1
|
Proctor JL, Xu S, Guo S, Piskoun B, Miller C, Roys S, Gullapalli RP, Fiskum G. Aeromedical evacuation-relevant hypobaria following traumatic brain injury in rats contributes to cerebral blood flow depression, altered neurochemistry and increased neuroinflammation. J Cereb Blood Flow Metab 2025; 45:945-959. [PMID: 39696912 PMCID: PMC11656461 DOI: 10.1177/0271678x241299985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 09/11/2024] [Accepted: 10/16/2024] [Indexed: 12/20/2024]
Abstract
Aircraft cabins are routinely pressurized to the equivalent of 8000 ft altitude. Exposure of lab animals to aeromedical evacuation relevant hypobaria after traumatic brain injury worsens neurological outcomes, which is paradoxically exacerbated by hyperoxia. This study tested the hypothesis that exposure of rats to hypobaria following cortical impact reduces cerebral blood flow, increases neuroinflammation, and alters brain neurochemistry. Rats were exposed to simulated ground (normobaric) or air (hypobaric 8000 ft) transport, under normoxia or hyperoxia, 24 hr after trauma. Hypobaria exposure resulted in lower cerebral blood flow to the contralateral cortex and bilateral thalamus during flight and increased delayed cortical inflammation (ED1 immunoreactivity) at 14 days post injury. Impacted rats exposed to hypobaria had higher cortical creatine levels compared rats maintained at sea level. Exposure to the combination of hyperoxia and hypobaria resulted in the greatest reduction in cortical blood flow and total creatine during flight which persisted up to two weeks. In conclusion, hypoperfusion during hypobaria exposure could contribute to worsening of neuroinflammation and neurochemical imbalances. The presence of excessive O2 during hypobaria results in long-term suppression of cerebral blood flow, indicating that supplemental O2 should be titrated during hypobaria to maintain normoxia.
Collapse
Affiliation(s)
- Julie L Proctor
- Department of Anesthesiology and the Center for Shock Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sijia Guo
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Boris Piskoun
- Department of Anesthesiology and the Center for Shock Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Catriona Miller
- Department of Anesthesiology and the Center for Shock Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Steven Roys
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rao P Gullapalli
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gary Fiskum
- Department of Anesthesiology and the Center for Shock Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Gober IG, Russell AL, Shick TJ, Vagni VA, Carlson JC, Kochanek PM, Wagner AK. Exploratory assessment of the effect of systemic administration of soluble glycoprotein 130 on cognitive performance and chemokine levels in a mouse model of experimental traumatic brain injury. J Neuroinflammation 2024; 21:149. [PMID: 38840141 PMCID: PMC11155101 DOI: 10.1186/s12974-024-03129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024] Open
Abstract
Uncontrolled neuroinflammation mediates traumatic brain injury (TBI) pathology and impairs recovery. Interleukin-6 (IL-6), a pleiotropic inflammatory regulator, is associated with poor clinical TBI outcomes. IL-6 operates via classical-signaling through membrane-bound IL-6 receptor (IL-6R) and trans-signaling through soluble IL-6 receptor (s)IL-6R. IL-6 trans-signaling specifically contributes to neuropathology, making it a potential precision therapeutic TBI target. Soluble glycoprotein 130 (sgp130) prevents IL-6 trans-signaling, sparing classical signaling, thus is a possible treatment. Mice received either controlled cortical impact (CCI) (6.0 ± 0.2 m/s; 2 mm; 50-60ms) or sham procedures. Vehicle (VEH) or sgp130-Fc was subcutaneously administered to sham (VEH or 1 µg) and CCI (VEH, 0.25 µg or 1 µg) mice on days 1, 4, 7, 10 and 13 post-surgery to assess effects on cognition [Morris Water Maze (MWM)] and ipsilateral hemisphere IL-6 related biomarkers (day 21 post-surgery). CCI + sgp130-Fc groups (0.25 µg and 1 µg) were combined for analysis given similar behavior/biomarker outcomes. CCI + VEH mice had longer latencies and path lengths to the platform and increased peripheral zone time versus Sham + VEH and Sham + sgp130-Fc mice, suggesting injury-induced impairments in learning and anxiety. CCI + sgp130-Fc mice had shorter platform latencies and path lengths and had decreased peripheral zone time, indicating a therapeutic benefit of sgp130-Fc after injury on learning and anxiety. Interestingly, Sham + sgp130-Fc mice had shorter platform latencies, path lengths and peripheral zone times than Sham + VEH mice, suggesting a beneficial effect of sgp130-Fc, independent of injury. CCI + VEH mice had increased brain IL-6 and decreased sgp130 levels versus Sham + VEH and Sham + sgp130-Fc mice. There was no treatment effect on IL-6, sIL6-R or sgp130 in Sham + VEH versus Sham + sgp130-Fc mice. There was also no treatment effect on IL-6 in CCI + VEH versus CCI + sgp130-Fc mice. However, CCI + sgp130-Fc mice had increased sIL-6R and sgp130 versus CCI + VEH mice, demonstrating sgp130-Fc treatment effects on brain biomarkers. Inflammatory chemokines (MIP-1β, IP-10, MIG) were increased in CCI + VEH mice versus Sham + VEH and Sham + sgp130-Fc mice. However, CCI + sgp130-Fc mice had decreased chemokine levels versus CCI + VEH mice. IL-6 positively correlated, while sgp130 negatively correlated, with chemokine levels. Overall, we found that systemic sgp130-Fc treatment after CCI improved learning, decreased anxiety and reduced CCI-induced brain chemokines. Future studies will explore sex-specific dosing and treatment mechanisms for sgp130-Fc therapy.
Collapse
Affiliation(s)
- Ian G Gober
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Ashley L Russell
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Tyler J Shick
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Vincent A Vagni
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenna C Carlson
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA.
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA.
- Center for Neuroscience, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Chandrasekaran S, Santibanez F, Long T, Nichols T, Kait J, Bruegge RV, 'Dale' Bass CR, Pinton G. Shear shock wave injury in vivo: High frame-rate ultrasound observation and histological assessment. J Biomech 2024; 166:112021. [PMID: 38479150 DOI: 10.1016/j.jbiomech.2024.112021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 04/13/2024]
Abstract
Using high frame-rate ultrasound and ¡1μm sensitive motion tracking we previously showed that shear waves at the surface of ex vivo and in situ brains develop into shear shock waves deep inside the brain, with destructive local accelerations. However post-mortem tissue cannot develop injuries and has different viscoelastodynamic behavior from in vivo tissue. Here we present the ultrasonic measurement of the high-rate shear shock biomechanics in the in vivo porcine brain, and histological assessment of the resulting axonal pathology. A new biomechanical model of brain injury was developed consisting of a perforated mylar surface attached to the brain and vibrated using an electromechanical shaker. Using a custom sequence with 8 interleaved wide beam emissions, brain imaging and motion tracking were performed at 2900 images/s. Shear shock waves were observed for the first time in vivo wherein the shock acceleration was measured to be 2.6 times larger than the surface acceleration ( 95g vs. 36g). Histopathology showed axonal damage in the impacted side of the brain from the brain surface, accompanied by a local shock-front acceleration of >70g. This shows that axonal injury occurs deep in the brain even though the shear excitation was at the brain surface, and the acceleration measurements support the hypothesis that shear shock waves are responsible for deep traumatic brain injuries.
Collapse
Affiliation(s)
| | - Francisco Santibanez
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| | - Tyler Long
- Departments of Medicine and Pathology and Laboratory Medicine at University of North Carolina at Chapel Hill, USA
| | - Tim Nichols
- Departments of Medicine and Pathology and Laboratory Medicine at University of North Carolina at Chapel Hill, USA
| | - Jason Kait
- Department of Biomedical Engineering, Duke University, USA
| | - Ruth Vorder Bruegge
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| | | | - Gianmarco Pinton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA.
| |
Collapse
|
4
|
Meyer S, Hummel R, Neulen A, Hirnet T, Thal SC. Influence of traumatic brain injury on ipsilateral and contralateral cortical perfusion in mice. Neurosci Lett 2023; 795:137047. [PMID: 36603737 DOI: 10.1016/j.neulet.2023.137047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/28/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023]
Abstract
Traumatic brain injury (TBI) is one of the most important causes of death in young adults. After brain injury cortical perfusion is impaired by cortical spreading depression, cerebral microvasospasm or microvascular thrombosis and contributes to secondary expansion of lesion into surrounding healthy brain tissue. The present study was designed to determine the regional cortical perfusion pattern after experimental TBI induced by controlled cortical impact (CCI) in male C57/BL6N mice. We performed a longitudinal time series analysis by Laser speckle contrast imaging (LSCI). Measurements were carried out before, immediately and 24 h after trauma. Immediately after CCI cortical perfusion in the lesion core dropped to 10 % of before injury (baseline; %BL) and to 21-24 %BL in the cortical area surrounding the core. Interestingly, cortical perfusion was also significantly reduced in the contralateral non-injured hemisphere (41-58 %BL) matching the corresponding brain region of the injured hemisphere. 24 h after CCI perfusion of the contralateral hemisphere returned to baseline level in the area corresponding to the lesion core, whereas the lateral area of the parietal cortex was hyperperfused (125 %BL). The lesion core region itself remained severely hypoperfused (18 to 26 %BL) during the observation period. TBI causes a maldistribution of both ipsi- and contralateral cerebral perfusion immediately after trauma, which persist for at least 24 h. Higher perfusion levels in the lesion core 24 h after trauma were associated with increased tissue damage, which supports the role of reperfusion injury for secondary brain damage after TBI.
Collapse
Affiliation(s)
- Simon Meyer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Regina Hummel
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Axel Neulen
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Tobias Hirnet
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Serge C Thal
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; Department of Anesthesiology, HELIOS University Hospital Wuppertal, University Witten/Herdecke, Heusnerstraße 40, 42283 Wuppertal, Germany.
| |
Collapse
|
5
|
Comparative study of brain damage and oxidative stress using two animal models of the shaken baby syndrome. Exp Gerontol 2022; 166:111874. [PMID: 35779807 DOI: 10.1016/j.exger.2022.111874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/26/2022] [Accepted: 06/21/2022] [Indexed: 11/21/2022]
Abstract
The objective was compare the morphological damages in brain and to evaluate the participation of oxidative stress, using two animal models of shaken baby syndrome (SBS). Five-day-old Wistar rats were used to develop two models of SBS as follows: Gyrotwister (GT) group was subjected to low intensity, high duration rotating movements and Ratshaker (RS) group made to undergo high intensity, low duration anteroposterior movements. Both groups presented respiratory distress, weight loss and shorter stature compared with the control group. In addition, involuntary movements occurred in both experimental models. Hemorrhage was observed in 10 % of the GT group and in 40 % of the RS group. This last group experienced lesser weight gain at 30 days. Glutathione decreased by 25.7 % (GT) and 59.96 (RT). Cell data analysis revealed the presence of crenate and pyknotic cells, characterized by apparent absence of nucleus and nucleolus as well as vacuolation in the GT group. In the RS group, there were a high number of angular, pyknotic and shrunken cells, and a lot of vacuolization. The severity of the brain damage can be related to the magnitude of biochemical modifications, specifically, those related to the production of reactive oxygen or nitrogen species, oxidative stress, oxidative damage.
Collapse
|
6
|
Goodfellow M, Medina JA, Proctor J, Xu S, Gullapalli RP, Rangghran P, Miller C, Vesselinov A, Fiskum G. Combined traumatic brain injury and hemorrhagic shock in ferrets leads to structural, neurochemical, and functional impairments. J Neurotrauma 2022; 39:1442-1452. [PMID: 35481784 DOI: 10.1089/neu.2022.0102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Aeromedical evacuation-relevant hypobaria after traumatic brain injury (TBI) leads to increased neurologic injury and mortality in rats relative to those maintained under normobaria. However, applicability of rodent brain injury research to humans may be limited by differences in neuroanatomy. Therefore, we developed a model in which ferrets are exposed to polytrauma consisting of controlled cortical impact TBI and hemorrhagic shock subjected 24 h later to 6 h of hypobaria or normobaria. Our objective was to determine if the deleterious effects of hypobaria observed in rats, with lissencephalic brains, are also present in a species with a human-like gyrencephalic brain. While no mortality was observed, magnetic resonance spectroscopy (MRS) results obtained 2 days post-injury indicated reduced cortical creatine, N-acetylaspartate, GABA, myo-inositol, and glutamate which was not affected by hypobaria. T2-weighted magnetic resonance imaging (MRI) quantification revealed increased hyperintensity volume representing cortical edema at the site of impact following polytrauma. Hypobaria did not exacerbate this focal edema but did lead to overall reductions in total cortical volume. Both normobaric and hypobaric ferrets exhibited impaired spatial memory 6 days post-injury on the Object Location Test, but no differences were noted between groups. Finally, cortical lesion volume was not exacerbated by hypobaria exposure on day 7 post-injury. Results suggest that air travel 24 h after polytrauma is associated with structural changes in the ferret brain. Future studies should investigate secondary injury from hypobaria following polytrauma in greater detail including alternative outcome measures, timepoints, and exposure to multiple flights.
