the changes in 18 proteins following PBBI and 17 proteins following MCAo out of a total of 998 screened proteins. Distinct differences were observed between the two models: five proteins were up- or down-regulated in both models, 23 proteins changed in only one model and one protein was differentially expressed. Western blots were used to verify HTPI results for selected proteins with measurable changes observed in both blood and brain for the proteins STAT3, Tau, PKA RII beta, 14-3-3 epsilon and p43/EMAPII. CONCLUSIONS These results suggest distinct post-injury protein profiles between brain injury types (traumatic vs. ischemic) that will facilitate strategies aimed at the differential diagnosis and prognosis of acute brain injury.
Collapse
Affiliation(s)
- Changping Yao
- Department of Applied Neurobiology, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
57
|
|
58
|
Timaru-Kast R, Meissner A, Heimann A, Hoelper B, Kempski O, Alessandri B. Acute subdural hematoma in pigs: role of volume on multiparametric neuromonitoring and histology. J Neurotrauma 2008; 25:1107-19. [PMID: 18771396 DOI: 10.1089/neu.2008.0517] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) is often complicated by acute subdural hemorrhage (ASDH) with a high mortality rate. The pathophysiological mechanisms behind such an injury type and the contribution of blood to the extent of an injury remain poorly understood. Therefore, the goals of this study were to establish a porcine ASDH model in order to investigate pathomechanisms of ASDH and to compare effects induced by blood or sheer volume. Thus, we infused 2, 5, and 9 mL of blood (up to 15% of intracranial volume), and we compared a 5-mL blood and paraffin oil volume to separate out effects of extravasated blood on brain tissue. An extended neuromonitoring was applied that lasted up to 12 h after injury and included intracranial pressure (ICP), cerebral perfusion pressure (CPP), tissue oxygen concentration (ptiO(2)), biochemical markers (glutamate, lactate), somatosensory evoked potentials (SEP), brain water content, and histological assessment (Lesion Index [LI]). Volume-dependent changes were detected mainly during the first hours after injury. ICP increased to significant levels (p < 0.05) of 36.89 +/- 1.59, 15.52 +/- 0.48, and 11.25 +/- 0.35 mm Hg after 9, 5, and 2 mL of subdural blood, respectively (sham, 4.85 +/- 0.06 mm Hg). The ptiO(2) dropped drastically after 9 mL of subdural blood without recovery in both hemispheres to below 20% of baseline, but was affected little after 2 and 5 mL in the acute monitoring period (maximal drop to 71% of baseline). Later, 5 mL of blood led to a significant increase of ptiO(2) compared to 2 mL ipsilaterally (p < 0.05). Glutamate and lactate showed a comparable pattern with a long-lasting increase after 9 mL of blood and short-lasting changes after 2 and 5 mL. The two smaller volumes caused an increased brain swelling (2 mL, 80.60 +/- 0.34%; 5 mL, 81.20 +/- 0.66%; p < 0.05 vs. sham), a significant LI (sham, 6.4 +/- 1.4; 2 mL, 30.0 +/- 0.95; 5 mL, 32.1 +/- 1.2; p < 0.05 vs. sham), and a reduced SEP amplitude (5 mL, p < 0.05 vs. baseline) at the end of the experiment. A 9-mL led to herniation during the experiment causing dramatical brain swelling and acute histological damage. Comparison of blood volume with paraffin oil showed no significance, indicating that volume alone determines the acute pathophysiological processes leading to a rapidly developing histological damage. Additional effects due to blood contact with brain tissue (e.g., inflammation) may be detected only at later time points (>12 h).
Collapse
Affiliation(s)
- Ralph Timaru-Kast
- Institute for Neurosurgical Pathophysiology, Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
59
|
Engel DC, Mies G, Terpolilli NA, Trabold R, Loch A, De Zeeuw CI, Weber JT, Maas AI, Plesnila N. Changes of Cerebral Blood Flow during the Secondary Expansion of a Cortical Contusion Assessed by14C-Iodoantipyrine Autoradiography in Mice Using a Non-Invasive Protocol. J Neurotrauma 2008; 25:739-53. [DOI: 10.1089/neu.2007.0480] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Doortje C. Engel
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Günter Mies
- Max-Planck-Institute for Neurological Research, Cologne, Germany
| | - Nicole A. Terpolilli
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Raimund Trabold
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany
| | - Alexander Loch
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - John T. Weber
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andrew I.R. Maas
- Department of Neurosurgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nikolaus Plesnila
- Department of Neurosurgery, University of Munich Medical Center, Munich, Germany
- Institute for Surgical Research, University of Munich Medical Center, Munich, Germany
| |
Collapse
|
60
|
Onyszchuk G, He YY, Berman NEJ, Brooks WM. Detrimental effects of aging on outcome from traumatic brain injury: a behavioral, magnetic resonance imaging, and histological study in mice. J Neurotrauma 2008; 25:153-71. [PMID: 18260798 DOI: 10.1089/neu.2007.0430] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Considerable evidence indicates that outcomes from traumatic brain injury (TBI) are worse in the elderly, but there has been little preclinical research to explore potential mechanisms. In this study, we examined the age-related effects on outcome in a mouse model of controlled cortical impact (CCI) injury. We compared the responses of adult (5-6 months old) and aged (21-24 months old) male mice following a moderate lateral CCI injury to the sensorimotor cortex. Sensorimotor function was evaluated with the rotarod, gridwalk and spontaneous forelimb behavioral tests. Acute edema was assessed from hyperintensity on T2-weighted magnetic resonance images. Blood-brain barrier opening was measured using anti-mouse immunoglobulin G (IgG) immunohistochemistry. Neurodegeneration was assessed by amino-cupric silver staining, and lesion cavity volumes were measured from histological images. Indicators of injury were generally worse in the aged than the adult mice. Acute edema, measured at 24 and 48 h post-injury, resolved more slowly in the aged mice (p < 0.01). Rotarod recovery (p < 0.05) and gridwalk deficits (p < 0.01) were significantly worse in aged mice. There was greater (p < 0.01 at 3 days) and more prolonged post-acute opening of the blood-brain barrier in the aged mice. Neurodegeneration was greater in the aged mice (p < 0.01 at 3 days). In contrast, lesion cavity volumes, measured at 3 days post-injury, were not different between injured groups. These results suggest that following moderate controlled cortical impact injury, the aged brain is more vulnerable than the adult brain to neurodegeneration, resulting in greater loss of function. Tissue loss at the impact site does not explain the increased functional deficits seen in the aged animals. Prolonged acute edema, increased opening of the blood-brain barrier and increased neurodegeneration found in the aged animals implicate secondary processes in age-related differences in outcome.
Collapse
Affiliation(s)
- Gregory Onyszchuk
- Hoglund Brain Imaging Center, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | |
Collapse
|
61
|
Thomale UW, Griebenow M, Mautes A, Beyer TF, Dohse NK, Stroop R, Sakowitz OW, Unterberg AW, Stover JF. Heterogeneous regional and temporal energetic impairment following controlled cortical impact injury in rats. Neurol Res 2008; 29:594-603. [PMID: 17535559 DOI: 10.1179/016164107x166272] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Following traumatic brain injury metabolic stability is impaired. Duration and reversibility of these changes might be important to guide specific interventions. METHODS To characterize temporal and regional changes in cerebral metabolism, 68 male Sprague-Dawley rats were subjected to a focal cortical contusion. Lesion progression and mitochondrial impairment were determined by magnetic resonance imaging (MRI) and triphenyl tetrazolium chloride (TTC) staining, respectively. Metabolic alterations were determined at hours 6 and 24 and day 7 by measuring extracellular glucose, lactate and hypoxanthine levels with microdialysis catheters placed adjacent and distant to the contusion and by quantifying changes in tissue ATP, lactate and glucose using bioluminescence imaging. RESULTS The cortical lesion reached its maximal extent at hour 24 and remained confined to the ipsilateral hemisphere. In microdialysate, at hour 6, extracellular hypoxanthine and lactate reached maximal values, thereafter hypoxanthine normalized while lactate remained increased. Extracellular glucose reached the highest values at hour 24 and remained elevated. Bioluminescence imaging revealed heterogeneous changes in areas distant to the contusion. No significant changes were found in ATP content. Slightly elevated tissue glucose until 24 hours in the ipsilateral hemisphere was observed. Following a continuous increase, lactate levels were the highest by 6 hours in the ipsilateral cortex and hippocampus. DISCUSSION CCI is associated with disturbances in energetic metabolism. Metabolic perturbation is not restricted to the early phase and the contusional region following focal cortical contusion, but also involves hippocampus and primarily uninjured parts of the hemisphere.
Collapse
Affiliation(s)
- Ulrich W Thomale
- Department of Neurosurgery, Charité Campus Virchow, Medical School of Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Diffusion tensor imaging reliably detects experimental traumatic axonal injury and indicates approximate time of injury. J Neurosci 2007; 27:11869-76. [PMID: 17978027 DOI: 10.1523/jneurosci.3647-07.2007] [Citation(s) in RCA: 344] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Traumatic axonal injury (TAI) may contribute greatly to neurological impairments after traumatic brain injury, but it is difficult to assess with conventional imaging. We quantitatively compared diffusion tensor imaging (DTI) signal abnormalities with histological and electron microscopic characteristics of pericontusional TAI in a mouse model. Two DTI parameters, relative anisotropy and axial diffusivity, were significantly reduced 6 h to 4 d after trauma, corresponding to relatively isolated axonal injury. One to 4 weeks after trauma, relative anisotropy remained decreased, whereas axial diffusivity "pseudo-normalized" and radial diffusivity increased. These changes corresponded to demyelination, edema, and persistent axonal injury. At every time point, DTI was more sensitive to injury than conventional magnetic resonance imaging, and relative anisotropy distinguished injured from control mice with no overlap between groups. Remarkably, DTI changes strongly predicted the approximate time since trauma. These results provide an important validation of DTI for pericontusional TAI and suggest novel clinical and forensic applications.