Collapse
Affiliation(s)
- Molly Goodfellow
- University of Maryland School of Medicine, 12264, Anesthesiology, Baltimore, Maryland, United States;
| | - Juliana A Medina
- University of Maryland School of Medicine, Anesthesiology, Baltimore, Maryland, United States;
| | - Julie Proctor
- University of Maryland School of Medicine, Anesthesiology, 685 W Baltimore St, 534 MSTF, Baltimore, Maryland, United States, 21201;
| | - Su Xu
- University of Maryland School of Medicine, Diagnostic Radiology & Nuclear Medicine, Baltimore, Maryland, United States;
| | - Rao P Gullapalli
- University of Maryland School of Medicine, 12264, Diagnostic Radiology & Nuclear Medicine, 670 W Batimore St, Baltimore, Maryland, United States, 21201;
| | - Parisa Rangghran
- University of Maryland School of Medicine, Anesthesiology, Baltimore, Maryland, United States;
| | - Catriona Miller
- University of Maryland School of Medicine, Anesthesiology, Baltimore, Maryland, United States;
| | - Alexandra Vesselinov
- University of Maryland School of Medicine, Anesthesiology, Baltimore, Maryland, United States;
| | - Gary Fiskum
- University of Maryland School of Medicine, 12264, Anesthesiology, Baltimore, Maryland, United States;
| |
Collapse
|
7
|
Wang J, Xie X, Wu Y, Zhou Y, Li Q, Li Y, Xu X, Wang M, Murdiyarso L, Houck K, Hilton T, Chung D, Li M, Zhang JN, Dong J. Brain-Derived Extracellular Vesicles Induce Vasoconstriction and Reduce Cerebral Blood Flow in Mice. J Neurotrauma 2022; 39:879-890. [PMID: 35316073 DOI: 10.1089/neu.2021.0274] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) impairs cerebrovascular autoregulation and reduces cerebral blood flow (CBF), leading to ischemic secondary injuries. We have shown that injured brains release brain-derived extracellular vesicles (BDEVs) into circulation, where they cause a systemic hypercoagulable state that rapidly turns into consumptive coagulopathy. BDEVs induce endothelial injury and permeability, leading to the hypothesis that they contribute to TBI-induced cerebrovascular dysregulation. In a study designed to test this hypothesis, we detected circulating BDEVs in C57BL/6J mice subjected to severe TBI, reaching peak levels of 3x104/µl at 3 hours post injury (71.2±21.5% of total annexin V-binding EVs). We further showed in an adaptive transfer model that 41.7±5.8% of non-injured mice died within 6 hours after being infused with 3x104/µl of BDEVs. BDEVs transmigrated through the vessel walls, induced rapid vasoconstriction by inducing calcium influx in vascular smooth muscle cells, and reduced CBF by 93.8±5.6% within 30 minutes after infusion. The CBF suppression was persistent in mice that eventually died but it recovered quickly in surviving mice. It was prevented by the calcium channel blocker nimodipine. When being separated, neither protein nor phospholipid components from the lethal number of BDEVs induced vasoconstriction, reduced CBF, and caused death. These results demonstrate a novel vasoconstrictive activity of BDEVs that depends on the structure of BDEVs and contributes to TBI-induced disseminated cerebral ischemia and sudden death.
Collapse
Affiliation(s)
- Jiwei Wang
- Tianjin Neurological Institute, 230967, Anshan road No.154, Tianjin, China, 300052;
| | - Xiaofeng Xie
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | - Yingang Wu
- University of Science and Technology of China, 12652, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine., Hefei, Anhui, China;
| | - Yuan Zhou
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Qifeng Li
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Ying Li
- Tianjin Neurological Institute, 230967, Tianjin, Tianjin, China;
| | - Xin Xu
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Min Wang
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | | | - Katie Houck
- Bloodworks Research institute, Seattle, United States;
| | | | - Dominic Chung
- Bloodworks Research institute, Seattle, United States;
| | - Min Li
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | - Jian-Ning Zhang
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Jingfei Dong
- Bloodworks Research Institute, Bloodworks Northwest, Seattle, Seattle, Washington, United States.,Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States;
| |
Collapse
|
8
|
Tatara Y, Shimada R, Kibayashi K. Effects of Preexisting Diabetes Mellitus on the Severity of Traumatic Brain Injury. J Neurotrauma 2020; 38:886-902. [PMID: 32998635 DOI: 10.1089/neu.2020.7118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Falls and traffic accidents can cause traumatic brain injury (TBI). Assessment of the injury severity is essential to determine the prognosis or the cause of death. Diabetes mellitus (DM) is a common preexisting disease in elderly adults. We hypothesized that preexisting DM exacerbates TBI secondary to prolonged inflammation. In this study, we investigated TBI-induced changes in nerve function and inflammatory cell migration to the injury site, and the extent of brain contusion in KK-Ay (DM) and C57BL/6J (non-DM) mice. A controlled cortical impact device was used to induce TBI in each mouse. The brain contusion volume was measured using magnetic resonance imaging. Nerve function changes were assessed using the following animal behavior tasks: neurological severity score (NSS), Morris water maze, forced swim test, and beam walking. Immunohistochemical examinations of brain sections were performed to assess the infiltration of neutrophils, astrocytes, microglia, and macrophages, and to detect apoptosis. These experiments were performed on post-injury days 1-90 (over five experiments/time-points in each group). Compared with non-DM mice, DM mice showed significantly greater brain contusion volume, greater deterioration in the NSS, and a higher number of neutrophils, macrophages, and apoptotic cells in the brain tissue specimens. This study indicates that the prognosis of normal mice and DM mice differs, even if they acquire a TBI of the same severity. Therefore, it is important to evaluate patients with TBI for DM and other preexisting diseases in order to provide adequate treatment or to determine the correct cause of death.
Collapse
Affiliation(s)
- Yuki Tatara
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Ryo Shimada
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuhiko Kibayashi
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
9
|
Washington PM, Lee C, Dwyer MKR, Konofagou EE, Kernie SG, Morrison B. Hyaluronidase reduced edema after experimental traumatic brain injury. J Cereb Blood Flow Metab 2020; 40:2026-2037. [PMID: 31648593 PMCID: PMC7786840 DOI: 10.1177/0271678x19882780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cerebral edema and the subsequent increased intracranial pressure are associated with mortality and poor outcome following traumatic brain injury. Previous in vitro studies have shown that the Gibbs-Donnan effect, which describes the tendency of a porous, negatively charged matrix to attract positive ions and water, applies to brain tissue and that enzymatic reduction of the fixed charge density can prevent tissue swelling. We tested whether hyaluronidase, an enzyme that degrades the large, negatively charged glycosaminoglycan hyaluronan, could reduce brain edema after traumatic brain injury. In vivo, intracerebroventricular injection of hyaluronidase after controlled cortical impact in mice reduced edema in the ipsilateral hippocampus at 24 h by both the wet-weight/dry-weight method (78.15 ± 0.65% vs. 80.4 ± 0.46%; p < 0.01) and T2-weighted magnetic resonance imaging (13.88 ± 3.09% vs. 29.23 ± 6.14%; p < 0.01). Hyaluronidase did not adversely affect blood-brain-barrier-integrity measured by dynamic contrast-enhanced magnetic resonance imaging, nor did hyaluronidase negatively affect functional recovery after controlled cortical impact measured with the rotarod or Morris water maze tasks. Reduction of fixed charge density by hyaluronidase was confirmed in cortical explants in vitro (5.46 ± 1.15 µg/mg vs. 7.76 ± 1.87 µg/mg; p < 0.05). These data demonstrate that targeting the fixed charge density with hyaluronidase reduced edema in an in vivo mouse model of traumatic brain injury.
Collapse
Affiliation(s)
- Patricia M Washington
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Changhee Lee
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Mary Kate R Dwyer
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Steven G Kernie
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Barclay Morrison III, Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY 10027, USA.
| |
Collapse
|
10
|
Bharadwaj VN, Copeland C, Mathew E, Newbern J, Anderson TR, Lifshitz J, Kodibagkar VD, Stabenfeldt SE. Sex-Dependent Macromolecule and Nanoparticle Delivery in Experimental Brain Injury. Tissue Eng Part A 2020; 26:688-701. [PMID: 32697674 PMCID: PMC7398445 DOI: 10.1089/ten.tea.2020.0040] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023] Open
Abstract
The development of effective therapeutics for brain disorders is challenging, in particular, the blood-brain barrier (BBB) severely limits access of the therapeutics into the brain parenchyma. Traumatic brain injury (TBI) may lead to transient BBB permeability that affords a unique opportunity for therapeutic delivery via intravenous administration ranging from macromolecules to nanoparticles (NPs) for developing precision therapeutics. In this regard, we address critical gaps in understanding the range/size of therapeutics, delivery window(s), and moreover, the potential impact of biological factors for optimal delivery parameters. Here we show, for the first time, to the best of our knowledge, that 24-h postfocal TBI female mice exhibit a heightened macromolecular tracer and NP accumulation compared with male mice, indicating sex-dependent differences in BBB permeability. Furthermore, we report for the first time the potential to deliver NP-based therapeutics within 3 days after focal injury in both female and male mice. The delineation of injury-induced BBB permeability with respect to sex and temporal profile is essential to more accurately tailor time-dependent precision and personalized nanotherapeutics. Impact statement In this study, we identified a sex-dependent temporal profile of blood/brain barrier disruption in a preclinical mouse model of traumatic brain injury (TBI) that contributes to starkly different macromolecule and nanoparticle delivery profiles post-TBI. The implications and potential impact of this work are profound and far reaching as it indicates that a demand of true personalized medicine for TBI is necessary to deliver the right therapeutic at the right time for the right patient.