Collapse
|
63
|
Bermpohl D, You Z, Lo EH, Kim HH, Whalen MJ. TNF alpha and Fas mediate tissue damage and functional outcome after traumatic brain injury in mice. J Cereb Blood Flow Metab 2007; 27:1806-18. [PMID: 17406655 DOI: 10.1038/sj.jcbfm.9600487] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tumor necrosis factor-alpha (TNFalpha) and Fas are induced after traumatic brain injury (TBI); however, their functional roles are incompletely understood. Using controlled cortical impact (CCI) and mice deficient in TNFalpha, Fas, or both (TNFalpha/Fas-/-), we hypothesized that TNFalpha and Fas receptor mediate secondary TBI in a redundant manner. Compared with wild type (WT), TNFalpha/Fas-/- mice had improved motor performance from 1 to 4 days (P<0.05), improved spatial memory acquisition at 8 to 14 days (P<0.05), and decreased brain lesion size at 2 and 6 weeks after CCI (P<0.05). Protection in TNFalpha/Fas-/- mice from histopathological and motor deficits was reversed by reconstitution with recombinant TNFalpha before CCI, and TNFalpha-/- mice administered anti-Fas ligand antibodies had improved spatial memory acquisition versus similarly treated WT mice (P<0.05). Tumor necrosis factor-alpha/Fas-/- mice had decreased the numbers of cortical cells with plasmalemma damage at 6 h (P<0.05 versus WT), and reduced matrix metalloproteinase-9 activity in injured brain at 48 and 72 h after CCI. In immature mice subjected to CCI, genetic inhibition of TNFalpha and Fas conferred beneficial effects on histopathology and spatial memory acquisition in adulthood (both P<0.05 versus WT), suggesting that the beneficial effects of TNFalpha/Fas inhibition may be permanent. The data suggest that redundant signaling pathways initiated by TNFalpha and Fas play pivotal roles in the pathogenesis of TBI, and that biochemical mechanisms downstream of TNFalpha/Fas may be novel therapeutic targets to limit neurological sequelae in children and adults with severe TBI.
Collapse
Affiliation(s)
- Daniela Bermpohl
- Neuroscience Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | |
Collapse
|
64
|
Hillary FG, Biswal B. The influence of neuropathology on the FMRI signal: a measurement of brain or vein? Clin Neuropsychol 2007; 21:58-72. [PMID: 17366278 DOI: 10.1080/13854040601064542] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
There is a rapidly growing literature using fMRI technology to investigate the various forms of behavioral impairment associated with brain injury and disease. Given this, surprisingly little work has been conducted to examine the influence of neuropathophysiological processes on the fMRI signal. This paper reviews the literature examining baseline alteration in cerebrovascular parameters associated with normal aging, brain injury, and brain disease. In addition, findings from three cases of individuals with severe brain trauma will be presented to show the influence of brain trauma on baseline cerebrovascular parameters measured by fMRI. The methods used here can be implemented by other investigators to accurately isolate specific hemodynamic changes that can influence the BOLD fMRI signal.
Collapse
Affiliation(s)
- F G Hillary
- Psychology Department, Pennsylvania State University, State College, PA , USA.
| | | |
Collapse
|
65
|
Onyszchuk G, Al-Hafez B, He YY, Bilgen M, Berman NEJ, Brooks WM. A mouse model of sensorimotor controlled cortical impact: characterization using longitudinal magnetic resonance imaging, behavioral assessments and histology. J Neurosci Methods 2006; 160:187-96. [PMID: 17049995 PMCID: PMC1941707 DOI: 10.1016/j.jneumeth.2006.09.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 08/23/2006] [Accepted: 09/01/2006] [Indexed: 10/24/2022]
Abstract
The present study establishes a new mouse model for traumatic brain injury (TBI), using an electromechanically driven linear motor impactor device to deliver a lateral controlled cortical impact (CCI) injury to the sensorimotor cortex. Lesion cavity size was measured, and inter-animal consistency demonstrated, at 14 days post injury. Qualitative information regarding damage progression over time was obtained by scanning with high field magnetic resonance imaging (MRI) at five time points following injury. Functional impairment and recovery were measured with the Rotarod, gridwalk and cylinder tests, and lesion cavity volume was measured post mortem with thionin-stained tissue sections. The study establishes the reliability of a linear-motor based device for producing repeatable damage in a CCI model, demonstrates the power of longitudinal MRI in studying damage evolution, and confirms that a simple battery of functional tests record sensorimotor impairment and recovery.
Collapse
Affiliation(s)
- Gregory Onyszchuk
- Hoglund Brain Imaging Center, University of Kansas Medical Center, 3901 Rainbow Boulevard, MS1052, Kansas City, KS 66160, USA
| | | | | | | | | | | |
Collapse
|
66
|
Igarashi T, Potts MB, Noble-Haeusslein LJ. Injury severity determines Purkinje cell loss and microglial activation in the cerebellum after cortical contusion injury. Exp Neurol 2006; 203:258-68. [PMID: 17045589 DOI: 10.1016/j.expneurol.2006.08.030] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 08/05/2006] [Accepted: 08/10/2006] [Indexed: 10/24/2022]
Abstract
Clinical evidence suggests that the cerebellum is damaged after traumatic brain injury (TBI) and experimental studies have validated these observations. We have previously shown cerebellar vulnerability, as demonstrated by Purkinje cell loss and microglial activation, after fluid percussion brain injury. In this study, we examine the effect of graded controlled cortical impact (CCI) injury on the cerebellum in the context of physiologic and anatomical parameters that have been shown by others to be sensitive to injury severity. Adult male rats received mild, moderate, or severe CCI and were euthanized 7 days later. We first validated the severity of the initial injury using physiologic criteria, including apnea and blood pressure, during the immediate postinjury period. Increasing injury severity was associated with an increased incidence of apnea and higher mortality. Severe injury also induced transient hypertension followed by hypotension, while lower grade injuries produced an immediate and sustained hypotension. We next evaluated the pattern of subcortical neuronal loss in response to graded injuries. There was significant neuronal loss in the ipsilateral cortex, hippocampal CA2/CA3, and laterodorsal thalamus that was injury severity-dependent and that paralleled microglial activation. Similarly, there was a distinctive pattern of Purkinje cell loss and microglial activation in the cerebellar vermis that varied with injury severity. Together, these findings emphasize the vulnerability of the cerebellum to TBI. That a selective pattern of Purkinje cell loss occurs regardless of the type of injury suggests a generalized response that is a likely determinant of recovery and a target for therapeutic intervention.
Collapse
Affiliation(s)
- Takuji Igarashi
- Department of Neurological Surgery, University of California, San Francisco, 521 Parnassus Avenue, Room C-224, San Francisco, CA 94143-0520, USA
| | | | | |
Collapse
|
67
|
Zweckberger K, Erös C, Zimmermann R, Kim SW, Engel D, Plesnila N. Effect of Early and Delayed Decompressive Craniectomy on Secondary Brain Damage after Controlled Cortical Impact in Mice. J Neurotrauma 2006; 23:1083-93. [PMID: 16866621 DOI: 10.1089/neu.2006.23.1083] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The timing of decompressive craniectomy for the treatment of increased intracranial pressure (ICP) after traumatic brain injury (TBI) is a widely discussed clinical issue. Although we showed recently that early decompression is beneficial following experimental TBI, it remains unclear to what degree decompression craniectomy reduces secondary brain damage and if craniectomy is still beneficial when it is delayed by several hours as often inevitable during daily clinical practice. The aim of the current study was therefore to investigate the influence of craniectomy on secondary contusion expansion and brain edema formation and to determine the therapeutic window of craniectomy. Male C57/Bl6 mice were subjected to controlled cortical impact injury. Contusion volume, brain edema formation, and opening of the blood-brain barrier were investigated 2, 6, 12, and 24 h and 7 days after trauma. The effect of decompression craniectomy on secondary brain damage was studied in control mice (closed skull) and in animals craniotomized immediately or with a delay of 1, 3, or 8 h after trauma. Twenty-four hours after trauma, the time point of maximal lesion expansion (+60% vs. 15 min after trauma) and brain edema formation (+3.0% water content vs. sham), contusion volume in craniotomized mice did not show any secondary expansion; that is, contusion volume was similar to that observed in mice sacrificed immediately after trauma (18.3 +/- 5.3 vs. 22.2 +/- 1.4 mm(3)). Furthermore, brain edema formation was reduced by 52% in craniotomized animals. The beneficial effect of craniectomy was still present even when treatment was delayed by up to 3 h after trauma (p < 0.05). The current study clearly demonstrates that early craniectomy prevents secondary brain damage and significantly reduces brain edema formation after experimental TBI. Evaluation of early craniectomy as a therapeutic option after TBI in humans may therefore be indicated.
Collapse
Affiliation(s)
- Klaus Zweckberger
- Laboratory of Experimental Neurosurgery, Department of Neurosurgery and Institute for Surgical Research, University of Munich Medical Center-Grosshadern, Munich, Germany
| | | | | | | | | | | |
Collapse
|
68
|
Statler KD, Alexander H, Vagni V, Dixon CE, Clark RSB, Jenkins L, Kochanek PM. Comparison of seven anesthetic agents on outcome after experimental traumatic brain injury in adult, male rats. J Neurotrauma 2006; 23:97-108. [PMID: 16430376 DOI: 10.1089/neu.2006.23.97] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Isoflurane is commonly used in experimental traumatic brain injury (TBI), both before and early after injury, yet it is rarely used clinically. Narcotics and benzodiazepines are frequently used after injury in clinical TBI. We compared seven anesthetic/sedative agents applied after injury in the controlled cortical impact model: diazepam, fentanyl, isoflurane, ketamine, morphine, pentobarbital, and propofol. Our objective was to provide insight into the relative degrees of neuroprotection provided by these agents in a standard model of TBI. We hypothesized that the choice of anesthetic/sedative early after experimental TBI critically impacts outcome and that the agents most commonly used clinically may be less neuroprotective than isoflurane. Rats treated with isoflurane had the best cognitive recovery (p < 0.05) and hippocampal neuronal survival (p < 0.05). Conversely, rats treated with ketamine had the most hippocampal neuronal death (p < 0.05). Morphine or propofol, two agents commonly used clinically, were associated with the poorest motor function on post-trauma day 1-5 (p < 0.05). Our data support beneficial effects of isoflurane early after experimental TBI. Our data suggest that the early post-TBI use of isoflurane, despite practical logistical issues, could potentially provide clinical benefits in TBI--versus other commonly used sedatives or analgesics. Furthermore, the choice of post-injury sedation and analgesia could have important implications on attempts to translate novel therapies from bench to field or bedside.
Collapse
Affiliation(s)
- Kimberly D Statler
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | | | |
Collapse
|
69
|
Statler KD, Alexander H, Vagni V, Holubkov R, Dixon CE, Clark RSB, Jenkins L, Kochanek PM. Isoflurane exerts neuroprotective actions at or near the time of severe traumatic brain injury. Brain Res 2006; 1076:216-24. [PMID: 16473332 DOI: 10.1016/j.brainres.2005.12.106] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 12/16/2005] [Accepted: 12/16/2005] [Indexed: 10/25/2022]
Abstract
Isoflurane improves outcome vs. fentanyl anesthesia, in experimental traumatic brain injury (TBI). We assessed the temporal profile of isoflurane neuroprotection and tested whether isoflurane confers benefit at the time of TBI. Adult, male rats were randomized to isoflurane (1%) or fentanyl (10 mcg/kg iv bolus then 50 mcg/kg/h) for 30 min pre-TBI. Anesthesia was discontinued, rats recovered to tail pinch, and TBI was delivered by controlled cortical impact. Immediately post-TBI, rats were randomized to 1 h of isoflurane, fentanyl, or no additional anesthesia, creating 6 anesthetic groups (isoflurane:isoflurane, isoflurane:fentanyl, isoflurane:none, fentanyl:isoflurane, fentanyl:fentanyl, fentanyl:none). Beam balance, beam walking, and Morris water maze (MWM) performances were assessed over post-trauma d1-20. Contusion volume and hippocampal survival were assessed on d21. Rats receiving isoflurane pre- and post-TBI exhibited better beam walking and MWM performances than rats treated with fentanyl pre- and any treatment post-TBI. All rats pretreated with isoflurane had better CA3 neuronal survival than rats receiving fentanyl pre- and post-TBI. In rats pretreated with fentanyl, post-traumatic isoflurane failed to affect function but improved CA3 neuronal survival vs. rats given fentanyl pre- and post-TBI. Post-traumatic isoflurane did not alter histopathological outcomes in rats pretreated with isoflurane. Rats receiving fentanyl pre- and post-TBI had the worst CA1 neuronal survival of all groups. Our data support isoflurane neuroprotection, even when used at the lowest feasible level before TBI (i.e., when discontinued with recovery to tail pinch immediately before injury). Investigators using isoflurane must consider its beneficial effects in the design and interpretation of experimental TBI research.
Collapse
Affiliation(s)
- Kimberly D Statler
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | |
Collapse
|
70
|
Leinenweber SB, Sheng H, Lynch JR, Wang H, Batinić-Haberle I, Laskowitz DT, Crapo JD, Pearlstein RD, Warner DS. Effects of a manganese (III) porphyrin catalytic antioxidant in a mouse closed head injury model. Eur J Pharmacol 2006; 531:126-32. [PMID: 16455070 DOI: 10.1016/j.ejphar.2005.12.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2005] [Revised: 12/14/2005] [Accepted: 12/15/2005] [Indexed: 10/25/2022]
Abstract
Closed head injury induces cerebral oxidative stress. The efficacy of a Mn (III) porphyrin catalytic antioxidant was assessed in a mouse closed head injury model. Mice were subjected to closed head injury and treated 15 min later with an i.v. bolus of vehicle or 3 mg/kg MnTE-2-PyP5+. Aconitase activity, Fluoro-Jade staining, glial fibrillary acidic protein immunoreactivity, and rotarod falling latencies were measured. Closed head injury altered all variables. MnTE-2-PyP5+ had no effect on any variable with the exception of attenuation of aconitase inactivation at 2 h post-closed head injury. In a second experiment, mice received 3 mg/kg or 6 mg/kg MnTE-2-PyP5+ or vehicle i.v. 15 min post-closed head injury. Rotarod and Morris water maze latencies were measured. Closed head injury altered performance in both tests. No statistically significant effect of MnTE-2-PyP5+ was observed. We conclude that single dose MnTE-2-PyP5+ does not alter outcome in this mouse closed head injury model.
Collapse
|
71
|
Kochanek PM, Hendrich KS, Jackson EK, Wisniewski SR, Melick JA, Shore PM, Janesko KL, Zacharia L, Ho C. Characterization of the effects of adenosine receptor agonists on cerebral blood flow in uninjured and traumatically injured rat brain using continuous arterial spin-labeled magnetic resonance imaging. J Cereb Blood Flow Metab 2005; 25:1596-612. [PMID: 15931163 DOI: 10.1038/sj.jcbfm.9600154] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Hypoperfusion after traumatic brain injury may exacerbate damage. Adenosine, a vasodilator, regulates cerebral blood flow (CBF). Treatment with adenosine receptor agonists has shown benefit in experimental CNS trauma; however, their effects on CBF after injury remain undefined. We used magnetic resonance imaging to assess CBF in uninjured rats both early and at 24 h after intrahippocampal administration of either the nonselective adenosine receptor agonist 2-chloroadenosine (2-CA, 12 nmol) or the A(2A)-receptor agonist 2-p-(2-carboxyethyl)-phenethylamino-5'-N-ethylcarbox-amidoadenosine (CGS 21680, 6 nmol). We also assessed the effects of these agents on cerebral metabolic rate for glucose (CMRglu). We then assessed the effect of 2-CA on CBF at 3.5 to 5 h after controlled cortical impact (CCI). Injection of 2-CA into uninjured rat brain produced marked increases in CBF in ipsilateral hippocampus and cortex versus vehicle (P<0.05); CBF increases persisted even at 24 h. Measurement of hippocampal levels of 2-CA showed persistent increases to 24 h. CGS 21680 produced even more marked global increases in CBF than seen with 2-CA (2-6-fold versus vehicle, P<0.05 in 10/12 regions of interest (ROIs)). Neither agonist altered CMRglu versus vehicle. After CCI, 2-CA increased CBF in ipsilateral hippocampal and hemispheric ROIs (P<0.05 versus vehicle), but the response was attenuated at severe injury levels. We report marked increases in CBF after injection of adenosine receptor agonists into uninjured rat brain despite unaltered CMRglu. 2-Chloroadenosine produced enduring increases in CBF in uninjured brain and attenuated posttraumatic hypoperfusion. Future studies of adenosine-related therapies in CNS injury should address the role of CBF.
Collapse
Affiliation(s)
- Patrick M Kochanek
- Safar Center for Resuscitation Research, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Wagner AK, Chen X, Kline AE, Li Y, Zafonte RD, Dixon CE. Gender and environmental enrichment impact dopamine transporter expression after experimental traumatic brain injury. Exp Neurol 2005; 195:475-83. [PMID: 16023635 DOI: 10.1016/j.expneurol.2005.06.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Revised: 06/03/2005] [Accepted: 06/15/2005] [Indexed: 11/26/2022]
Abstract
Dopamine (DA) systems are implicated in cognitive deficits following traumatic brain injury (TBI). Rodent studies have demonstrated that both environmental enrichment (EE) and sex hormones can influence DA systems. The dopamine transporter (DAT) plays a crucial role in regulating DA transmission, and previous work shows that DAT is decreased after TBI in males. Therefore, the purpose of this study was to examine the effects of gender and EE on frontal cortex and striatal DAT expression after TBI. Sprague-Dawley male (n = 24) and cycling female rats (n = 24) were placed into EE or standard housing after controlled cortical impact (2.7 mm, 4.0 m/s) injury or sham surgery (eight groups, n = 6/group). Four weeks post-surgery, bilateral frontal cortex and striatal DAT expression was examined via Western blot. Results demonstrated that there was a significant effect of injury, EE, and region on DAT expression (P < 0.05 all comparisons) on female groups. There were no significant DAT decreases in any region as a result of injury, however, EE did promote significant post-injury DAT decreases in the striatum and ipsilateral frontal cortex (P < 0.05 all comparisons) compared to female shams housed in the standard environment. For males, there was a significant effect of injury, EE, and region for male groups (P < 0.05 all comparisons). There were decreases in DAT expression in three regions studied for injured males housed in the standard environment compared to sham males in the standard environment (P < 0.05 all comparisons), however, EE did not add significantly to post-injury DAT decreases in these regions. These results suggest that CCI causes larger relative decreases in DAT expression for males compared to females and that treatment with EE has larger effects on post-injury DAT expression for females than males. These findings may have some relevance to treatment paradigms using dopaminergic neurostimulants after TBI.
Collapse
Affiliation(s)
- Amy K Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | |
Collapse
|
73
|
Morales DM, Marklund N, Lebold D, Thompson HJ, Pitkanen A, Maxwell WL, Longhi L, Laurer H, Maegele M, Neugebauer E, Graham DI, Stocchetti N, McIntosh TK. Experimental models of traumatic brain injury: do we really need to build a better mousetrap? Neuroscience 2005; 136:971-89. [PMID: 16242846 DOI: 10.1016/j.neuroscience.2005.08.030] [Citation(s) in RCA: 240] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Revised: 06/08/2005] [Accepted: 08/04/2005] [Indexed: 11/19/2022]
Abstract
Approximately 4000 human beings experience a traumatic brain injury each day in the United States ranging in severity from mild to fatal. Improvements in initial management, surgical treatment, and neurointensive care have resulted in a better prognosis for traumatic brain injury patients but, to date, there is no available pharmaceutical treatment with proven efficacy, and prevention is the major protective strategy. Many patients are left with disabling changes in cognition, motor function, and personality. Over the past two decades, a number of experimental laboratories have attempted to develop novel and innovative ways to replicate, in animal models, the different aspects of this heterogenous clinical paradigm to better understand and treat patients after traumatic brain injury. Although several clinically-relevant but different experimental models have been developed to reproduce specific characteristics of human traumatic brain injury, its heterogeneity does not allow one single model to reproduce the entire spectrum of events that may occur. The use of these models has resulted in an increased understanding of the pathophysiology of traumatic brain injury, including changes in molecular and cellular pathways and neurobehavioral outcomes. This review provides an up-to-date and critical analysis of the existing models of traumatic brain injury with a view toward guiding and improving future research endeavors.