Collapse
Affiliation(s)
- Vimala N. Bharadwaj
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, Arizona, USA
| | - Connor Copeland
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, Arizona, USA
| | - Ethan Mathew
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, Arizona, USA
| | - Jason Newbern
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Trent R. Anderson
- Basic Medical Sciences, University of Arizona, College of Medicine–Phoenix, Phoenix, Arizona, USA
| | - Jonathan Lifshitz
- Department of Child Health, University of Arizona, College of Medicine–Phoenix, Phoenix, Arizona, USA
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA
- Phoenix VA Health Care System, Phoenix, Arizona, USA
| | - Vikram D. Kodibagkar
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, Arizona, USA
| | - Sarah E. Stabenfeldt
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
11
|
Abrahamson EE, Poloyac SM, Dixon CE, Dekosky ST, Ikonomovic MD. Acute and chronic effects of single dose memantine after controlled cortical impact injury in adult rats. Restor Neurol Neurosci 2020; 37:245-263. [PMID: 31177251 DOI: 10.3233/rnn-190909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Altered glutamatergic neurotransmission after traumatic brain injury (TBI) contributes to excitotoxic cell damage and death. Prevention or suppression of such changes is a desirable goal for treatment of TBI. Memantine (3,5-dimethyl-1-adamantanamine), an uncompetitive NMDA receptor antagonist with voltage-dependent open channel blocking kinetics, was reported to be neuroprotective in preclinical models of excitotoxicity, brain ischemia, and in TBI when administered prophylactically, immediately, or within minutes after injury. METHODS The current study examined effects of memantine administered by single intraperitoneal injection to adult male rats at a more clinically relevant delay of one hour after moderate-severe controlled cortical impact (CCI) injury or sham surgery. Histopathology was assessed on days 1, 7, 21, and 90, vestibulomotor function (beam balance and beam walk) was assessed on days 1-5 and 71-75, and spatial memory (Morris water maze test, MWM) was assessed on days 14-21 and 83-90 after CCI injury or sham surgery. RESULTS When administered at 10 mg/kg, but not 2.5 or 5 mg/kg, memantine preserved cortical tissue and reduced neuronal degeneration 1 day after injury, and attenuated loss of synaptophysin immunoreactivity in the hippocampus 7 days after injury. No effects of 10 mg/kg memantine were observed on histopathology at 21 and 90 days after CCI injury or sham surgery, or on vestibulomotor function and spatial memory acquisition assessed during any of the testing periods. However, 10 mg/kg memantine resulted in trends for improved search strategy in the MWM memory retention probe trial. CONCLUSIONS Administration of memantine at a clinically-relevant delay after moderate-severe CCI injury has beneficial effects on acute outcomes, while more significant improvement on subacute and chronic outcomes may require repeated drug administration or its combination with another therapy.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh PA, USA.,Department of Neurology, University of Pittsburgh, Pittsburgh PA, USA
| | - Samuel M Poloyac
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh PA, USA
| | - C Edward Dixon
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh PA, USA.,Department of Neurosurgery, University of Pittsburgh, Pittsburgh PA, USA
| | - Steven T Dekosky
- Department of Neurology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh PA, USA.,Department of Neurology, University of Pittsburgh, Pittsburgh PA, USA.,Department of Psychiatry, University of Pittsburgh, Pittsburgh PA, USA
| |
Collapse
|
12
|
Sharma S, Ifergan I, Kurz JE, Linsenmeier RA, Xu D, Cooper JG, Miller SD, Kessler JA. Intravenous Immunomodulatory Nanoparticle Treatment for Traumatic Brain Injury. Ann Neurol 2020; 87:442-455. [PMID: 31925846 PMCID: PMC7296512 DOI: 10.1002/ana.25675] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 12/29/2019] [Accepted: 01/05/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVE There are currently no definitive disease-modifying therapies for traumatic brain injury (TBI). In this study, we present a strong therapeutic candidate for TBI, immunomodulatory nanoparticles (IMPs), which ablate a specific subset of hematogenous monocytes (hMos). We hypothesized that prevention of infiltration of these cells into brain acutely after TBI would attenuate secondary damage and preserve anatomic and neurologic function. METHODS IMPs, composed of US Food and Drug Administration-approved 500nm carboxylated-poly(lactic-co-glycolic) acid, were infused intravenously into wild-type C57BL/6 mice following 2 different models of experimental TBI, controlled cortical impact (CCI), and closed head injury (CHI). RESULTS IMP administration resulted in remarkable preservation of both tissue and neurological function in both CCI and CHI TBI models in mice. After acute treatment, there was a reduction in the number of immune cells infiltrating into the brain, mitigation of the inflammatory status of the infiltrating cells, improved electrophysiologic visual function, improved long-term motor behavior, reduced edema formation as assessed by magnetic resonance imaging, and reduced lesion volumes on anatomic examination. INTERPRETATION Our findings suggest that IMPs are a clinically translatable acute intervention for TBI with a well-defined mechanism of action and beneficial anatomic and physiologic preservation and recovery. Ann Neurol 2020;87:442-455.
Collapse
Affiliation(s)
- Sripadh Sharma
- Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago
| | - Igal Ifergan
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago
| | - Jonathan E Kurz
- Davee Pediatric Neurocritical Care Program, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago
| | - Robert A Linsenmeier
- Department of Biomedical Engineering, Northwestern University, Evanston, IL
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - Dan Xu
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago
| | - John G Cooper
- Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago
| | - John A Kessler
- Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago
| |
Collapse
|
13
|
Jiang Q, Chen J, Long X, Yao X, Zou X, Yang Y, Huang G, Zhang H. Phillyrin protects mice from traumatic brain injury by inhibiting the inflammation of microglia via PPARγ signaling pathway. Int Immunopharmacol 2020; 79:106083. [PMID: 31923823 DOI: 10.1016/j.intimp.2019.106083] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/04/2019] [Accepted: 11/24/2019] [Indexed: 12/29/2022]
Abstract
The neuroinflammatory response induced by microglia plays a vital role in causing secondary brain damage after traumatic brain injury (TBI). Previous studies have found that the improved regulation of activated microglia could reduce neurological damage post-TBI. Phillyrin (Phi) is one of the main active ingredients extracted from the fruits of the medicinal plant Forsythia suspensa (Thunb.) with anti-inflammatory effects. Our study attempted to investigate the effects of phillyrin on microglial activation and neuron damage after TBI. The TBI model was applied to induce brain injury in mice, and neurological scores, brain water content, hematoxylin and eosin staining and Nissl staining were employed to determine the neuroprotective effects of phillyrin. Immunofluorescent staining and western blot analysis were used to detect nuclear factor-kappa B (NF-κB) and peroxisome proliferator-activated receptor gamma (PPARγ) expression and nuclear translocation, and the inflammation-related proteins and mRNAs were assessed by western blot analysis and quantitative real-time PCR. The results revealed that phillyrin not only inhibited the proinflammatory response induced by activated microglia but also attenuated neurological impairment and brain edema in vivo in a mouse TBI model. Additionally, phillyrin suppressed the phosphorylation of NF-κB in microglia after TBI insult. These effects of phillyrin were mostly abolished by the antagonist of PPARγ. Our results reveal that phillyrin could prominently inhibit the inflammation of microglia via the PPARγ signaling pathway, thus leading to potential neuroprotective treatment after traumatic brain injury.
Collapse
Affiliation(s)
- Qian Jiang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Jun Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Xiaobing Long
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Xiaolong Yao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China; Department of Neurosurgery, Taikang Tongji Hospital, Wuhan 430050, PR China
| | - Xin Zou
- Department of Traditional Chinese Medicine, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Yiping Yang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China
| | - Guangying Huang
- Department of Traditional Chinese Medicine, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China.
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
14
|
Lu L, Mao H. Quantifying the Effect of Repeated Impacts and Lateral Tip Movements on Brain Responses during Controlled Cortical Impact. J Neurotrauma 2019; 36:1828-1835. [DOI: 10.1089/neu.2018.5929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Lihong Lu
- Department of Mechanical and Materials Engineering, Western University, London, Ontario, Canada
| | - Haojie Mao
- Department of Mechanical and Materials Engineering, Western University, London, Ontario, Canada
| |
Collapse
|
15
|
Zhang Y, Chopp M, Rex CS, Simmon VF, Sarraf ST, Zhang ZG, Mahmood A, Xiong Y. A Small Molecule Spinogenic Compound Enhances Functional Outcome and Dendritic Spine Plasticity in a Rat Model of Traumatic Brain Injury. J Neurotrauma 2018; 36:589-600. [PMID: 30014757 DOI: 10.1089/neu.2018.5790] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The tetra (ethylene glycol) derivative of benzothiazole aniline (SPG101) has been shown to improve dendritic spine density and cognitive memory in the triple transgenic mouse model of Alzheimer disease (AD) when administered intraperitoneally. The present study was designed to investigate the therapeutic effects of SPG101 on dendritic spine density and morphology and sensorimotor and cognitive functional recovery in a rat model of traumatic brain injury (TBI) induced by controlled cortical impact (CCI). Young adult male Wistar rats with CCI were randomly divided into the following two groups (n = 7/group): (1) Vehicle, and (2) SPG101. SPG101 (30 mg/kg) dissolved in vehicle (1% dimethyl sulfoxide in phosphate buffered saline) or Vehicle were intraperitoneally administered starting at 1 h post-injury and once daily for the next 34 days. Sensorimotor deficits were assessed using a modified neurological severity score and adhesive removal and foot fault tests. Cognitive function was measured by Morris water maze, novel object recognition (NOR), and three-chamber social recognition tests. The animals were sacrificed 35 days after injury, and their brains were processed for measurement of dendritic spine density and morphology using ballistic dye labeling. Compared with the vehicle treatment, SPG101 treatment initiated 1 h post-injury significantly improved sensorimotor functional recovery (days 7-35, p < 0.0001), spatial learning (days 32-35, p < 0.0001), NOR (days 14 and 35, p < 0.0001), social recognition (days 14 and 35, p < 0.0001). Further, treatment significantly increased dendritic spine density in the injured cortex (p < 0.05), decreased heterogeneous distribution of spine lengths in the injured cortex and hippocampus (p < 0.0001), modifications that are associated with the promotion of spine maturation in these brain regions. In summary, treatment with SPG101 initiated 1 h post-injury and continued for an additional 34 days improves both sensorimotor and cognitive functional recovery, indicating that SPG101 acts as a spinogenic agent and may have potential as a novel treatment of TBI.
Collapse
Affiliation(s)
- Yanlu Zhang
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Michael Chopp
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan.,3 Department of Physics, Oakland University , Rochester, Michigan
| | | | | | | | - Zheng Gang Zhang
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan
| | - Asim Mahmood
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Ye Xiong
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| |
Collapse
|
16
|
Aneja RK, Alcamo AM, Cummings J, Vagni V, Feldman K, Wang Q, Dixon CE, Billiar TR, Kochanek PM. Lack of Benefit on Brain Edema, Blood-Brain Barrier Permeability, or Cognitive Outcome in Global Inducible High Mobility Group Box 1 Knockout Mice Despite Tissue Sparing after Experimental Traumatic Brain Injury. J Neurotrauma 2018; 36:360-369. [PMID: 30045665 DOI: 10.1089/neu.2018.5664] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a prototypical danger-associated molecular pattern molecule that is considered a late mediator of neuro-inflammation after traumatic brain injury (TBI). Prior studies have suggested that targeting HMGB1 may lead to neuroprotective effects, but none of these studies have reported cognitive outcomes. We hypothesized that loss of HMGB1 before and after TBI would markedly attenuate post-traumatic brain edema, blood-brain barrier (BBB) permeability, improve functional deficits and long-term neuropathology versus control mice. Using the controlled cortical impact model and conditional global HMGB1 knockout (HMGB1 KO) mice, we demonstrate that there was a neuroprotective effect seen in the HMGB1 KO versus wild-type control evidenced by a significant reduction in contusion volume. However, two surprising findings were 1) the lack of benefit on either post-traumatic brain edema or BBB permeability, and 2) that spatial memory performance was impaired in HMGB1 KO naïve mice such that the behavioral effects of HMGB1 deletion in uninjured naïve mice were similar to those observed after TBI. Our data suggest the possibility that the role of HMGB1 in TBI is a "double-edged sword"; that is, despite the benefits on selected aspects of secondary injury, the sustained absence of HMGB1 may impair cognitive function, even in naïve mice. Given the pleiotropic actions of extracellular and intracellular HMGB1, when evaluating the potential use of therapies targeting HMGB1, effects on long-term cognitive outcome should be carefully evaluated. It also may be prudent in future studies to examine cell-specific effects of manipulating the HMGB1 pathway.