Collapse
Affiliation(s)
- D M Morales
- Traumatic Brain Injury Laboratory, Department of Neurosurgery, University of Pennsylvania, 3320 Smith Walk, 105C Hayden Hall, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Wagner AK, Sokoloski JE, Ren D, Chen X, Khan AS, Zafonte RD, Michael AC, Dixon CE. Controlled cortical impact injury affects dopaminergic transmission in the rat striatum. J Neurochem 2005; 95:457-65. [PMID: 16190869 DOI: 10.1111/j.1471-4159.2005.03382.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The therapeutic benefits of dopamine (DA) agonists after traumatic brain injury (TBI) imply a role for DA systems in mediating functional deficits post-TBI. We investigated how experimental TBI affects striatal dopamine systems using fast scan cyclic voltammetry (FSCV), western blot, and d-amphetamine-induced rotational behavior. Adult male Sprague-Dawley rats were injured by a controlled cortical impact (CCI) delivered unilaterally to the parietal cortex, or were naïve controls. Amphetamine-induced rotational behavior was assessed 10 days post-CCI. Fourteen days post-CCI, animals were anesthetized and underwent FSCV with bilateral striatal carbon fiber microelectrode placement and stimulating electrode placement in the medial forebrain bundle (MFB). Evoked DA overflow was assessed in the striatum as the MFB was electrically stimulated at 60 Hz for 10 s. In 23% of injured animals, but no naïve animals, rotation was observed with amphetamine administration. Compared with naïves, striatal evoked DA overflow was lower for injured animals in the striatum ipsilateral to injury (p < 0.05). Injured animals exhibited a decrease in V(max) (52% of naïve, p < 0.05) for DA clearance in the hemisphere ipsilateral to injury compared with naïves. Dopamine transporter (DAT) expression was proportionally decreased in the striatum ipsilateral to injury compared with naïve animals (60% of naïve, p < 0.05), despite no injury-related changes in vesicular monoamine transporter or D2 receptor expression (DRD2) in this region. Collectively, these data appear to confirm that the clinical efficacy of dopamine agonists in the treatment of TBI may be related to disruptions in the activity of subcortical dopamine systems.
Collapse
Affiliation(s)
- A K Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
Magnetic resonance imaging (MRI) continues to have a large impact on the diagnosis and management of a number of diseases, especially diseases associated with brain injury. The strengths of MRI are the unique contrast that can be obtained, and the fact that it is not harmful and that it can be readily applied to human and animal models. The past decade has seen development of functional MRI techniques that measure aspects of hemodynamics and water diffusion that are playing an important role. Indeed, these techniques are having a major impact on management of brain injury. The development of MRI continues at a rapid pace and a renewed push to increased spatial and temporal resolution will extend the applicability of anatomical and functional MRI. Increased interest in molecular imaging using MRI is increasing the number of processes that can be imaged in the brain. This work reviews some new developments that are being made in anatomical, functional, and molecular MRI of the brain, with comments about usefulness for work in the area of neuroprotection.
Collapse
Affiliation(s)
- Alan P Koretsky
- Laboratory of Functional and Molecular Imaging, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20812, USA.
| |
Collapse
|
76
|
Thomale UW, Griebenow M, Kroppenstedt SN, Unterberg AW, Stover JF. Small volume resuscitation with HyperHaes improves pericontusional perfusion and reduces lesion volume following controlled cortical impact injury in rats. J Neurotrauma 2005; 21:1737-46. [PMID: 15684765 DOI: 10.1089/neu.2004.21.1737] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The hyperosmolar and hyperoncotic properties of HyperHaes (HHES) might improve impaired posttraumatic cerebral perfusion. Possible beneficial effects on pericontusional perfusion, brain edema, and contusion volume were investigated in rats subjected to controlled cortical impact (CCI). Male Sprague-Dawley rats (n = 60) anesthetized with isoflurane were subjected to a left temporoparietal CCI. Thereafter, rats were randomized to receive HHES (10% hydroxyethylstarch, 7.5% NaCl) or physiological saline solution (4 mL/kg body weight) intravenously. Mean arterial blood pressure (MABP) and intracranial pressure (ICP) were determined before and following CCI, after drug administration and 24 h later. Regional pericontusional cortical perfusion was determined by scanning laser Doppler flowmetry before CCI, and 30 min, 4 and 24 h after injury. At 24 h brain swelling and water content were measured gravimetrically. At 7 days, cortical contusion volume was determined planimetrically. MABP was not influenced by HHES. ICP was significantly decreased immediately after HHES infusion (5.7 +/- 0.4 vs. 7.1 +/- 1.0 mm Hg; p < 0.05). Pericontusional cortical perfusion was significantly decreased by 44% compared to pre-injury levels (p < 0.05). HHES significantly improved cortical perfusion at 4 h after CCI, approaching baseline values (85 +/- 12%). While increased posttraumatic brain edema was not reduced by HHES at 24 h, cortical contusion volume was significantly decreased in the HHES-treated rats at 7 days after CCI (23.4 +/- 3.5 vs. 39.6 +/- 6.2 mm3; p < 0.05). Intravaneous administration of HHES within 15 min after CCI has a neuroprotective potential, as it significantly attenuated impaired pericontusional perfusion and markedly reduced the extent of induced structural damage.
Collapse
Affiliation(s)
- Ulrich-Wilhelm Thomale
- Department of Neurosurgery, Charité, Virchow Medical Center, Humboldt University Berlin, Germany.
| | | | | | | | | |
Collapse
|
77
|
Rennaker RL, Street S, Ruyle AM, Sloan AM. A comparison of chronic multi-channel cortical implantation techniques: manual versus mechanical insertion. J Neurosci Methods 2005; 142:169-76. [PMID: 15698656 DOI: 10.1016/j.jneumeth.2004.08.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Revised: 08/05/2004] [Accepted: 08/05/2004] [Indexed: 10/26/2022]
Abstract
High-density multi-channel intra-cortical electrode arrays allow researchers to record simultaneously from populations of neurons for the purpose of understanding neural coding and plasticity. These devices have tens to hundreds of electrodes spaced within a few square millimeters. During insertion, the high-density probes can compress the cortex several millimeters prior to breaking through the pia. Compression of cortical tissue has been demonstrated to result in traumatic brain injury (TBI) which may be a major contributor to low electrode yield and decreased recording longevity. Two insertion techniques for chronically implanting multi-wire electrode arrays in layer IV of primary auditory cortex were compared. A mechanical insertion device, capable of rapidly inserting the electrode array without visible compression of the brain, was constructed. The neural responses to broadband clicks and pure tones recorded from the arrays inserted with the mechanical device were compared to the results from a manual insertion method using a micromanipulator. Both techniques result in a similar number of active channels directly following surgery with a mean signal-to-noise ratio of approximately 4.5. Over 60% of the animals implanted with the mechanical insertion device had driven activity at week 6 whereas none of the animals with manually inserted arrays exhibited functional responses after 3 weeks. This report provides initial evidence that mechanical insertion devices, which prevent cortical compression, increase electrode recording longevity.
Collapse
Affiliation(s)
- R L Rennaker
- Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, OK 73019, USA.
| | | | | | | |
Collapse
|
78
|
Knoblach SM, Alroy DA, Nikolaeva M, Cernak I, Stoica BA, Faden AI. Caspase inhibitor z-DEVD-fmk attenuates calpain and necrotic cell death in vitro and after traumatic brain injury. J Cereb Blood Flow Metab 2004; 24:1119-32. [PMID: 15529012 DOI: 10.1097/01.wcb.0000138664.17682.32] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In studies designed to evaluate the therapeutic window for treatment of traumatic brain injury, the caspase 3 inhibitor z-DEVD-fmk improved neurologic function and reduced lesion volumes when administered at 1 but not at 4, 8, or 24 hours after injury. Moreover, neither caspase 3 nor PARP, a caspase 3 substrate, were cleaved in injured, untreated cortex from 1 to 72 hours after injury. Few cortical neurons expressed active caspase 3 or were TUNEL positive from 6 to 24 hours after injury, and TUNEL staining was primarily Type I (necrotic). Nissl staining revealed extensive neuronal necrosis in the injured cortex from 6 to 24 hours after impact. Considered together, these data suggested that z-DEVD-fmk may reduce neuronal necrosis, so we used an in vitro model of necrotic cell death induced by maitotoxin to test this further and explore the potential mechanism(s) involved. Z-DEVD-fmk (1 nM-100 microM) significantly attenuated maitotoxin induced neuronal cell death and markedly reduced expression of the 145 kD calpain-mediated alpha-spectrin breakdown product after maitotoxin injury. Neither the 120 kD caspase-mediated alpha-spectrin cleavage product nor cathepsin B were expressed after maitotoxin injury. In a cell free assay, z-DEVD-fmk reduced hydrolysis of casein by purified calpain I. Finally, z-DEVD-fmk reduced expression of the 145 kD calpain-mediated alpha-spectrin cleavage fragment after traumatic brain injury in vivo. These data suggest that neuroprotection by z-DEVD-fmk may, in part, reflect inhibition of calpain-related necrotic cell death.
Collapse
Affiliation(s)
- Susan M Knoblach
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA.
| | | | | | | | | | | |
Collapse
|
79
|
Stoffel M, Blau C, Reinl H, Breidt J, Gersonde K, Baethmann A, Plesnila N. Identification of brain tissue necrosis by MRI: validation by histomorphometry. J Neurotrauma 2004; 21:733-40. [PMID: 15253801 DOI: 10.1089/0897715041269678] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The volume of an experimental necrotic lesion of the cortex expands up to 400% of its initial size within the first 24 h after the insult. Lesion expansion, a clinically well known phenomenon, is often accompanied by perifocal brain edema and consequently difficult to image and to analyze by magnetic resonance imaging (MRI). Therefore we aimed to validate a T(2)-weighted spin echo sequence upon its ability to distinguish necrotic from edematous brain tissue. Male Sprague-Dawley rats (n = 5 per group) were subjected to a cortical freezing lesion leading to immediate tissue necrosis with subsequent perifocal vasogenic brain edema. Immediately and 4, 12, and 24 h after the lesion the maximal area of necrosis was quantified longitudinally by coronal T(2)-weighted spin echo MRI-scans. After the last scan, animals were sacrificed for direct comparison of the lesion area obtained by MRI and histomorphometry. In parallel groups of animals, lesion expansion was quantified by histology. The acquired T(2)-maps clearly distinguish the cortical necrosis from perifocal edema and healthy brain. Focal freezing led to a cortical lesion of 5.24 +/- 0.36 mm(2) immediately after trauma (0 h; 100%) which expanded progressively to a maximum of 6.82 +/- 0.34 mm(2) after 24 h (131%; *p < 0.01 vs. 0 h). Lesion expansion quantified by histology was almost identical (132% within 24 h). Histological assessment resulted in smaller absolute lesion areas compared to MRI, most likely due to shrinking during tissue processing (4.72 +/- 0.26 mm(2) vs. 6.82 +/- 0.34 mm(2), p < 0.01). The current study shows that necrotic brain tissue can be distinguished from surrounding brain edema by T(2)-mapping. The technique is sensitive enough to detect small changes in necrosis expansion in vivo as validated by histology. The presented technique may be a useful future tool for the non-invasive identification of necrotic brain tissue following brain injury (e. g., from trauma or ischemia).
Collapse
Affiliation(s)
- Michael Stoffel
- Department of Neurosurgery, Rheinische Friedrich-Wilhelms-University, Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
80
|
Chen SF, Richards HK, Smielewski P, Johnström P, Salvador R, Pickard JD, Harris NG. Relationship between flow-metabolism uncoupling and evolving axonal injury after experimental traumatic brain injury. J Cereb Blood Flow Metab 2004; 24:1025-36. [PMID: 15356423 DOI: 10.1097/01.wcb.0000129415.34520.47] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Blood flow-metabolism uncoupling is a well-documented phenomenon after traumatic brain injury, but little is known about the direct consequences for white matter. The aim of this study was to quantitatively assess the topographic interrelationship between local cerebral blood flow (LCBF) and glucose metabolism (LCMRglc) after controlled cortical impact injury and to determine the degree of correspondence with the evolving axonal injury. LCMRglc and LCBF measurements were obtained at 3 hours in the same rat from 18F-fluorodeoxyglucose and 14C-iodoantipyrine coregistered autoradiographic images, and compared to the density of damaged axonal profiles in adjacent sections and in an additional group at 24 hours using beta-amyloid precursor protein (beta-APP) immunohistochemistry. LCBF was significantly reduced over the ipsilateral hemisphere by 48 +/- 15% compared with sham-controls, whereas LCMRglc was unaffected, apart from foci of elevated LCMRglc in the contusion margin. Flow-metabolism was uncoupled, indicated by a significant 2-fold elevation in the LCMRglc/LCBF ratio within most ipsilateral structures. There was a significant increase in beta-APP-stained axons from 3 to 24 hours, which was negatively correlated with LCBF and positively correlated with the LCMRglc/LCBF ratio at 3 hours in the cingulum and corpus callosum. Our study indicates a possible dependence of axonal outcome on flow-metabolism in the acute injury stage.
Collapse
Affiliation(s)
- Szu-Fu Chen
- Academic Neurosurgery, Center for Brain Repair, University of Cambridge, Robinson Way, UK
| | | | | | | | | | | | | |
Collapse
|
81
|
Lundblad C, Grände PO, Bentzer P. A Mouse Model for Evaluation of Capillary Perfusion, Microvascular Permeability, Cortical Blood Flow, and Cortical Edema in the Traumatized Brain. J Neurotrauma 2004; 21:741-53. [PMID: 15253802 DOI: 10.1089/0897715041269614] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genetically engineered mice have successfully been used to investigate molecular and cellular mechanisms associated with cell dysfunction following brain trauma. Such animals may also offer a possibility to investigate mechanisms involved in posttraumatic hemodynamic alterations. The objective of the study was to establish a mouse model in which important hemodynamic alterations following trauma could be analyzed. C57/BL6 male mice were subjected to controlled cortical impact injury (CCI) or sham-injury. Distribution of blood flow was estimated by determining number of perfused capillaries using FITC-dextran as an intravascular marker. Cortical blood flow was measured using [(14)C]-iodoantipyrine, brain water content (BWC) was measured using a wet vs. dry weight method, and permeability surface area product (PS) was estimated by the transfer constant for [(51)Cr]-EDTA. Number of perfused capillaries in the contusion area was progressively reduced during the first 24 h following trauma by at most 60% relative to a value of 329 +/- 61/mm(2) in sham-injured animals. Blood flow in the contusion area decreased simultaneously by at most 50% relative to a control value of 1.8 +/- 0.4 mL.min(-1).g(-1), and was reduced further in subregions within the contusion area. BWC in the injured hemisphere increased from 79.3 +/- 0.5% at control to at most 79.9 +/- 0.6% at 24 h post trauma. PS in the injured hemisphere increased by 71% at 3 h post trauma relative to a control value of 0.45 +/- 0.1 microL.min(-1).g(-1), and was close to control at 24 h. The present study demonstrates that brain trauma in addition to a reduction in cortical blood flow, reduces number of perfused capillaries, which most likely affects exchange of nutrients and fluid. The CCI in mouse is likely to be a useful tool to elucidate mechanisms involved in hemodynamic alterations following brain trauma.
Collapse
Affiliation(s)
- C Lundblad
- Department of Physiological Sciences, University of Lund and Lund University Hospital, Lund, Sweden.
| | | | | |
Collapse
|
82
|
Hoover RC, Motta M, Davis J, Saatman KE, Fujimoto ST, Thompson HJ, Stover JF, Dichter MA, Twyman R, White HS, McIntosh TK. Differential Effects of the Anticonvulsant Topiramate on Neurobehavioral and Histological Outcomes following Traumatic Brain Injury in Rats. J Neurotrauma 2004; 21:501-12. [PMID: 15165359 DOI: 10.1089/089771504774129847] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The efficacy of topiramate, a novel therapeutic agent approved for the treatment of seizure disorders, was evaluated in a model of traumatic brain injury (TBI). Adult male rats were anesthetized (sodium pentobarbital, 60 mg/kg, i.p.), subjected to lateral fluid percussion brain injury (n = 60) or sham injury (n = 47) and randomized to receive either topiramate or vehicle at 30 min (30 mg/kg, i.p.), and 8, 20 and 32 h postinjury (30 mg/kg, p.o.). In Study A, memory was evaluated using a Morris water maze at 48 h postinjury, after which brain tissue was evaluated for regional cerebral edema. In Study B, animals were evaluated for motor function at 48 h and 1, 2, 3, and 4 weeks postinjury using a composite neuroscore and the rotating pole test and for learning ability at 4 weeks. Brains were analyzed for hemispheric tissue loss and hippocampal CA3 cell loss. Topiramate had no effect on posttraumatic cerebral edema or histologic damage when compared to vehicle. At 48 h, topiramate treatment improved memory function in sham but not brain-injured animals, while at one month postinjury it impaired learning performance in brain-injured but not sham animals. Topiramate significantly improved composite neuroscores at 4 weeks postinjury and rotating pole performance at 1 and 4 weeks postinjury, suggesting a potentially beneficial effect on motor function following TBI.
Collapse
Affiliation(s)
- Rachel C Hoover
- Head Injury Center, Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania 19103, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Alessandri B, Heimann A, Filippi R, Kopacz L, Kempski O. Moderate controlled cortical contusion in pigs: effects on multi-parametric neuromonitoring and clinical relevance. J Neurotrauma 2004; 20:1293-305. [PMID: 14748978 DOI: 10.1089/089771503322686094] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Over the last decade, routine neuromonitoring of ICP and CPP has been extended with new on-line techniques such as microdialysis, tissue oxygen (ptiO(2)), acid-base balance (ptiCO(2), pH) and CBF measurements, which so far have not lead to clear-cut therapy approaches in the neurointensive care unit. This is partially due to the complex pathophysiology following a wide-range of brain injuries, and the lack of suitable animal models allowing simultaneous, clinically relevant neuromonitoring under controlled conditions. Therefore, a controlled cortical impact (CCI) model in large animals (pig) has been developed. After placement of microdialysis, ptiO(2), temperature and ICP catheters, an unilateral CCI injury (2.6-2.8 m/sec velocity, 9 mm depth, 400 ms dwell time) was applied and neuromonitoring continued for 10 h. CCI caused a rapid drop in CPP, ptiO(2) and glucose, whereas ICP, glutamate and lactate increased significantly. Most parameters returned to baseline values within hours. Lactate stayed elevated significantly throughout the experiment, but the lactate-to-pyruvate ratio (LPR) changed only slightly, indicating no severely ischemic CBF. Contralateral parameters were not affected significantly. Evaluation of brain water content and histology (12 h post-CCI) showed ipsilateral brain swelling by 5% and massive cell damage underneath the injury site which correlated with changes of ICP, CPP, glutamate, lactate, and ptiO(2) within the first hours post-CCI. Moderate controlled cortical contusion in pigs induced a complex pattern of pathophysiological processes which led to 'early' histological damage. Thus, this new large animal model will enable us to investigate the effect of therapeutic interventions on multi-parametric neuromonitoring and histological outcome, and to translate the data into clinical practice.