Collapse
Affiliation(s)
- Rajesh K Aneja
- 1 Department of Critical Care Medicine, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania.,2 Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Alicia M Alcamo
- 1 Department of Critical Care Medicine, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania.,2 Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Jessica Cummings
- 1 Department of Critical Care Medicine, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania.,2 Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Vincent Vagni
- 1 Department of Critical Care Medicine, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Keri Feldman
- 1 Department of Critical Care Medicine, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Qingde Wang
- 3 Department of Surgery, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - C Edward Dixon
- 4 Department of Neurological Surgery, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Timothy R Billiar
- 3 Department of Surgery, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- 1 Department of Critical Care Medicine, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| |
Collapse
|
17
|
Jha RM, Molyneaux BJ, Jackson TC, Wallisch JS, Park SY, Poloyac S, Vagni VA, Janesko-Feldman KL, Hoshitsuki K, Minnigh MB, Kochanek PM. Glibenclamide Produces Region-Dependent Effects on Cerebral Edema in a Combined Injury Model of Traumatic Brain Injury and Hemorrhagic Shock in Mice. J Neurotrauma 2018; 35:2125-2135. [PMID: 29648981 PMCID: PMC6098411 DOI: 10.1089/neu.2016.4696] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cerebral edema is critical to morbidity/mortality in traumatic brain injury (TBI) and is worsened by hypotension. Glibenclamide may reduce cerebral edema by inhibiting sulfonylurea receptor-1 (Sur1); its effect on diffuse cerebral edema exacerbated by hypotension/resuscitation is unknown. We aimed to determine if glibenclamide improves pericontusional and/or diffuse edema in controlled cortical impact (CCI) (5m/sec, 1 mm depth) plus hemorrhagic shock (HS) (35 min), and compare its effects in CCI alone. C57BL/6 mice were divided into five groups (n = 10/group): naïve, CCI+vehicle, CCI+glibenclamide, CCI+HS+vehicle, and CCI+HS+glibenclamide. Intravenous glibenclamide (10 min post-injury) was followed by a subcutaneous infusion for 24 h. Brain edema in injured and contralateral hemispheres was subsequently quantified (wet-dry weight). This protocol brain water (BW) = 80.4% vehicle vs. 78.3% naïve, p < 0.01) but was not reduced by glibenclamide (I%BW = 80.4%). Ipsilateral edema also developed in CCI alone (I%BW = 80.2% vehicle vs. 78.3% naïve, p < 0.01); again unaffected by glibenclamide (I%BW = 80.5%). Contralateral (C) %BW in CCI+HS was increased in vehicle (78.6%) versus naive (78.3%, p = 0.02) but unchanged in CCI (78.3%). At 24 h, glibenclamide treatment in CCI+HS eliminated contralateral cerebral edema (C%BW = 78.3%) with no difference versus naïve. By 72 h, contralateral cerebral edema had resolved (C%BW = 78.5 ± 0.09% vehicle vs. 78.3 ± 0.05% naïve). Glibenclamide decreased 24 h contralateral cerebral edema in CCI+HS. This beneficial effect merits additional exploration in the important setting of TBI with polytrauma, shock, and resuscitation. Contralateral edema did not develop in CCI alone. Surprisingly, 24 h of glibenclamide treatment failed to decrease ipsilateral edema in either model. Interspecies dosing differences versus prior studies may play an important role in these findings. Mechanisms underlying brain edema may differ regionally, with pericontusional/osmolar swelling refractory to glibenclamide but diffuse edema (via Sur1) from combined injury and/or resuscitation responsive to this therapy. TBI phenotype may mandate precision medicine approaches to treat brain edema.
Collapse
Affiliation(s)
- Ruchira M. Jha
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurosurgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bradley J. Molyneaux
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurosurgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Travis C. Jackson
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessica S. Wallisch
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Seo-Young Park
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Samuel Poloyac
- Department of Pharmacy and Therapeutics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vincent A. Vagni
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Keri L. Janesko-Feldman
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Keito Hoshitsuki
- Department of Pharmacy and Therapeutics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - M. Beth Minnigh
- Department of Pharmacy and Therapeutics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patrick M. Kochanek
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Anesthesia, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
18
|
Intraoperative contrast-enhanced ultrasonography for microcirculatory evaluation in rhesus monkey with spinal cord injury. Oncotarget 2018; 8:40756-40764. [PMID: 28489576 PMCID: PMC5522262 DOI: 10.18632/oncotarget.17252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/03/2017] [Indexed: 11/25/2022] Open
Abstract
This study tried to quantify spinal cord perfusion by using contrast-enhanced ultrasound (CEUS) in rhesus monkey models with acute spinal cord injury. Acute spinal cord perfusion after injury was detected by CEUS, coupling with conventional ultrasound (US) and Color Doppler US (CDFI). Time-intensity curves and perfusion parameters were obtained by autotracking contrast quantification (ACQ) software in the epicenter and adjacent regions of injury, respectively. Neurological and histological examinations were performed to confirm the severity of injury. US revealed spinal cords were hypoechoic and homogeneous, whereas dura maters, pia maters, and cerebral aqueducts were hyperechoic. After spinal cord contusion, the injured spinal cord was hyperechoic on US, and intramedullary vessels of adjacent region of injury were increased and dilated on CDFI. On CEUS hypoperfusion were found in the epicenter of injury, while hyperperfusion in its adjacent region. Quantitative analysis showed that peak intensity (PI) decreased in epicenters of injury but significantly increased in adjacent regions at all time points (p < 0.05). Functional evaluation demonstrated significant deterioration compared to pre-contusion (p < 0.05). Quantitative analysis with CEUS is a promising method for monitoring perfusion changes of spinal cord injury in overall views and real-time.
Collapse
|
19
|
Abutarboush R, Mullah SH, Saha BK, Haque A, Walker PB, Aligbe C, Pappas G, Tran Ho LTV, Arnaud FG, Auker CR, McCarron RM, Scultetus AH, Moon-Massat P. Brain oxygenation with a non-vasoactive perfluorocarbon emulsion in a rat model of traumatic brain injury. Microcirculation 2018; 25:e12441. [DOI: 10.1111/micc.12441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Rania Abutarboush
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Saad H. Mullah
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Biswajit K. Saha
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Ashraful Haque
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Peter B. Walker
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Chioma Aligbe
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | - Georgina Pappas
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | | | - Francoise G. Arnaud
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | - Charles R. Auker
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Richard M. McCarron
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | - Anke H. Scultetus
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | - Paula Moon-Massat
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| |
Collapse
|
20
|
Chiluwal A, Narayan RK, Chaung W, Mehan N, Wang P, Bouton CE, Golanov EV, Li C. Neuroprotective Effects of Trigeminal Nerve Stimulation in Severe Traumatic Brain Injury. Sci Rep 2017; 7:6792. [PMID: 28754973 PMCID: PMC5533766 DOI: 10.1038/s41598-017-07219-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/23/2017] [Indexed: 12/25/2022] Open
Abstract
Following traumatic brain injury (TBI), ischemia and hypoxia play a major role in further worsening of the damage, a process referred to as 'secondary injury'. Protecting neurons from causative factors of secondary injury has been the guiding principle of modern TBI management. Stimulation of trigeminal nerve induces pressor response and improves cerebral blood flow (CBF) by activating the rostral ventrolateral medulla. Moreover, it causes cerebrovasodilation through the trigemino-cerebrovascular system and trigemino-parasympathetic reflex. These effects are capable of increasing cerebral perfusion, making trigeminal nerve stimulation (TNS) a promising strategy for TBI management. Here, we investigated the use of electrical TNS for improving CBF and brain oxygen tension (PbrO2), with the goal of decreasing secondary injury. Severe TBI was produced using controlled cortical impact (CCI) in a rat model, and TNS treatment was delivered for the first hour after CCI. In comparison to TBI group, TBI animals with TNS treatment demonstrated significantly increased systemic blood pressure, CBF and PbrO2 at the hyperacute phase of TBI. Furthermore, rats in TNS-treatment group showed significantly reduced brain edema, blood-brain barrier disruption, lesion volume, and brain cortical levels of TNF-α and IL-6. These data provide strong early evidence that TNS could be an effective neuroprotective strategy.
Collapse
Affiliation(s)
- Amrit Chiluwal
- Northwell Neuromonitoring Laboratory, The Feinstein Institute for Medical Research, Manhasset, NY, USA
- Department of Neurosurgery, Hofstra Northwell School of Medicine, Hempstead, NY, USA
| | - Raj K Narayan
- Northwell Neuromonitoring Laboratory, The Feinstein Institute for Medical Research, Manhasset, NY, USA
- Department of Neurosurgery, Hofstra Northwell School of Medicine, Hempstead, NY, USA
- Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Wayne Chaung
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Neal Mehan
- Northwell Neuromonitoring Laboratory, The Feinstein Institute for Medical Research, Manhasset, NY, USA
- Department of Neurosurgery, Hofstra Northwell School of Medicine, Hempstead, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Chad E Bouton
- Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Eugene V Golanov
- Department of Neurosurgery, The Houston Methodist Research Institute, Houston, Texas, USA
| | - Chunyan Li
- Northwell Neuromonitoring Laboratory, The Feinstein Institute for Medical Research, Manhasset, NY, USA.
- Department of Neurosurgery, Hofstra Northwell School of Medicine, Hempstead, NY, USA.
- Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
21
|
Blood-brain barrier breakdown and neovascularization processes after stroke and traumatic brain injury. Curr Opin Neurol 2016; 28:556-64. [PMID: 26402408 DOI: 10.1097/wco.0000000000000248] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Angiogenesis or vascular reorganization plays a role in recovery after stroke and traumatic brain injury (TBI). In this review, we have focused on two major events that occur during stroke and TBI from a vascular perspective - what is the process and time course of blood-brain barrier (BBB) breakdown? and how does the surrounding vasculature recover and facilitate repair? RECENT FINDINGS Despite differences in the primary injury, the BBB changes overlap between stroke and TBI. Disruption of BBB involves a series of events: formation of caveolae, trans and paracellular disruption, tight junction breakdown and vascular disruption. Confounding factors that need careful assessment and standardization are the severity, duration and extent of the stroke and TBI that influences BBB disruption. Vascular repair proceeds through long-term neovascularization processes: angiogenesis, arteriogenesis and vasculogenesis. Enhancing each of these processes may impart beneficial effects in endogenous recovery. SUMMARY Our understanding of BBB breakdown acutely after the cerebrovascular injury has come a long way; however, we lack a clear understanding of the course of BBB disruption and BBB recovery and the evolution of individual cellular events associated with BBB change. Neovascularization responses have been widely studied in stroke for their role in functional recovery but the role of vascular reorganization after TBI in recovery is much less defined.
Collapse
|
22
|
Osier ND, Dixon CE. The Controlled Cortical Impact Model: Applications, Considerations for Researchers, and Future Directions. Front Neurol 2016; 7:134. [PMID: 27582726 PMCID: PMC4987613 DOI: 10.3389/fneur.2016.00134] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/02/2016] [Indexed: 12/26/2022] Open
Abstract
Controlled cortical impact (CCI) is a mechanical model of traumatic brain injury (TBI) that was developed nearly 30 years ago with the goal of creating a testing platform to determine the biomechanical properties of brain tissue exposed to direct mechanical deformation. Initially used to model TBIs produced by automotive crashes, the CCI model rapidly transformed into a standardized technique to study TBI mechanisms and evaluate therapies. CCI is most commonly produced using a device that rapidly accelerates a rod to impact the surgically exposed cortical dural surface. The tip of the rod can be varied in size and geometry to accommodate scalability to difference species. Typically, the rod is actuated by a pneumatic piston or electromagnetic actuator. With some limits, CCI devices can control the velocity, depth, duration, and site of impact. The CCI model produces morphologic and cerebrovascular injury responses that resemble certain aspects of human TBI. Commonly observed are graded histologic and axonal derangements, disruption of the blood-brain barrier, subdural and intra-parenchymal hematoma, edema, inflammation, and alterations in cerebral blood flow. The CCI model also produces neurobehavioral and cognitive impairments similar to those observed clinically. In contrast to other TBI models, the CCI device induces a significantly pronounced cortical contusion, but is limited in the extent to which it models the diffuse effects of TBI; a related limitation is that not all clinical TBI cases are characterized by a contusion. Another perceived limitation is that a non-clinically relevant craniotomy is performed. Biomechanically, this is irrelevant at the tissue level. However, craniotomies are not atraumatic and the effects of surgery should be controlled by including surgical sham control groups. CCI devices have also been successfully used to impact closed skulls to study mild and repetitive TBI. Future directions for CCI research surround continued refinements to the model through technical improvements in the devices (e.g., minimizing mechanical sources of variation). Like all TBI models, publications should report key injury parameters as outlined in the NIH common data elements (CDEs) for pre-clinical TBI.
Collapse
Affiliation(s)
- Nicole D. Osier
- Department of Acute and Tertiary Care, University of Pittsburgh School of Nursing, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, Pittsburgh, PA, USA
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, Pittsburgh, PA, USA
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Jullienne A, Obenaus A, Ichkova A, Savona-Baron C, Pearce WJ, Badaut J. Chronic cerebrovascular dysfunction after traumatic brain injury. J Neurosci Res 2016; 94:609-22. [PMID: 27117494 PMCID: PMC5415378 DOI: 10.1002/jnr.23732] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 02/11/2016] [Accepted: 02/28/2016] [Indexed: 12/12/2022]
Abstract
Traumatic brain injuries (TBI) often involve vascular dysfunction that leads to long-term alterations in physiological and cognitive functions of the brain. Indeed, all the cells that form blood vessels and that are involved in maintaining their proper function can be altered by TBI. This Review focuses on the different types of cerebrovascular dysfunction that occur after TBI, including cerebral blood flow alterations, autoregulation impairments, subarachnoid hemorrhage, vasospasms, blood-brain barrier disruption, and edema formation. We also discuss the mechanisms that mediate these dysfunctions, focusing on the cellular components of cerebral blood vessels (endothelial cells, smooth muscle cells, astrocytes, pericytes, perivascular nerves) and their known and potential roles in the secondary injury cascade. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California
| | - Andre Obenaus
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, California
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California Riverside, Riverside, California
| | | | | | - William J Pearce
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Jerome Badaut
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, California
- CNRS UMR5287, University of Bordeaux, Bordeaux, France
| |
Collapse
|
24
|
Hernandez A, Donovan V, Grinberg YY, Obenaus A, Carson MJ. Differential detection of impact site versus rotational site injury by magnetic resonance imaging and microglial morphology in an unrestrained mild closed head injury model. J Neurochem 2016; 136 Suppl 1:18-28. [PMID: 26806371 PMCID: PMC5047732 DOI: 10.1111/jnc.13402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 01/31/2023]
Abstract
Seventy‐five percent of all traumatic brain injuries are mild and do not cause readily visible abnormalities on routine medical imaging making it difficult to predict which individuals will develop unwanted clinical sequelae. Microglia are brain‐resident macrophages and early responders to brain insults. Their activation is associated with changes in morphology or expression of phenotypic markers including P2Y12 and major histocompatibility complex class II. Using a murine model of unrestrained mild closed head injury (mCHI), we used microglia as reporters of acute brain injury at sites of impact versus sites experiencing rotational stress 24 h post‐mCHI. Consistent with mild injury, a modest 20% reduction in P2Y12 expression was detected by quantitative real‐time PCR (qPCR) analysis but only in the impacted region of the cortex. Furthermore, neither an influx of blood‐derived immune cells nor changes in microglial expression of CD45, TREM1, TREM2, major histocompatibility complex class II or CD40 were detected. Using magnetic resonance imaging (MRI), small reductions in T2 weighted values were observed but only near the area of impact and without overt tissue damage (blood deposition, edema). Microglial morphology was quantified without cryosectioning artifacts using ScaleA2 clarified brains from CX3CR1‐green fluorescence protein (GFP) mice. The cortex rostral to the mCHI impact site receives greater rotational stress but neither MRI nor molecular markers of microglial activation showed significant changes from shams in this region. However, microglia in this rostral region did display signs of morphologic activation equivalent to that observed in severe CHI. Thus, mCHI‐triggered rotational stress is sufficient to cause injuries undetectable by routine MRI that could result in altered microglial surveillance of brain homeostasis.
Acute changes in microglial morphology reveal brain responses to unrestrained mild traumatic brain injury
In areas subjected to rotational stress distant from impact site In the absence of detectable changes in standard molecular indicators of brain damage, inflammation or microglial activation. That might result in decreased surveillance of brain function and increased susceptibility to subsequent brain insults.
Collapse
Affiliation(s)
- Alfredo Hernandez
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,MarcU Program, University of California Riverside, Riverside, California, USA.,Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, California, USA
| | - Virgina Donovan
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, California, USA.,Cell Molecular and Developmental Biology Program, University of California Riverside, Riverside, California, USA.,Loma Linda University School of Medicine, Loma Linda California, Loma Linda, CA, USA
| | - Yelena Y Grinberg
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, California, USA
| | - Andre Obenaus
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,Cell Molecular and Developmental Biology Program, University of California Riverside, Riverside, California, USA.,Loma Linda University School of Medicine, Loma Linda California, Loma Linda, CA, USA
| | - Monica J Carson
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, California, USA.,Cell Molecular and Developmental Biology Program, University of California Riverside, Riverside, California, USA
| |
Collapse
|
25
|
Bondi CO, Semple BD, Noble-Haeusslein LJ, Osier ND, Carlson SW, Dixon CE, Giza CC, Kline AE. Found in translation: Understanding the biology and behavior of experimental traumatic brain injury. Neurosci Biobehav Rev 2015; 58:123-46. [PMID: 25496906 PMCID: PMC4465064 DOI: 10.1016/j.neubiorev.2014.12.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 10/26/2014] [Accepted: 12/02/2014] [Indexed: 12/14/2022]
Abstract
The aim of this review is to discuss in greater detail the topics covered in the recent symposium entitled "Traumatic brain injury: laboratory and clinical perspectives," presented at the 2014 International Behavioral Neuroscience Society annual meeting. Herein, we review contemporary laboratory models of traumatic brain injury (TBI) including common assays for sensorimotor and cognitive behavior. New modalities to evaluate social behavior after injury to the developing brain, as well as the attentional set-shifting test (AST) as a measure of executive function in TBI, will be highlighted. Environmental enrichment (EE) will be discussed as a preclinical model of neurorehabilitation, and finally, an evidence-based approach to sports-related concussion will be considered. The review consists predominantly of published data, but some discussion of ongoing or future directions is provided.
Collapse
Affiliation(s)
- Corina O Bondi
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bridgette D Semple
- Neurological Surgery and the Graduate Program in Physical Medicine & Rehabilitation Science, University of California, San Francisco, San Francisco, CA, United States; Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC, Australia
| | - Linda J Noble-Haeusslein
- Neurological Surgery and the Graduate Program in Physical Medicine & Rehabilitation Science, University of California, San Francisco, San Francisco, CA, United States
| | - Nicole D Osier
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; School of Nursing, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shaun W Carlson
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - C Edward Dixon
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States; Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - Christopher C Giza
- Pediatric Neurology and Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States; UCLA Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anthony E Kline
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States; Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
26
|
Schwarzmaier SM, Gallozzi M, Plesnila N. Identification of the Vascular Source of Vasogenic Brain Edema following Traumatic Brain Injury Using In Vivo 2-Photon Microscopy in Mice. J Neurotrauma 2015; 32:990-1000. [PMID: 25585052 DOI: 10.1089/neu.2014.3775] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Vasogenic brain edema due to vascular leakage is one of the most important factors determining the clinical outcome of patients following acute brain injury. To date, performing a detailed in vivo quantification of vascular leakage has not been possible. Here, we used in vivo 2-photon microscopy (2-PM) to determine the spatial (3D) and temporal development of vasogenic brain edema following traumatic brain injury (TBI) in mice; in addition, we identified the vessel types involved in vascular leakage. Thirteen male Tie2-GFP mice (6-8 weeks old) were subjected to controlled cortical impact (CCI) or a sham operation; subsequently, a cranial window was prepared adjacent to the injury site, and tetramethylrhodamine-dextran (TMRM, 40 mg/kg, MW 40,000) was injected intravenously to visualize blood plasma leakage. Parenchymal fluorescence intensity was monitored in three regions for 2-4 h post-CCI, reaching from the surface of the brain to a depth of 300 μm, and TMRM leakage was measured as an increase in TMRM fluorescence intensity outside the vessel lumen and in the parenchyma. In the CCI group, vascular leakage was detected in all investigated regions as early as 2.5 h post-injury. This leakage increased over time and was more pronounced proximal to the primary contusion. Both arterioles and venules contributed similarly to brain edema formation and their contribution was independent of vessel size; however, capillaries were the major contributor to leakage. In summary, using 2-PM to perform in vivo 3D deep-brain imaging, we found that TBI induces vascular leakage from capillaries, venules, and arterioles. Thus, all three vessel types are involved in trauma-induced brain edema and should be considered when developing novel therapies for preventing vasogenic brain edema.
Collapse
Affiliation(s)
- Susanne M Schwarzmaier
- 1 Department of Neurodegeneration, Royal College of Surgeons in Ireland (RCSI) , Dublin, Ireland .,2 Department of Anesthesiology, University of Munich Medical Center , Germany .,3 Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center , Germany
| | - Micaela Gallozzi
- 1 Department of Neurodegeneration, Royal College of Surgeons in Ireland (RCSI) , Dublin, Ireland
| | - Nikolaus Plesnila
- 1 Department of Neurodegeneration, Royal College of Surgeons in Ireland (RCSI) , Dublin, Ireland .,3 Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center , Germany .,4 Munich Cluster of Symptoms Neurology (Synergy) , Munich, Germany
| |
Collapse
|
27
|
Preservation of the blood brain barrier and cortical neuronal tissue by liraglutide, a long acting glucagon-like-1 analogue, after experimental traumatic brain injury. PLoS One 2015; 10:e0120074. [PMID: 25822252 PMCID: PMC4379006 DOI: 10.1371/journal.pone.0120074] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 02/02/2015] [Indexed: 01/04/2023] Open
Abstract
Cerebral edema is a common complication following moderate and severe traumatic brain injury (TBI), and a significant risk factor for development of neuronal death and deterioration of neurological outcome. To this date, medical approaches that effectively alleviate cerebral edema and neuronal death after TBI are not available. Glucagon-like peptide-1 (GLP-1) has anti-inflammatory properties on cerebral endothelium and exerts neuroprotective effects. Here, we investigated the effects of GLP-1 on secondary injury after moderate and severe TBI. Male Sprague Dawley rats were subjected either to TBI by Controlled Cortical Impact (CCI) or sham surgery. After surgery, vehicle or a GLP-1 analogue, Liraglutide, were administered subcutaneously twice daily for two days. Treatment with Liraglutide (200 μg/kg) significantly reduced cerebral edema in pericontusional regions and improved sensorimotor function 48 hours after CCI. The integrity of the blood-brain barrier was markedly preserved in Liraglutide treated animals, as determined by cerebral extravasation of Evans blue conjugated albumin. Furthermore, Liraglutide reduced cortical tissue loss, but did not affect tissue loss and delayed neuronal death in the thalamus on day 7 post injury. Together, our data suggest that the GLP-1 pathway might be a promising target in the therapy of cerebral edema and cortical neuronal injury after moderate and severe TBI.
Collapse
|
28
|
Thal SC, Neuhaus W. The blood-brain barrier as a target in traumatic brain injury treatment. Arch Med Res 2014; 45:698-710. [PMID: 25446615 DOI: 10.1016/j.arcmed.2014.11.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI) is one of the most frequent causes of death in the young population. Several clinical trials have unsuccessfully focused on direct neuroprotective therapies. Recently immunotherapeutic strategies shifted into focus of translational research in acute CNS diseases. Cross-talk between activated microglia and blood-brain barrier (BBB) could initiate opening of the BBB and subsequent recruitment of systemic immune cells and mediators into the brain. Stabilization of the BBB after TBI could be a promising strategy to limit neuronal inflammation, secondary brain damage and acute neurodegeneration. This review provides an overview on the pathophysiology of TBI and brain edema formation including definitions and classification of TBI, current clinical treatment strategies, as well as current understanding on the underlying cellular processes. A summary of in vivo and in vitro models to study different aspects of TBI is presented. Three mechanisms proposed for stabilization of the BBB, myosin light chain kinases, glucocorticoid receptors and peroxisome proliferator-activated receptors are reviewed for their influence on barrier-integrity and outcome after TBI. In conclusion, the BBB is recommended as a promising target for the treatment of traumatic brain injury, and it is suggested that a combination of BBB stabilization and neuroprotectants may improve therapeutic success.
Collapse
Affiliation(s)
- Serge C Thal
- Department of Anesthesia and Critical Care, Johannes Gutenberg University, Mainz, Germany
| | - Winfried Neuhaus
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse, Vienna, Austria; Department of Anesthesia and Critical Care, University Hospital Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
29
|
Schwarzmaier SM, Plesnila N. Contributions of the immune system to the pathophysiology of traumatic brain injury - evidence by intravital microscopy. Front Cell Neurosci 2014; 8:358. [PMID: 25408636 PMCID: PMC4219391 DOI: 10.3389/fncel.2014.00358] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) results in immediate brain damage that is caused by the mechanical impact and is non-reversible. This initiates a cascade of delayed processes which cause additional—secondary—brain damage. Among these secondary mechanisms, the inflammatory response is believed to play an important role, mediating actions that can have both protective and detrimental effects on the progression of secondary brain damage. Histological data generated extensive information; however, this is only a snapshot of processes that are, in fact, very dynamic. In contrast, in vivo microscopy provides detailed insight into the temporal and spatial patterns of cellular dynamics. In this review, we aim to summarize data which was generated by in vivo microscopy, specifically investigating the immune response following brain trauma, and its potential effects on secondary brain damage.