Collapse
Affiliation(s)
- Beat Alessandri
- Institute for Neurosurgical Pathophysiology, Johannes Gutenberg-University Mainz, Mainz, Germany.
| | | | | | | | | |
Collapse
|
84
|
Wagner AK, Willard LA, Kline AE, Wenger MK, Bolinger BD, Ren D, Zafonte RD, Dixon CE. Evaluation of estrous cycle stage and gender on behavioral outcome after experimental traumatic brain injury. Brain Res 2004; 998:113-21. [PMID: 14725974 DOI: 10.1016/j.brainres.2003.11.027] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Female sex hormones are acutely neuroprotective in experimental models of traumatic brain injury (TBI). Because hormonal profiles are known to vary with estrous cycle stage, the purpose of this study was to evaluate how pre-injury estrous stage affects motor and cognitive performance after experimental TBI. We also sought to compare post-injury behavioral performance in males vs. females. Under anesthesia, male (n=18) and female (n=35) Sprague-Dawley rats underwent either controlled cortical impact (CCI) injury (2.7 mm; 4 m/s) or sham operations. Females were grouped according to estrous stage (proestrous or non-proestrous) at the time of surgery. Motor function was assessed pre-injury and for the first 5 days after surgery using beam balance and walking tasks. Spatial memory was assessed beginning 14 days post-injury utilizing the Morris water maze (MWM) task. No significant differences were found on any task between injured females regardless of estrous cycle stage. Females performed significantly better than males on both motor tasks, but gender did not influence MWM performance. Mixed effects multivariate analysis corroborated these results by showing that pre-injury serum hormone levels had little affect on behavioral performance. The results suggest that the presence of endogenous circulating hormones, rather than hormonal status at time of injury, may confer early neuroprotection in females after TBI. The impact of early neuroprotection on later behavioral outcome and the anatomic structural specificity of hormonal neuroprotection require further study.
Collapse
Affiliation(s)
- Amy K Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, 3471 Fifth Avenue, Suite 201 Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Statler KD, Janesko KL, Melick JA, Clark RSB, Jenkins LW, Kochanek PM. Hyperglycolysis is exacerbated after traumatic brain injury with fentanyl vs. isoflurane anesthesia in rats. Brain Res 2003; 994:37-43. [PMID: 14642446 DOI: 10.1016/j.brainres.2003.09.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Despite common use of narcotics in the clinical management of severe traumatic brain injury (TBI), in experimental models rats treated with fentanyl have exhibited worse functional outcome and more CA1 hippocampal death than rats treated with standard isoflurane anesthesia. We hypothesized that greater post-traumatic excitotoxicity, reflected by cerebral glucose utilization (CMRglu), may account for detrimental effects of fentanyl vs. isoflurane. Rats were anesthetized with either isoflurane (1% by inhalation) or fentanyl (10 mcg/kg iv bolus then 50 mcg/kg/h infusion). 14C-deoxyglucose autoradiography was performed 45 min after controlled cortical impact (CCI) to left parietal cortex (n=4 per anesthetic group) or in uninjured rats after 45 min of anesthesia (n=3 per anesthetic group). Uninjured rats treated with fentanyl vs. isoflurane showed 35-45% higher CMRglu in all brain structures (p<0.05) except CA3. After TBI in rats treated with isoflurane, CMRglu increased significantly only in ipsilateral CA1 and ipsilateral parietal cortex (p<0.05 vs. isoflurane uninjured). Conversely, after TBI in rats treated with fentanyl, CMRglu increased markedly and bilaterally in CA1 and CA3 (p<0.05 vs. fentanyl uninjured), but not ipsilateral parietal cortex. In contralateral CA1, CMRglu was nearly two times greater after TBI in fentanyl vs. isoflurane treated rats (p<0.05). Hyperglycolysis was exacerbated in CA1 and CA3 hippocampus after TBI in rats treated with fentanyl vs. isoflurane anesthesia. This post-traumatic hyperglycolysis suggests greater excitotoxicity and concurs with reports of worse functional outcome and more CA1 hippocampal death after TBI with fentanyl vs. isoflurane anesthesia.
Collapse
Affiliation(s)
- Kimberly D Statler
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, 3434 Fifth Avenue, Pittsburgh, PA 15260, USA
| | | | | | | | | | | |
Collapse
|
86
|
Rogatsky GG, Sonn J, Kamenir Y, Zarchin N, Mayevsky A. Relationship between Intracranial Pressure and Cortical Spreading Depression following Fluid Percussion Brain Injury in Rats. J Neurotrauma 2003; 20:1315-25. [PMID: 14748980 DOI: 10.1089/089771503322686111] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) is known to be accompanied by an increase in intracranial pressure (ICP) and in some cases, by spontaneous generation of cortical spreading depression (CSD) cycles. However, the role of CSD in the pathophysiology of cerebral contusion is still unknown. A multiparametric monitoring assembly was placed on the right hemisphere of the rat brain to evaluate ICP, DC potential, extracellular K(+), cerebral blood flow (CBF), and electrocorticogram in 27 rats during 5 h. Fluid percussion brain injury (FPBI) with the magnitude of the impact 2.9, 3.3, 4.1, and 5.0 atmospheres was induced to the left parietal cortex in animal groups A, B, C, and D, respectively. A slow increase in ICP was evident, and was pronounced in group C and especially in group D, where four of nine animals died during the monitoring. At the end of the 5 h experiment, the mean ICP levels were 6.75 +/- 2.87, 8.40 +/- 2.70, 12.75 +/- 4.03, 29.56 +/- 9.25, and the mean total number of CSD cycles was 2.00 +/- 1.41, 4.29 +/- 4.23, 11.71 +/- 13.29, and 20.11 +/- 19.26 in groups A, B, C, and D, respectively. The maximal level of intensity of CSD cycle generation after FPBI was obtained in group D, where almost constant activity was maintained until the end of the experiment. A significant coefficient of correlation between ICP level and total number of CSD cycles was found for all ICP measurements (r = 0.47-0.63, p < 0.05, n = 27), however more significant (p < 0.001) was the coefficient during the period of monitoring between 2 and 4 h after FPBI. Our results suggest that numerous repeating CSD cycles are typical phenomena in moderate and especially severe forms of FPBI. The rising number of CSD cycles under condition of an ICP level >/=20 mm Hg may demonstrate, with high probability, the unfavorable development of TBI, caused by growing secondary hypoxic insult.
Collapse
Affiliation(s)
- G G Rogatsky
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | | | | |
Collapse
|
87
|
Van Landeghem FKH, Schreiber S, Unterberg AW, Von Deimling A, Stover JF. Differential Concentration-Dependent Effects of Prolonged Norepinephrine Infusion on Intraparenchymal Hemorrhage and Cortical Contusion in Brain-Injured Rats. J Neurotrauma 2003; 20:1327-37. [PMID: 14748981 DOI: 10.1089/089771503322686120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Under clinical conditions catecholamines are infused to elevate cerebral perfusion pressure and improve impaired posttraumatic cerebral microcirculation. This, however, is associated with the risk of additional hemorrhage in the acute phase following traumatic brain injury. In the present study we investigated the dose-dependent effects of prolonged norepinephrine infusion on arterial blood pressure, blood glucose, and structural damage in brain-injured rats. At 4 h following induction of a focal cortical contusion (CCI), 40 rats were randomized to receive low (0.15), medium (0.3), or high dose (1 microg/kg/min) norepinephrine. Control rats were given equal volume of NaCl. Norepinephrine and NaCl were infused intravenously via Alzet osmotic pumps for 44 h. Mean arterial blood pressure (MABP), blood gases and blood glucose were determined before, at 4, 24, 48 h after CCI in repeatedly anesthetized rats (n = 28). Systolic arterial blood pressure (SABP) was measured using the tail cuff method in awake, restrained rats (n = 12). Cortical contusion and intraparenchymal hemorrhage volume were quantified at 48 h in all rats. MABP determined in anesthetized rats was only marginally increased. SABP was significantly elevated during infusion of medium and high dose norepinephrine in awake rats, exceeding 140 mm Hg. Medium and high dose norepinephrine significantly increased cortical hemorrhage by 157% and 142%, without increasing the cortical contusion volume. Low dose norepinephrine significantly reduced the cortical contusion by 44%. Norepinephrine aggravates the underlying brain damage during the acute posttraumatic phase. Future studies are needed to determine the least deleterious norepinephrine concentration.
Collapse
Affiliation(s)
- Frank K H Van Landeghem
- Institute of Neuropathology, Charité Campus Virchow Klinikum, Humboldt-University, 13353 Berlin, Germany.
| | | | | | | | | |
Collapse
|
88
|
Schuhmann MU, Stiller D, Skardelly M, Bernarding J, Klinge PM, Samii A, Samii M, Brinker T. Metabolic changes in the vicinity of brain contusions: a proton magnetic resonance spectroscopy and histology study. J Neurotrauma 2003; 20:725-43. [PMID: 12965052 DOI: 10.1089/089771503767869962] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Proton MR spectroscopy (1H-MRS) has been previously used to monitor metabolic changes in areas of diffuse brain injury. We studied metabolism in the close vicinity of experimental traumatic brain contusions and remote on the contralateral side from 1h to 28d post-injury. Changes of creatine and phosphocreatine (Cr&PCr), N-acetylaspartate (NAA), choline (Cho), inositol (Ino), taurine (Tau), glutamate (Glu), and lactate (Lac) were assessed and compared to neuronal, glial and inflammatory changes in histology. In the pericontusional zone Cr&PCr, NAA, and Glu decreased immediately after trauma by -35%, -60%, and -37%, respectively, related to primary cell disintegration and secondary perturbations as reflected in histology. These metabolites partially recovered at 7d (-15%, -37%, and -21% respectively), in parallel to indicators of repair in immunhistochemistry. Control levels were not regained at 28d, in correlation to a decrease of viable neurons. Cho and Ino, initially lowered by -26% and -31% respectively, increased at 7d by +74% and 31%, reflecting glial activation and proliferation. The signal including the lactate resonance increased by >1000% with a maximum at 7d, possibly related to energy failure, inflammation and glial activation. A partial contribution of lipids to this signal cannot be fully excluded. The contralateral side showed mild astroglial activation in histology, but no changes in 1H-MRS. The study demonstrates the feasibility of volume selective 1H-MRS using the LCModel (Linear Combination of Model in vitro spectra of metabolites solutions) to monitor metabolic changes close to focal traumatic lesions and suggests how metabolic alterations can be differentiated in cause.
Collapse
Affiliation(s)
- Martin Ulrich Schuhmann
- Department of Pediatric Neurosurgery, Wayne State University School of Medicine, Children's Hospital of Michigan, Detroit, Michigan, USA.