Collapse
Affiliation(s)
- Susanne M Schwarzmaier
- Department of Anesthesiology, University of Munich Medical Center Munich, Germany ; Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center Munich, Germany ; Munich Cluster of Systems Neurology Munich, Germany
| |
Collapse
|
30
|
Lu H, Lei X. The apparent diffusion coefficient does not reflect cytotoxic edema on the uninjured side after traumatic brain injury. Neural Regen Res 2014; 9:973-7. [PMID: 25206920 PMCID: PMC4146228 DOI: 10.4103/1673-5374.133150] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2014] [Indexed: 01/20/2023] Open
Abstract
After traumatic brain injury, vasogenic and cytotoxic edema appear sequentially on the involved side. Neuroimaging investigations of edema on the injured side have employed apparent diffusion coefficient measurements in diffusion tensor imaging. We investigated the changes occurring on the injured and uninjured sides using diffusion tensor imaging/apparent diffusion coefficient and histological samples in rats. We found that, on the injured side, that vasogenic edema appeared at 1 hour and intracellular edema appeared at 3 hours. Mixed edema was observed at 6 hours, worsening until 12–24 hours post-injury. Simultaneously, microglial cells proliferated at the trauma site. Apparent diffusion coefficient values increased at 1 hour, decreased at 6 hours, and increased at 12 hours. The uninjured side showed no significant pathological change at 1 hour after injury. Cytotoxic edema appeared at 3 hours, and vasogenic edema was visible at 6 hours. Cytotoxic edema persisted, but vasogenic edema tended to decrease after 12–24 hours. Despite this complex edema pattern on the uninjured side with associated pathologic changes, no significant change in apparent diffusion coefficient values was detected over the first 24 hours. Apparent diffusion coefficient values accurately detected the changes on the injured side, but did not detect the changes on the uninjured side, giving a false-negative result.
Collapse
Affiliation(s)
- Hong Lu
- Department of Radiology, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, Hainan Province, China
| | - Xiaoyan Lei
- Department of Radiology, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, Hainan Province, China
| |
Collapse
|
31
|
Turtzo LC, Lescher J, Janes L, Dean DD, Budde MD, Frank JA. Macrophagic and microglial responses after focal traumatic brain injury in the female rat. J Neuroinflammation 2014; 11:82. [PMID: 24761998 PMCID: PMC4022366 DOI: 10.1186/1742-2094-11-82] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/06/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND After central nervous system injury, inflammatory macrophages (M1) predominate over anti-inflammatory macrophages (M2). The temporal profile of M1/M2 phenotypes in macrophages and microglia after traumatic brain injury (TBI) in rats is unknown. We subjected female rats to severe controlled cortical impact (CCI) and examined the postinjury M1/M2 time course in their brains. METHODS The motor cortex (2.5 mm left laterally and 1.0 mm anteriorly from the bregma) of anesthetized female Wistar rats (ages 8 to 10 weeks; N = 72) underwent histologically moderate to severe CCI with a 5-mm impactor tip. Separate cohorts of rats had their brains dissociated into cells for flow cytometry, perfusion-fixed for immunohistochemistry (IHC) and ex vivo magnetic resonance imaging or flash-frozen for RNA and protein analysis. For each analytical method used, separate postinjury times were included for 24 hours; 3 or 5 days; or 1, 2, 4 or 8 weeks. RESULTS By IHC, we found that the macrophagic and microglial responses peaked at 5 to 7 days post-TBI with characteristics of mixed populations of M1 and M2 phenotypes. Upon flow cytometry examination of immunological cells isolated from brain tissue, we observed that peak M2-associated staining occurred at 5 days post-TBI. Chemokine analysis by multiplex assay showed statistically significant increases in macrophage inflammatory protein 1α and keratinocyte chemoattractant/growth-related oncogene on the ipsilateral side within the first 24 hours after injury relative to controls and to the contralateral side. Quantitative RT-PCR analysis demonstrated expression of both M1- and M2-associated markers, which peaked at 5 days post-TBI. CONCLUSIONS The responses of macrophagic and microglial cells to histologically severe CCI in the female rat are maximal between days 3 and 7 postinjury. The response to injury is a mixture of M1 and M2 phenotypes.
Collapse
Affiliation(s)
- L Christine Turtzo
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Gold EM, Su D, López-Velázquez L, Haus DL, Perez H, Lacuesta GA, Anderson AJ, Cummings BJ. Functional assessment of long-term deficits in rodent models of traumatic brain injury. Regen Med 2014; 8:483-516. [PMID: 23826701 DOI: 10.2217/rme.13.41] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) ranks as the leading cause of mortality and disability in the young population worldwide. The annual US incidence of TBI in the general population is estimated at 1.7 million per year, with an estimated financial burden in excess of US$75 billion a year in the USA alone. Despite the prevalence and cost of TBI to individuals and society, no treatments have passed clinical trial to clinical implementation. The rapid expansion of stem cell research and technology offers an alternative to traditional pharmacological approaches targeting acute neuroprotection. However, preclinical testing of these approaches depends on the selection and characterization of appropriate animal models. In this article we consider the underlying pathophysiology for the focal and diffuse TBI subtypes, discuss the existing preclinical TBI models and functional outcome tasks used for assessment of injury and recovery, identify criteria particular to preclinical animal models of TBI in which stem cell therapies can be tested for safety and efficacy, and review these criteria in the context of the existing TBI literature. We suggest that 2 months post-TBI is the minimum period needed to evaluate human cell transplant efficacy and safety. Comprehensive review of the published TBI literature revealed that only 32% of rodent TBI papers evaluated functional outcome ≥1 month post-TBI, and only 10% evaluated functional outcomes ≥2 months post-TBI. Not all published papers that evaluated functional deficits at a minimum of 2 months post-TBI reported deficits; hence, only 8.6% of overall TBI papers captured in this review demonstrated functional deficits at 2 months or more postinjury. A 2-month survival and assessment period would allow sufficient time for differentiation and integration of human neural stem cells with the host. Critically, while trophic effects might be observed at earlier time points, it will also be important to demonstrate the sustainability of such an effect, supporting the importance of an extended period of in vivo observation. Furthermore, regulatory bodies will likely require at least 6 months survival post-transplantation for assessment of toxicology/safety, particularly in the context of assessing cell abnormalities.
Collapse
Affiliation(s)
- Eric M Gold
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine 2030 Gross Hall, CA 92697-1705, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Traumatic brain injury (TBI) affects all age groups in a population and is an injury generating scientific interest not only as an acute event, but also as a complex brain disease with several underlying neurobehavioral and neuropathological characteristics. We review early and long-term alterations after juvenile and adult TBI with a focus on changes in the neurovascular unit (NVU), including neuronal interactions with glia and blood vessels at the blood-brain barrier (BBB). Post-traumatic changes in cerebral blood-flow, BBB structures and function, as well as mechanistic pathways associated with brain aging and neurodegeneration are presented from clinical and experimental reports. Based on the literature, increased attention on BBB changes should be integrated in studies characterizing TBI outcome and may provide a meaningful therapeutic target to resolve detrimental post-traumatic dysfunction.
Collapse
Affiliation(s)
- V Pop
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354 USA
| | | |
Collapse
|
34
|
Badaut J, Bix GJ. Vascular neural network phenotypic transformation after traumatic injury: potential role in long-term sequelae. Transl Stroke Res 2013; 5:394-406. [PMID: 24323723 DOI: 10.1007/s12975-013-0304-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 01/11/2023]
Abstract
The classical neurovascular unit (NVU), composed primarily of endothelium, astrocytes, and neurons, could be expanded to include smooth muscle and perivascular nerves present in both the up- and downstream feeding blood vessels (arteries and veins). The extended NVU, which can be defined as the vascular neural network (VNN), may represent a new physiological unit to consider for therapeutic development in stroke, traumatic brain injury, and other brain disorders (Zhang et al., Nat Rev Neurol 8(12):711-716, 2012). This review is focused on traumatic brain injury and resultant post-traumatic changes in cerebral blood flow, smooth muscle cells, matrix, blood-brain barrier structures and function, and the association of these changes with cognitive outcomes as described in clinical and experimental reports. We suggest that studies characterizing TBI outcomes should increase their focus on changes to the VNN, as this may yield meaningful therapeutic targets to resolve posttraumatic dysfunction.
Collapse
Affiliation(s)
- J Badaut
- Department of Pediatrics, Loma Linda University School of Medicine, Coleman Pavilion, Room A1120, 11175 Campus Street, Loma Linda, CA, 92354, USA,
| | | |
Collapse
|
35
|
Shen X, Li A, Zhang Y, Dong X, Shan T, Wu Y, Jia J, Hu Y. The effect of different intensities of treadmill exercise on cognitive function deficit following a severe controlled cortical impact in rats. Int J Mol Sci 2013; 14:21598-612. [PMID: 24185909 PMCID: PMC3856023 DOI: 10.3390/ijms141121598] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/12/2013] [Accepted: 10/17/2013] [Indexed: 11/16/2022] Open
Abstract
Exercise has been proposed for the treatment of traumatic brain injury (TBI). However, the proper intensity of exercise in the early phase following a severe TBI is largely unknown. To compare two different treadmill exercise intensities on the cognitive function following a severe TBI in its early phase, rats experienced a controlled cortical impact (CCI) and were forced to treadmill exercise for 14 days. The results revealed that the rats in the low intensity exercise group had a shorter latency to locate a platform and a significantly better improvement in spatial memory in the Morris water maze (MWM) compared to the control group (p < 0.05). The high intensity exercise group showed a longer latency and a mild improvement in spatial memory compared to the control group rats in the MWM; however, this difference was not statistically significant (p > 0.05). The brain-derived neurotrophic factor (BDNF) and p-CREB protein levels in the contralateral hippocampus were increased significantly in the low intensity exercise group. Our results suggest that 2 weeks of low intensity of treadmill exercise is beneficial for improving cognitive function and increasing hippocampal BDNF expression after a severe TBI in its early phase.
Collapse
Affiliation(s)
- Xiafeng Shen
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
- Department of Rehabilitation, Shanghai Yangpu District Geriatric Hospital, Shanghai 200090, China
| | - Aiping Li
- Rehabilitation Medicine Center, Nanjing Military Region Sanatorium of Hangzhou, Hangzhou 310007, Zhejiang, China; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-571-8734-8142; Fax: +86-571-8734-8114
| | - Yuling Zhang
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| | - XiaoMin Dong
- Rehabilitation Medicine Center, Nanjing Military Region Sanatorium of Hangzhou, Hangzhou 310007, Zhejiang, China; E-Mail:
| | - Tian Shan
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| | - Yi Wu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| | - Jie Jia
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| | - Yongshan Hu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| |
Collapse
|
36
|
Moro N, Ghavim S, Harris NG, Hovda DA, Sutton RL. Glucose administration after traumatic brain injury improves cerebral metabolism and reduces secondary neuronal injury. Brain Res 2013; 1535:124-36. [PMID: 23994447 DOI: 10.1016/j.brainres.2013.08.044] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 08/21/2013] [Accepted: 08/22/2013] [Indexed: 01/08/2023]
Abstract
Clinical studies have indicated an association between acute hyperglycemia and poor outcomes in patients with traumatic brain injury (TBI), although optimal blood glucose levels needed to maximize outcomes for these patients' remain under investigation. Previous results from experimental animal models suggest that post-TBI hyperglycemia may be harmful, neutral, or beneficial. The current studies determined the effects of single or multiple episodes of acute hyperglycemia on cerebral glucose metabolism and neuronal injury in a rodent model of unilateral controlled cortical impact (CCI) injury. In Experiment 1, a single episode of hyperglycemia (50% glucose at 2 g/kg, i.p.) initiated immediately after CCI was found to significantly attenuate a TBI-induced depression of glucose metabolism in cerebral cortex (4 of 6 regions) and subcortical regions (2 of 7) as well as to significantly reduce the number of dead/dying neurons in cortex and hippocampus at 24 h post-CCI. Experiment 2 examined effects of more prolonged and intermittent hyperglycemia induced by glucose administrations (2 g/kg, i.p.) at 0, 1, 3 and 6h post-CCI. The latter study also found significantly improved cerebral metabolism (in 3 of 6 cortical and 3 of 7 subcortical regions) and significant neuroprotection in cortex and hippocampus 1 day after CCI and glucose administration. These results indicate that acute episodes of post-TBI hyperglycemia can be beneficial and are consistent with other recent studies showing benefits of providing exogenous energy substrates during periods of increased cerebral metabolic demand.