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Plesnila N, Friedrich D, Eriskat J, Baethmann A, Stoffel M. Relative cerebral blood flow during the secondary expansion of a cortical lesion in rats. Neurosci Lett 2003; 345:85-8. [PMID: 12821177 DOI: 10.1016/s0304-3940(03)00396-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The size of a cerebral contusion is not finite at the moment of trauma, but liable to secondary increase during the following hours and days. In the present study we investigated whether this phenomenon may be related to changes in cortical blood flow (cCBF). In rats a cortical lesion grew to 140% of its initial volume during the first 24 h after injury. During the time of most rapid lesion expansion (<6 h after the insult) marked hypoperfusion (approximately 30% of baseline) was found in the ipsilateral hemisphere by laser Doppler scanning fluxmetry. In the peri-contusional area cCBF slowly recovered to approximately 80% of baseline, while in the distant brain not affected by delayed cell death, significant hyperperfusion (approximately 160% of baseline) was observed. Thus, early hypoperfusion might be an important mechanism for secondary lesion expansion.
Collapse
Affiliation(s)
- Nikolaus Plesnila
- Institute for Surgical Research, Ludwig-Maximilians University, Marchioninistrasse 15, 81366 Munich, Germany.
| | | | | | | | | |
Collapse
|
90
|
Bentzer P, Venturoli D, Carlsson O, Grände PO. Low-dose prostacyclin improves cortical perfusion following experimental brain injury in the rat. J Neurotrauma 2003; 20:447-61. [PMID: 12803977 DOI: 10.1089/089771503765355522] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
It was recently shown that prostacyclin at a low dose reduces cortical cell death following brain trauma in the rat. Conceivably, prostacyclin with its vasodilatory, anti-aggregatory, anti-adhesive and permeability-reducing properties improved a compromised perfusion caused by post-traumatic vasoconstriction, microthrombosis and increased microvascular permeability. The objective of the present study was therefore to investigate the hemodynamic effects of low-dose prostacyclin in the traumatized rat cortex. Following a fluid percussion brain injury or a sham procedure, animals were treated with a continuous intravenous infusion of prostacyclin of 1 or 2 ng x kg(-1) x min(-1), or vehicle. Blood flow ([(14)C]-iodoantipyrine), the permeability-surface area product (PS) for [(51)Cr]-EDTA, and brain water content were measured after 3 or 48 h of treatment. Blood flow values in the injured cortex were transiently reduced to 0.42 +/- 0.2 mL x min(-1) in the vehicle group 3 h following trauma from a corresponding value of about 1.6 mL x min(-1) in the sham group, with recovery of blood flow after 48 h. Prostacyclin treatment caused a dose-dependent increase in blood flow which reached statistical significance 48 h following trauma. Brain water content and PS increased in the injured cortex post trauma and the higher dose of prostacyclin increased these parameters further at 48 h compared to the vehicle group (p < 0.05). The latter effects of prostacyclin cannot be attributed to an increase in permeability, as prostacyclin did not influence PS or brain water content following sham trauma. In fact prostacyclin has been shown to have permeability-decreasing properties. We conclude that prostacyclin improves cortical perfusion following brain trauma. The simultaneous aggravation of brain edema can be explained by an increased surface area, perhaps in combination with increased capillary hydrostatic pressure.
Collapse
Affiliation(s)
- Peter Bentzer
- Department of Physiological Sciences, University of Lund and Lund University Hospital, Lund, Sweden.
| | | | | | | |
Collapse
|
91
|
Maxwell WL, Dhillon K, Harper L, Espin J, MacIntosh TK, Smith DH, Graham DI. There is differential loss of pyramidal cells from the human hippocampus with survival after blunt head injury. J Neuropathol Exp Neurol 2003; 62:272-9. [PMID: 12638731 DOI: 10.1093/jnen/62.3.272] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The experimental literature has shown that neurons within sub-fields of the hippocampus possess differential sensitivities to cell loss after different types of insult to the brain. In humans, after blunt head injury, differential neuronal responses between sub-fields of the hippocampus up to 72 hours after injury have been documented. But, in only a small part of the literature have data for alterations in real numbers of neurons been provided. In this study the hypothesis was tested that, after severe blunt head injury in humans, the total number of neurons within a defined volume of brain tissue differed between different sub-fields of the hippocampus and between groups of patients with differing post-traumatic survivals. Stereological methods were used to measure total cross-sectional area of sub-fields of the hippocampus taken at the level of the lateral geniculate nucleus and count numbers of neurons within each of the CA1, CA2, CA3, and CA4 sub-fields of the hippocampus in patients. The patients used in this study were categorized as follows: Group 1 (early) had survived for 1 week or less; Group 2 (late) survived 6 months or longer after fatal severe head injury; and Group 3 (controls) consisted of age-matched patients that had no history of head injury or disease prior to death. There was a significant loss in cross-sectional area in sub-fields CA3 and CA4 at 1 week or less after injury and in sub-field CA1 at 6 months and greater survival. There was no change in CA2. There was loss of neurons from within a predefined volume of brain tissue in sub-fields CA1, CA3, and CA4 one week or less after injury. But there was no loss in CA2. There was continued loss of neurons from sub-fields CA1 and CA4 between 1 week and 6 months and greater survival, but there was no loss of neurons in sub-fields CA2 and CA3 within the same period. These novel data show that after human severe head injury there is first an acute loss (1 week or less survival) of pyramidal neurons in all hippocampal sub-fields except CA2. Second, there is an ongoing loss of neurons in sub-field CA1 and, most notably, in sub-field CA4, in patients surviving for more than 6 months. However, in neither group of patients is there loss of neurons from sub-field CA2.
Collapse
Affiliation(s)
- W L Maxwell
- Department of Anatomy, IBLS, University of Glasgow, Glasgow, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
92
|
Leker RR, Shohami E, Constantini S. Experimental models of head trauma. ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 83:49-54. [PMID: 12442621 DOI: 10.1007/978-3-7091-6743-4_9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Traumatic brain injury is one of the most common causes for chronic disability in young people. Despite this there are currently no widely available modes of therapy that would limit the extent of brain damage secondary to trauma. Therefore, new insights into the pathological mechanisms involved in head trauma possibly leading to the identification of new therapeutic targets are urgently needed. In order to attain these goals adequate animal models for traumatic brain injury are needed. In the following paper the authors will review the various animal models for head trauma and emphasize their potential strengths and weaknesses.
Collapse
Affiliation(s)
- R R Leker
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hebrew University-Hadassah Medical School, Hadassah University Hospital, Jerusalem, Israel
| | | | | |
Collapse
|
93
|
Schuhmann MU, Stiller D, Skardelly M, Mokktarzadeh M, Thomas S, Brinker T, Samii M. Determination of contusion and oedema volume by MRI corresponds to changes of brain water content following controlled cortical impact injury. ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 81:213-5. [PMID: 12168307 DOI: 10.1007/978-3-7091-6738-0_55] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The time-course of brain contusion/oedema development as visualised by high-resolution MRI was compared to brain water content following experimental brain contusion. 36 Sprague-Dawley rats underwent Controlled Cortical Impact Injury (CCII), 24 served as controls. In 16 animals serial T2 weighted MRI investigations at 1 h, 4 h, 24 h and 7 d after CCII were performed, in 44 rats hemispheric brain water content was determined at the same time points by wet dry weight method. MRI lesion volume (mm3) and brain water content of injured hemisphere (%) showed for absolute and relative values a strictly parallel course. Significant posttraumatic increases had a maximum at 24 hours. Values on day 7 were below those of 1st h in both methods. The simple non-invasive MRI method quantifies contusion and surrounding penumbra according to elevated tissue water signal. The invasive wet dry weight method quantifies changes of hemispheric brain water content that are likely to take place in contusion core and surrounding penumbra. Therefore, from a theoretical aspect both methods seem comparable. Following experimental brain contusion, the simple MRI method might be an equally sufficient way to describe post-traumatic or post-therapeutic changes of lesion size and brain oedema.
Collapse
Affiliation(s)
- M U Schuhmann
- Department of Neurosurgery, Medical School Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
94
|
Thomale UW, Schaser K, Kroppenstedt SN, Unterberg AW, Stover JF. Cortical hypoperfusion precedes hyperperfusion following controlled cortical impact injury. ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 81:229-31. [PMID: 12168311 DOI: 10.1007/978-3-7091-6738-0_59] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Impaired cerebral perfusion contributes to tissue damage following traumatic brain injury. In this longitudinal study persistence of reduced cortical perfusion employing laser doppler flowmetry was investigated following controlled cortical impact injury (CCII). Before, 30 minutes, 6, 24, and 48 hours after CCII, perfusion in pericontusional cortex was determined by moving a laser doppler probe in 50 x 0.2 mm steps over the traumatized hemisphere in 5 rats. Arterial blood gases and mean arterial blood pressure were monitored. Mean arterial blood pressure and arterial blood gases remained stable during the entire experiments. At 30 minutes and 6 hours following CCII, cortical perfusion was significantly diminished by 24% and 43% (p < 0.05), respectively compared to pre-trauma levels. At 24 and 48 hours after CCII, pericontusional blood flow was significantly increased by 64% and 123%. Cortical hypoperfusion found within the early phase following trauma is reversible and precedes a long lasting phase of hyperperfusion. Changes in tissue mediators (endothelin, acidosis, NO) could account for these findings.
Collapse
Affiliation(s)
- U W Thomale
- Department of Neurosurgery, Charité, Virchow Medical Center, Humboldt-University of Berlin, Germany
| | | | | | | | | |
Collapse
|
95
|
Abstract
The main aim of this review is to describe some of the many animal models that have proved to be valuable from a neuroimaging perspective. This paper complements other articles in this volume, with a focus on animal models of the pathology of human brain disorders for investigations with modern non-invasive neuroimaging techniques. The use of animal model systems forms a fundamental part of neuroscience research efforts to improve the prevention, diagnosis, understanding and treatment of neurological conditions. Without such models it would be impossible to investigate such topics as the underlying mechanisms of neuronal cell damage and death, or to screen compounds for possible anticonvulsant properties. The adequacy of any one particular model depends on the suitability of information gained during experimental conditions. It is important, therefore, to understand the various types of animal model available and choose an appropriate model for the research question.
Collapse
Affiliation(s)
- Mark F Lythgoe
- RCS Unit of Biophysics, Institute of Child Health, University College London, UK
| | | | | |
Collapse
|
96
|
Kroppenstedt SN, Sakowitz OW, Thomale UW, Unterberg AW, Stover JF. Influence of norepinephrine and dopamine on cortical perfusion, EEG activity, extracellular glutamate, and brain edema in rats after controlled cortical impact injury. J Neurotrauma 2002; 19:1421-32. [PMID: 12490007 DOI: 10.1089/089771502320914651] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Following traumatic brain injury, catecholamines given to ameliorate cerebral perfusion may induce brain damage via cerebral arteriolar constriction and increased neuronal excitation. In the present study the acute effects of norepinephrine and dopamine on pericontusional cortical perfusion (rCBF), electroencephalographic (EEG) activity, extracellular glutamate, and brain edema were investigated in rats following controlled cortical impact injury (CCI). rCBF, cerebral perfusion pressure (CPP), EEG activity, and glutamate were determined before, during, and after infusing norepinephrine or dopamine, increasing MABP to 120 mm Hg for 90 min at 4 h after CCI. Control rats received physiological saline. At 8 h after CCI, hemispheric swelling and water content were determined gravimetrically. Following CCI, rCBF was significantly decreased. In parallel to elevating MABP and CPP, rCBF was significantly increased by norepinephrine and dopamine, being mostly pronounced with norepinephrine (+44% vs. +29%). In controls, rCBF remained diminished (-45%). EEG activity was significantly increased by norepinephrine and dopamine, while pericontusional glutamate was only elevated by norepinephrine (28 +/- 6 vs. 8 +/- 4 microM). Brain edema was not increased compared to control rats. Despite significantly increasing MABP and CPP to the same extent, norepinephrine and dopamine seem to differentially influence pericontusional cortical perfusion and glutamatergic transmission. In addition to the pressure-passive increase in CPP local cerebral effects seem to account for the sustained norepinephrine-induced increase in pericontusional cortical perfusion. The significantly elevated pericontusional glutamate concentrations in conjunction with the increased EEG activity suggest a sustained metabolically driven increase in cortical perfusion during norepinephrine infusion.
Collapse
|
97
|
Zhang F, Sprague SM, Farrokhi F, Henry MN, Son MG, Vollmer DG. Reversal of attenuation of cerebrovascular reactivity to hypercapnia by a nitric oxide donor after controlled cortical impact in a rat model of traumatic brain injury. J Neurosurg 2002; 97:963-9. [PMID: 12405388 DOI: 10.3171/jns.2002.97.4.0963] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Traumatic brain injury (TBI) attenuates the cerebral vasodilation to hypercapnia. Cortical spreading depression (CSD) also transiently reduces hypercapnic vasodilation. The authors sought to determine whether the CSD elicited by a controlled cortical impact (CCI) injury masks the true effect of TBI on hypercapnic vasodilation, and whether a nitric oxide (NO) donor can reverse the attenuation of hypercapnic vasodilation following CCI. METHODS Anesthetized rats underwent moderate CCI. Cerebral blood flow was monitored with laser Doppler flowmetry and the response to hypercapnia was determined for injured and sham-injured animals. The effect of the NO donor, S-nitroso-N-acetylpenicillamine (SNAP), on this response was also assessed. At an uninjured cortical site ipsilateral to the CCI, a single wave of CSD was recorded and the CO2 response at this location was significantly attenuated for up to 30 minutes (seven rats, p < 0.05). At the injured cortex, hypercapnic vasodilation continued to be attenuated for 7 hours. The cerebral vasodilation to CO2 was 37 +/- 5% in injured rats (six) compared with 84 +/- 10% in the sham-injured group (five rats, p < 0.05). After 30 minutes of topical superfusion with SNAP, hypercapnic vasodilation was restored to 74 +/- 7% (nine rats, p > 0.1 compared with that in the sham-injured group). In contrast, papaverine, an NO-independent vasodilator, failed to reverse the attenuation of the CO2 response to CCI. CONCLUSIONS The authors conclude that CSD elicited by CCI can mask the true effect of TBI on hypercapnic vasodilation for at least 30 minutes. Exogenous NO, but not papaverine, can reverse the attenuation of cerebrovascular reactivity to CO2 caused by TBI. This result supports the hypothesis that NO production is reduced after TBI and that the NO donor has a potential beneficial role in the clinical management of head injury.
Collapse
Affiliation(s)
- Fangyi Zhang
- Department of Surgery, University of Texas Health Science Center, San Antonio 78229-3900, USA.
| | | | | | | | | | | |
Collapse
|
98
|
Kochanek PM, Hendrich KS, Dixon CE, Schiding JK, Williams DS, Ho C. Cerebral blood flow at one year after controlled cortical impact in rats: assessment by magnetic resonance imaging. J Neurotrauma 2002; 19:1029-37. [PMID: 12482116 DOI: 10.1089/089771502760341947] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Progressive tissue loss and delayed cognitive deficits are seen in rats during the initial year after experimental traumatic brain injury (TBI). As much as 10% of parenchymal volume is lost even in the contralateral hemisphere by 1 year after controlled cortical impact (CCI) in rats. Progressive declines in cerebral blood flow (CBF) are also associated with advanced age and neurodegenerative diseases. Surprisingly, the long-term effects of TBI on CBF remain undefined. CBF was quantified by continuous arterial spin-labeled magnetic resonance imaging (MRI) and measurements of spin-lattice relaxation time in a slice through the plane of injury at 1 year after experimental TBI produced by CCI (n = 4) or sham surgery (n = 4) in rats. CBF was quantified in six regions of interest (ROIs) that were anatomically identified on the control images in each hemisphere and included a medial cortical segment (contusion-enriched, beneath the impact site, on the ipsilateral side) cortex, hippocampus, thalamus, amygdala/pyriform cortex, and hemisphere. At 1 year after injury, CBF was dramatically (96%) reduced in structures within the large cystic lesion that was seen in three of four rats and variably included cortex and hippocampus. Overall, there was an 80% reduction in CBF in the ipsilateral medial cortical segment comparing CCI and sham groups. Similarly, 52% and 67% reductions were seen in CBF in the cortical and hippocampal ROIs ipsilateral to impact (CCI vs. sham), respectively. These are regions both with marked CBF disturbances early after injury and that ultimately suffer considerable tissue loss over the 1-year interval. However, at 1 year after CCI, CBF was not different from sham in other ROIs, including ipsilateral thalamus, or either contralateral hippocampus or hemisphere. We conclude that, at 1 year after CCI, CBF is reduced in anatomic structures at or near the impact site, including injured cortex and hippocampus, and this translates into a reduction in hemispheric CBF. However, despite both significant occult tissue loss ipsilateral and contralateral to the injury and delayed cognitive deficits, widespread reductions in CBF are not observed. This suggests the possibility of remodeling or repackaging of the brain that preserves CBF outside of the cystic lesion.
Collapse
Affiliation(s)
- Patrick M Kochanek
- Department of Critical Care Medicine and Anesthesiology, Center for Resuscitation Research, Pittsburgh, Pennsylvania 15260, USA.
| | | | | | | | | | | |
Collapse
|
99
|
Rose ME, Huerbin MB, Melick J, Marion DW, Palmer AM, Schiding JK, Kochanek PM, Graham SH. Regulation of interstitial excitatory amino acid concentrations after cortical contusion injury. Brain Res 2002; 943:15-22. [PMID: 12088834 DOI: 10.1016/s0006-8993(02)02471-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Increases in brain interstitial excitatory amino acid (EAA(I)) concentrations after ischemia are ameliorated by use-dependent Na+ channel antagonists and by supplementing interstitial glucose, but the regulation of EAA(I) after traumatic brain injury (TBI) is unknown. We studied the regulation of EAA(I) after TBI using the controlled cortical impact model in rats. To monitor changes in EAA(I), microdialysis probes were placed in the cortex adjacent to the contusion and in the ipsilateral hippocampus. Significant increases in dialysate EAA(I) after TBI were found compared to levels measured in sham controls. Treatment with the use-dependent Na+ channel antagonist 619C89 (30 mg/kg i.v.) did not significantly decrease dialysate glutamate compared to vehicle controls in hippocampus (10.4+/-2.4 vs. 11.9+/-1.6 microM), but there was significant decrease in dialysate glutamate in cortex after 619C89 treatment (19.3+/-3 vs. 12.6+/-1.1 microM, P<0.05). Addition of 30 mM glucose to the dialysate, a treatment that decreases EAA(I) after ischemia, had no significant effect upon dialysate glutamate after TBI in cortex (20.0+/-4.9 vs. 11.7+/-3.4 microM) or in hippocampus (10.9+/-2.0 vs. 8.9+/-2.4 microM). These results suggest that neither increased release of EAAs due to Na+ channel-mediated depolarization nor failure of glutamate reuptake due to glucose deprivation can explain the majority of the increase in EAA(I) following TBI.
Collapse
Affiliation(s)
- Marie E Rose
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Kiening KL, van Landeghem FKH, Schreiber S, Thomale UW, von Deimling A, Unterberg AW, Stover JF. Decreased hemispheric Aquaporin-4 is linked to evolving brain edema following controlled cortical impact injury in rats. Neurosci Lett 2002; 324:105-8. [PMID: 11988338 DOI: 10.1016/s0304-3940(02)00180-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The cerebral Aquaporin-4 (AQP4) water channel is suggested to be involved in brain edema formation aggravated by reduced cerebral blood flow early after traumatic brain injury (TBI). Therefore, the temporal profile of brain edema formation, AQP4 expression, and cortical perfusion were investigated following focal TBI in rats. Brain edema was maximal by 24 h. Concurrently, AQP4 protein expression was decreased in both hemispheres, being more pronounced in the traumatized hemisphere (-50%) 48 h after trauma. Cortical perfusion was only decreased in the ipsilateral cortex (-40%) between 4 and 8 h after trauma, reaching baseline values at 24 h. Globally reduced AQP4 expression following induction of a focal contusion coincides with edema development and seems to be independent of changes in cortical perfusion.
Collapse
Affiliation(s)
- Karl L Kiening
- Department of Neurosurgery, Virchow Medical Center, Charité, Humboldt-University at Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|