Collapse
Affiliation(s)
- Nobuhiro Moro
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-7039, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, Box 957039, Los Angeles, CA 90095-7039, USA.
| | | | | | | | | |
Collapse
|
37
|
Boulet T, Kelso ML, Othman SF. Long-term in vivo imaging of viscoelastic properties of the mouse brain after controlled cortical impact. J Neurotrauma 2013; 30:1512-20. [PMID: 23534701 DOI: 10.1089/neu.2012.2788] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) presents a variety of causes and symptoms, thus making the development of reliable diagnostic methods and therapeutic treatments challenging. Magnetic resonance elastography (MRE) is a technique that allows for a noninvasive assessment of the mechanical properties of soft biological tissue, such as tissue stiffness, storage modulus, and loss modulus. Importantly, by quantifying the changes in the stiffness of tissue that is often associated with disease, MRE is able to detect tissue pathologies at early stages. Recent improvements in instrumentation have allowed for the investigation of small samples with microscopic resolution (μMRE). We hypothesize that μMRE can sensitively detect variations in micromechanical properties in the brain caused by the compressive and shearing forces sustained during TBI. To test this hypothesis, we randomized 13 C57BL mice to receive a controlled cortical impact at a 0.5 mm or 0.75 mm depth, with both sham and naïve mice as controls. Our objective was to propagate mechanical shear waves throughout the brain for in vivo TBI μMRE imaging. The mechanical properties of the injured brain tissue were determined at days 0, 1, 7, and 28 post-injury. For both groups, we observed a significant drop in the stiffness of the impacted region immediately following the injury; the 0.75 mm animals experienced increased tissue softness that lasted longer than that for the 0.5 mm group. Although the shear stiffness, storage modulus, and loss modulus parameters all followed the same trend, the tissue stiffness yielded the most statistically significant results. Overall, this article introduces a transformative technique for mechanically mapping the brain and detecting brain diseases and injury.
Collapse
Affiliation(s)
- Thomas Boulet
- Department of Engineering Mechanics, University of Nebraska-Lincoln, Lincoln, Nebraksa 43353, USA
| | | | | |
Collapse
|
38
|
Sword J, Masuda T, Croom D, Kirov SA. Evolution of neuronal and astroglial disruption in the peri-contusional cortex of mice revealed by in vivo two-photon imaging. Brain 2013; 136:1446-61. [PMID: 23466395 PMCID: PMC3634194 DOI: 10.1093/brain/awt026] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 12/08/2012] [Accepted: 12/27/2012] [Indexed: 12/14/2022] Open
Abstract
In traumatic brain injury mechanical forces applied to the cranium and brain cause irreversible primary neuronal and astroglial damage associated with terminal dendritic beading and spine loss representing acute damage to synaptic circuitry. Oedema develops quickly after trauma, raising intracranial pressure that results in a decrease of blood flow and consequently in cerebral ischaemia, which can cause secondary injury in the peri-contusional cortex. Spreading depolarizations have also been shown to occur after traumatic brain injury in humans and in animal models and are thought to accelerate and exacerbate secondary tissue injury in at-risk cortical territory. Yet, the mechanisms of acute secondary injury to fine synaptic circuitry within the peri-contusional cortex after mild traumatic brain injury remain unknown. A mild focal cortical contusion model in adult mouse sensory-motor cortex was implemented by the controlled cortical impact injury device. In vivo two-photon microscopy in the peri-contusional cortex was used to monitor via optical window yellow fluorescent protein expressing neurons, enhanced green fluorescent protein expressing astrocytes and capillary blood flow. Dendritic beading in the peri-contusional cortex developed slowly and the loss of capillary blood flow preceded terminal dendritic injury. Astrocytes were swollen indicating oedema and remained swollen during the next 24 h throughout the imaging session. There were no recurrent spontaneous spreading depolarizations in this mild traumatic brain injury model; however, when spreading depolarizations were repeatedly induced outside the peri-contusional cortex by pressure-injecting KCl, dendrites undergo rapid beading and recovery coinciding with passage of spreading depolarizations, as was confirmed with electrophysiological recordings in the vicinity of imaged dendrites. Yet, accumulating metabolic stress resulting from as few as four rounds of spreading depolarization significantly added to the fraction of beaded dendrites that were incapable to recover during repolarization, thus facilitating terminal injury. In contrast, similarly induced four rounds of spreading depolarization in another set of control healthy mice caused no accumulating dendritic injury as dendrites fully recovered from beading during repolarization. Taken together, our data suggest that in the mild traumatic brain injury the acute dendritic injury in the peri-contusional cortex is gated by the decline in the local blood flow, most probably as a result of developing oedema. Furthermore, spreading depolarization is a specific mechanism that could accelerate injury to synaptic circuitry in the metabolically compromised peri-contusional cortex, worsening secondary damage following traumatic brain injury.
Collapse
Affiliation(s)
- Jeremy Sword
- 1 Graduate Program in Neuroscience, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | - Tadashi Masuda
- 2 Brain and Behaviour Discovery Institute, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | - Deborah Croom
- 3 Department of Neurosurgery, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | - Sergei A. Kirov
- 2 Brain and Behaviour Discovery Institute, Georgia Health Sciences University, Augusta, Georgia 30912, USA
- 3 Department of Neurosurgery, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| |
Collapse
|
39
|
Turtzo LC, Budde MD, Gold EM, Lewis BK, Janes L, Yarnell A, Grunberg NE, Watson W, Frank JA. The evolution of traumatic brain injury in a rat focal contusion model. NMR IN BIOMEDICINE 2013; 26:468-479. [PMID: 23225324 PMCID: PMC3596464 DOI: 10.1002/nbm.2886] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 08/28/2012] [Accepted: 10/05/2012] [Indexed: 06/01/2023]
Abstract
Serial MRI facilitates the in vivo analysis of the intra- and intersubject evolution of traumatic brain injury lesions. Despite the availability of MRI, the natural history of experimental focal contusion lesions in the controlled cortical impact (CCI) rat model has not been well described. We performed CCI on rats and MRI during the acute to chronic stages of cerebral injury to investigate the time course of changes in the brain. Female Wistar rats underwent CCI of their left motor cortex with a flat impact tip driven by an electromagnetic piston. In vivo MRI was performed at 7 T serially over 6 weeks post-CCI. The appearances of CCI-induced lesions and lesion-associated cortical volumes were variable on MRI, with the percentage change in cortical volume of the CCI ipsilateral side relative to the contralateral side ranging from 18% within 2 h of injury on day 0 to a peak of 35% on day 1, and a trough of -28% by week 5/6, with an average standard deviation of ± 14% at any given time point. In contrast, the percentage change in cortical volume of the ipsilateral side relative to the contralateral side in control rats was not significant (1 ± 2%). Hemorrhagic conversion within and surrounding the CCI lesion occurred between days 2 and 9 in 45% of rats, with no hemorrhage noted on the initial scan. Furthermore, hemorrhage and hemosiderin within the lesion were positive for Prussian blue and highly autofluorescent on histological examination. Although some variation in injuries may be technique related, the divergence of similar lesions between initial and final scans demonstrates the inherent biological variability of the CCI rat model.
Collapse
Affiliation(s)
- L. Christine Turtzo
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Matthew D. Budde
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Eric M. Gold
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Bobbi K. Lewis
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay Janes
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Angela Yarnell
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Neil E. Grunberg
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - William Watson
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Joseph A. Frank
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
40
|
Alessandri B, Schwandt E, Kamada Y, Nagata M, Heimann A, Kempski O. The neuroprotective effect of lactate is not due to improved glutamate uptake after controlled cortical impact in rats. J Neurotrauma 2012; 29:2181-91. [PMID: 22888957 DOI: 10.1089/neu.2011.2067] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
For many years lactate was considered to be a waste product of glycolysis. Data are accumulating that suggest that lactate is an important energy substrate for neurons during activation. In severe traumatic brain injury (TBI) glutamate release and ischemic cerebral blood flow (CBF) are major factors for a mismatch between energy demand and supply and for neuronal cell death. Although ATP and behavior could be improved by lactate treatment after TBI, no histological correlate nor any linkage to better astrocytic glutamate uptake or CBF as possible mechanisms have been described. We subjected male rats to a controlled cortical impact (CCI; 5 m/sec, 2.5 mm). To study the effects of lactate treatment on lesion volume, glutamate release, and CBF, animals were infused with either NaCl or 100 mM lactate for up to 3 h. The role of endogenous lactate was investigated by inhibiting transport with α-cyano-4-hydroxy-cinnamic acid (4-CIN; 90 mg/kg). Lactate treatment 15 min post-CCI reduced lesion volume from 21.1±2.8 mm³ to 12.1±1.9 mm³ at day 2 after CCI. Contusion produced a significant three- to fourfold increase of glutamate in microdialysates, but there was no significant difference between treatments that began 30 min before CCI. In this experiment lesion volume was significantly reduced by lactate at day 7 post-CCI (23.7±4 to 9.3±1-2 mm³). CBF increased immediately after CCI and dropped thereafter below baseline in all animals. Lactate infusion 15 min post-CCI elevated CBF for 20 min in 7 of 10 animals, whereas 7 of 8 NaCl-treated animals showed a further CBF decline. Neuroprotection was achieved by lactate treatment following contusion injury, whereas blocking of endogenous lactate transport exerted no adverse effects. Neuroprotection was not achieved by improved glutamate uptake into astrocytes, but was supported by augmented CBF following CCI. Due to its neuroprotective property, lactate might be a beneficial pharmacological treatment for TBI patients.
Collapse
Affiliation(s)
- Beat Alessandri
- Institute for Neurosurgical Pathophysiology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany.
| | | | | | | | | | | |
Collapse
|
41
|
Harris NG, Mironova YA, Chen SF, Richards HK, Pickard JD. Preventing flow-metabolism uncoupling acutely reduces axonal injury after traumatic brain injury. J Neurotrauma 2012; 29:1469-82. [PMID: 22321027 PMCID: PMC3335110 DOI: 10.1089/neu.2011.2161] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We have previously presented evidence that the development of secondary traumatic axonal injury is related to the degree of local cerebral blood flow (LCBF) and flow-metabolism uncoupling. We have now tested the hypothesis that augmenting LCBF in the acute stages after brain injury prevents further axonal injury. Data were acquired from rats with or without acetazolamide (ACZ) that was administered immediately following controlled cortical impact injury to increase cortical LCBF. Local cerebral metabolic rate for glucose (LCMRglc) and LCBF measurements were obtained 3 h post-trauma in the same rat via ¹⁸F-fluorodeoxyglucose and ¹⁴C-iodoantipyrine co-registered autoradiographic images, and compared to the density of damaged axonal profiles in adjacent sections, and in additional groups at 24 h used to assess different populations of injured axons stereologically. ACZ treatment significantly and globally elevated LCBF twofold above untreated-injured rats at 3 h (p<0.05), but did not significantly affect LCMRglc. As a result, ipsilateral LCMRglc:LCBF ratios were reduced by twofold to sham-control levels, and the density of β-APP-stained axons at 24 h was significantly reduced in most brain regions compared to the untreated-injured group (p<0.01). Furthermore, early LCBF augmentation prevented the injury-associated increase in the number of stained axons from 3-24 h. Additional robust stereological analysis of impaired axonal transport and neurofilament compaction in the corpus callosum and cingulum underlying the injury core confirmed the amelioration of β-APP axon density, and showed a trend, but no significant effect, on RMO14-positive axons. These data underline the importance of maintaining flow-metabolism coupling immediately after injury in order to prevent further axonal injury, in at least one population of injured axons.
Collapse
Affiliation(s)
- Neil G Harris
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Box 957039, Los Angeles, CA 90095-7039, USA.
| | | | | | | | | |
Collapse
|
42
|
Schoch KM, Evans HN, Brelsfoard JM, Madathil SK, Takano J, Saido TC, Saatman KE. Calpastatin overexpression limits calpain-mediated proteolysis and behavioral deficits following traumatic brain injury. Exp Neurol 2012; 236:371-82. [PMID: 22572592 DOI: 10.1016/j.expneurol.2012.04.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 03/23/2012] [Accepted: 04/23/2012] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) results in abrupt, initial cell damage leading to delayed neuronal death. The calcium-activated proteases, calpains, are known to contribute to this secondary neurodegenerative cascade. Although the specific inhibitor of calpains, calpastatin, is present within neurons, normal levels of calpastatin are unable to fully prevent the damaging proteolytic activity of calpains after injury. In this study, increased calpastatin expression was achieved using transgenic mice that overexpress the human calpastatin (hCAST) construct under control of a calcium-calmodulin-dependent kinase II α promoter. Naïve hCAST transgenic mice exhibited enhanced neuronal calpastatin expression and significantly reduced protease activity. Acute calpain-mediated spectrin proteolysis in the cortex and hippocampus induced by controlled cortical impact brain injury was significantly attenuated in calpastatin overexpressing mice. Aspects of posttraumatic motor and cognitive behavioral deficits were also lessened in hCAST transgenic mice compared to their wildtype littermates. However, volumetric analyses of neocortical contusion revealed no histological neuroprotection at either acute or long-term time points. Partial hippocampal neuroprotection observed at a moderate injury severity was lost after severe TBI. This study underscores the effectiveness of calpastatin overexpression in reducing calpain-mediated proteolysis and behavioral impairment after TBI, supporting the therapeutic potential for calpain inhibition. In addition, the reduction in spectrin proteolysis without accompanied neocortical neuroprotection suggests the involvement of other factors that are critical for neuronal survival after contusion brain injury.
Collapse
Affiliation(s)
- Kathleen M Schoch
- Spinal Cord and Brain Injury Research Center, University of Kentucky, B416 Biomedical and Biological Sciences Research Building, 741 South Limestone Street, Lexington, KY 40536-0509, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Kenne E, Erlandsson A, Lindbom L, Hillered L, Clausen F. Neutrophil depletion reduces edema formation and tissue loss following traumatic brain injury in mice. J Neuroinflammation 2012; 9:17. [PMID: 22269349 PMCID: PMC3292978 DOI: 10.1186/1742-2094-9-17] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 01/23/2012] [Indexed: 12/24/2022] Open
Abstract
Background Brain edema as a result of secondary injury following traumatic brain injury (TBI) is a major clinical concern. Neutrophils are known to cause increased vascular permeability leading to edema formation in peripheral tissue, but their role in the pathology following TBI remains unclear. Methods In this study we used controlled cortical impact (CCI) as a model for TBI and investigated the role of neutrophils in the response to injury. The outcome of mice that were depleted of neutrophils using an anti-Gr-1 antibody was compared to that in mice with intact neutrophil count. The effect of neutrophil depletion on blood-brain barrier function was assessed by Evan's blue dye extravasation, and analysis of brain water content was used as a measurement of brain edema formation (24 and 48 hours after CCI). Lesion volume was measured 7 and 14 days after CCI. Immunohistochemistry was used to assess cell death, using a marker for cleaved caspase-3 at 24 hours after injury, and microglial/macrophage activation 7 days after CCI. Data were analyzed using Mann-Whitney test for non-parametric data. Results Neutrophil depletion did not significantly affect Evan's blue extravasation at any time-point after CCI. However, neutrophil-depleted mice exhibited a decreased water content both at 24 and 48 hours after CCI indicating reduced edema formation. Furthermore, brain tissue loss was attenuated in neutropenic mice at 7 and 14 days after injury. Additionally, these mice had a significantly reduced number of activated microglia/macrophages 7 days after CCI, and of cleaved caspase-3 positive cells 24 h after injury. Conclusion Our results suggest that neutrophils are involved in the edema formation, but not the extravasation of large proteins, as well as contributing to cell death and tissue loss following TBI in mice.
Collapse
Affiliation(s)
- Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
44
|
Ohayon S, Boyko M, Saad A, Douvdevani A, Gruenbaum BF, Melamed I, Shapira Y, Teichberg VI, Zlotnik A. Cell-Free DNA as a Marker for Prediction of Brain Damage in Traumatic Brain Injury in Rats. J Neurotrauma 2012; 29:261-7. [DOI: 10.1089/neu.2011.1938] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Affiliation(s)
- Sharon Ohayon
- Division of Anesthesiology and Critical Care, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Matthew Boyko
- Division of Anesthesiology and Critical Care, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Amit Saad
- Department of Philosophy, Haifa University, Haifa, Israel
| | - Amos Douvdevani
- Department of Clinical Biochemistry, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Benjamin F. Gruenbaum
- Division of Anesthesiology and Critical Care, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Israel Melamed
- Department of Neurosurgery, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yoram Shapira
- Division of Anesthesiology and Critical Care, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Alexander Zlotnik
- Division of Anesthesiology and Critical Care, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
45
|
Jeter CB, Hergenroeder GW, Ward NH, Moore AN, Dash PK. Human Traumatic Brain Injury Alters Circulating L-Arginine and Its Metabolite Levels: Possible Link to Cerebral Blood Flow, Extracellular Matrix Remodeling, and Energy Status. J Neurotrauma 2012; 29:119-27. [DOI: 10.1089/neu.2011.2029] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Cameron B. Jeter
- Department of Neurobiology & Anatomy, The Vivian L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, Texas
| | - Georgene W. Hergenroeder
- Department of Neurobiology & Anatomy, The Vivian L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, Texas
| | - Norman H. Ward
- Department of Neurobiology & Anatomy, The Vivian L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, Texas
| | - Anthony N. Moore
- Department of Neurobiology & Anatomy, The Vivian L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, Texas
| | - Pramod K. Dash
- Department of Neurobiology & Anatomy, The Vivian L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, Texas
| |
Collapse
|
46
|
Dijkhuizen RM. Advances in MRI-Based Detection of Cerebrovascular Changes after Experimental Traumatic Brain Injury. Transl Stroke Res 2011; 2:524-32. [PMID: 22207884 PMCID: PMC3236292 DOI: 10.1007/s12975-011-0130-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 10/25/2011] [Accepted: 10/26/2011] [Indexed: 01/04/2023]
Abstract
Traumatic brain injury is a heterogeneous and multifaceted neurological disorder that involves diverse pathophysiological pathways and mechanisms. Thorough characterization and monitoring of the brain’s status after neurotrauma is therefore highly complicated. Magnetic resonance imaging (MRI) provides a versatile tool for in vivo spatiotemporal assessment of various aspects of central nervous system injury, such as edema formation, perfusion disturbances and structural tissue damage. Moreover, recent advances in MRI methods that make use of contrast agents have opened up additional opportunities for measurement of events at the level of the cerebrovasculature, such as blood–brain barrier permeability, leukocyte infiltration, cell adhesion molecule upregulation and vascular remodeling. It is becoming increasingly clear that these cerebrovascular alterations play a significant role in the progression of post-traumatic brain injury as well as in the process of post-traumatic brain repair. Application of advanced multiparametric MRI strategies in experimental, preclinical studies may significantly aid in the elucidation of pathomechanisms, monitoring of treatment effects, and identification of predictive markers after traumatic brain injury.
Collapse
Affiliation(s)
- Rick M. Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, Building Nieuw Gildestein, Yalelaan 2, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
47
|
Viano DC, Hamberger A, Bolouri H, Säljö A. Evaluation of three animal models for concussion and serious brain injury. Ann Biomed Eng 2011; 40:213-26. [PMID: 22012080 DOI: 10.1007/s10439-011-0386-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 08/22/2011] [Indexed: 10/16/2022]
Abstract
Three animal models were evaluated in this study involving head impacts of the rat, including the Marmarou drop-weight and two momentum-exchange techniques. In series 1, 36 Wistar rats were hit on the side of the free-moving head using Marmarou's 450 g impact mass at 4.4, 5.4, and 6.3 m/s. Head acceleration was measured and injuries were observed. The 6.3-m/s side impact resulted in no deaths, no skull fractures, infrequent contusions, and some injuries consistent with diffuse axonal injury. In series 2, 57 Marmarou drop-weight tests were conducted to study head biomechanical responses. Marmarou's technique involves a head impact followed by prolonged loading into a foam pad under the animal. Based on the literature, the 2 m (6.3 m/s) Marmarou drop causes death, skull fracture, brain and spinal cord contusions, and diffuse axonal injury. These injuries are more severe than that occurring with impact of similar mass and velocity to the free-moving head. Impacts to the free-moving head provide more realistic animal models to study concussion and severe brain injury.
Collapse
Affiliation(s)
- David C Viano
- ProBiomechanics LLC, 265 Warrington Rd., Bloomfield Hills, MI 48304, USA.
| | | | | | | |
Collapse
|
48
|
Boulet T, Kelso ML, Othman SF. Microscopic magnetic resonance elastography of traumatic brain injury model. J Neurosci Methods 2011; 201:296-306. [PMID: 21871490 DOI: 10.1016/j.jneumeth.2011.08.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Revised: 08/01/2011] [Accepted: 08/01/2011] [Indexed: 01/11/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability for which there is no cure. One of the issues inhibiting clinical trial success is the lack of targeting specific patient populations due to inconsistencies between clinical diagnostic tools and underlying pathophysiology. The development of reliable, noninvasive markers of TBI severity and injury mechanisms may better identify these populations, thereby improving clinical trial design. Magnetic resonance elastography (MRE), by assessing tissue mechanical properties, can potentially provide such marker. MRE synchronizes mechanical excitations with a phase contrast imaging pulse sequence to noninvasively register shear wave propagation, from which local values of tissue viscoelastic properties can be deduced. The working hypothesis of this study is that TBI involves a compression of brain tissue large enough to bring the material out of its elastic range, sufficiently altering mechanical properties to generate contrast on MRE measurements. To test this hypothesis, we combined microscopic MRE with brain tissue collected from adult male rats subjected to a controlled cortical impact injury. Measurements were made in different regions of interest (somatosensory cortex, hippocampus, and thalamus), and at different time points following the injury (immediate, 24 h, 7 days, 28 days). Values of stiffness in the somatosensory cortex were found to be 23-32% lower in the injured hemisphere than in the healthy one, when no significant difference was observed in the case of sham brains. A preliminary in vivo experiment is also presented, as well as alternatives to improve the faithfulness of stiffness recovery.
Collapse
Affiliation(s)
- Thomas Boulet
- Department of Engineering Mechanics, W303 Nebraska Hall, University of Nebraska-Lincoln, Lincoln, NE 68588, USA.
| | | | | |
Collapse
|
49
|
|
50
|
Mao H, Jin X, Zhang L, Yang KH, Igarashi T, Noble-Haeusslein LJ, King AI. Finite element analysis of controlled cortical impact-induced cell loss. J Neurotrauma 2010; 27:877-88. [PMID: 20199194 DOI: 10.1089/neu.2008.0616] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The controlled cortical impact (CCI) model has been extensively used to study region-specific patterns of neuronal injury and cell death after a focal traumatic brain injury. Although external parameters such as impact velocity and depth of penetration have been defined in this injury model, little is known about the intracranial mechanical responses within cortical and subcortical brain regions where neuronal loss is prevalent. At present, one of the best methods to determine the internal responses of the brain is finite element (FE) modeling. A previously developed and biomechanically validated detailed three-dimensional FE rat brain model, consisting of 255,700 hexahedral elements and representing all essential anatomical features of a rat brain, was used to study intracranial responses in a series of CCI experiments in which injury severity ranged from mild to severe. A linear relationship was found between the percentage of the neuronal loss observed in vivo and the FE model-predicted maximum principal strain (R(2) = 0.602). Interestingly, the FE model also predicted some risk of injury in the cerebellum, located remote from the point of impact, with a 25% neuronal loss for the "severe" impact condition. More research is needed to examine other regions that do not have histological data for comparison with FE model predictions before this injury mechanism and the associated injury threshold can be fully established.
Collapse
Affiliation(s)
- Haojie Mao
- Bioengineering Center, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | |
Collapse
|