1
|
Nanda U, Zhang G, Underhill D, Pangarkar S. Management of Pain and Headache After Traumatic Brain Injury. Phys Med Rehabil Clin N Am 2024; 35:573-591. [PMID: 38945652 DOI: 10.1016/j.pmr.2024.02.009] [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] [Indexed: 07/02/2024]
Abstract
This article will identify common causes of pain following traumatic brain injury (TBI), discuss current treatment strategies for these complaints, and help tailor treatments for both acute and chronic settings. We will also briefly discuss primary and secondary headache disorders, followed by common secondary pain disorders that may be related to trauma.
Collapse
Affiliation(s)
- Udai Nanda
- Department of Physical Medicine and Rehabilitation, Pain Management, Headache Center of Excellence, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA; Division of Physical Medicine and Rehabilitation, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Grace Zhang
- Division of Physical Medicine and Rehabilitation, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - David Underhill
- Division of Physical Medicine and Rehabilitation, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Sanjog Pangarkar
- Division of Physical Medicine and Rehabilitation, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA; Department of Physical Medicine and Rehabilitation, Pain Management, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
2
|
Varghese N, Morrison B. Partial Depletion of Microglia Attenuates Long-Term Potentiation Deficits following Repeated Blast Traumatic Brain Injury in Organotypic Hippocampal Slice Cultures. J Neurotrauma 2023; 40:547-560. [PMID: 36508265 PMCID: PMC10081725 DOI: 10.1089/neu.2022.0284] [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] [Indexed: 12/14/2022] Open
Abstract
Blast-induced traumatic brain injury (bTBI) has been a health concern in both military and civilian populations due to recent military and geopolitical conflicts. Military service members are frequently exposed to repeated bTBI throughout their training and deployment. Our group has previously reported compounding functional deficits as a result of increased number of blast exposures. In this study, we further characterized the decrease in long-term potentiation (LTP) by varying the blast injury severity and the inter-blast interval between two blast exposures. LTP deficits were attenuated with increasing inter-blast intervals. We also investigated changes in microglial activation; expression of CD68 was increased and expression of CD206 was decreased after multiple blast exposures. Expression of macrophage inflammatory protein (MIP)-1α, interleukin (IL)-1β, monocyte chemoattractant protein (MCP)-1, interferon gamma-inducible protein (IP)-10, and regulated on activation, normal T cell expressed and secreted (RANTES) increased, while expression of IL-10 decreased in the acute period after both single and repeated bTBI. By partially depleting microglia prior to injury, LTP deficits after injury were significantly reduced. Treatment with the novel drug, MW-189, prevented LTP deficits when administered immediately following a repeated bTBI and even when administered only for an acute period (24 h) between two blast injuries. These findings could inform the development of therapeutic strategies to treat the neurological deficits of repeated bTBI suggesting that microglia play a major role in functional neuronal deficits and may be a viable therapeutic target to lessen the neurophysiological deficits after bTBI.
Collapse
Affiliation(s)
- Nevin Varghese
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| |
Collapse
|
3
|
Varghese N, Morrison B. Inhibition of cyclooxygenase and EP3 receptor improved long term potentiation in a rat organotypic hippocampal model of repeated blast traumatic brain injury. Neurochem Int 2023; 163:105472. [PMID: 36599378 DOI: 10.1016/j.neuint.2022.105472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/09/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Blast-induced traumatic brain injury (bTBI) is a health concern in military service members who are exposed to multiple blasts throughout their training and deployment. Our group has previously reported decreased long term potentiation (LTP) following repeated bTBI in a rat organotypic hippocampal slice culture (OHSC) model. In this study, we investigated changes in inflammatory markers like cyclooxygenase (COX) and tested the efficacy of COX or prostaglandin EP3 receptor (EP3R) inhibitors in attenuating LTP deficits. Expression of COX-2 was increased 48 h following repeated injury, whereas COX-1 expression was unchanged. EP3R expression was upregulated, and cyclic adenosine monophosphate (cAMP) concentration was decreased after repeated blast exposure. Post-traumatic LTP deficits improved after treatment with a COX-1 specific inhibitor, SC-560, a COX-2 specific inhibitor, rofecoxib, a pan-COX inhibitor, ibuprofen, or an EP3R inhibitor, L-798,106. Delayed treatment with ibuprofen and L-798,106 also prevented LTP deficits. These findings suggest that bTBI induced neuroinflammation may be responsible for some functional deficits that we have observed in injured OHSCs. Additionally, COX and EP3R inhibition may be viable therapeutic strategies to reduce neurophysiological deficits after repeated bTBI.
Collapse
Affiliation(s)
- Nevin Varghese
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, 10027, USA.
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, 10027, USA.
| |
Collapse
|
4
|
Snapper DM, Reginauld B, Liaudanskaya V, Fitzpatrick V, Kim Y, Georgakoudi I, Kaplan DL, Symes AJ. Development of a novel bioengineered 3D brain-like tissue for studying primary blast-induced traumatic brain injury. J Neurosci Res 2023; 101:3-19. [PMID: 36200530 DOI: 10.1002/jnr.25123] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/04/2022] [Accepted: 08/29/2022] [Indexed: 11/08/2022]
Abstract
Primary blast injury is caused by the direct impact of an overpressurization wave on the body. Due to limitations of current models, we have developed a novel approach to study primary blast-induced traumatic brain injury. Specifically, we employ a bioengineered 3D brain-like human tissue culture system composed of collagen-infused silk protein donut-like hydrogels embedded with human IPSC-derived neurons, human astrocytes, and a human microglial cell line. We have utilized this system within an advanced blast simulator (ABS) to expose the 3D brain cultures to a blast wave that can be precisely controlled. These 3D cultures are enclosed in a 3D-printed surrogate skull-like material containing media which are then placed in a holder apparatus inside the ABS. This allows for exposure to the blast wave alone without any secondary injury occurring. We show that blast induces an increase in lactate dehydrogenase activity and glutamate release from the cultures, indicating cellular injury. Additionally, we observe a significant increase in axonal varicosities after blast. These varicosities can be stained with antibodies recognizing amyloid precursor protein. The presence of amyloid precursor protein deposits may indicate a blast-induced axonal transport deficit. After blast injury, we find a transient release of the known TBI biomarkers, UCHL1 and NF-H at 6 h and a delayed increase in S100B at 24 and 48 h. This in vitro model will enable us to gain a better understanding of clinically relevant pathological changes that occur following primary blast and can also be utilized for discovery and characterization of biomarkers.
Collapse
Affiliation(s)
- Dustin M Snapper
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, USA
| | - Bianca Reginauld
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, USA
| | - Volha Liaudanskaya
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Yeonho Kim
- Preclinical Behavior and Modeling Core, Uniformed Services University, Bethesda, Maryland, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Srinivasan G, Brafman DA. The Emergence of Model Systems to Investigate the Link Between Traumatic Brain Injury and Alzheimer's Disease. Front Aging Neurosci 2022; 13:813544. [PMID: 35211003 PMCID: PMC8862182 DOI: 10.3389/fnagi.2021.813544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous epidemiological studies have demonstrated that individuals who have sustained a traumatic brain injury (TBI) have an elevated risk for developing Alzheimer's disease and Alzheimer's-related dementias (AD/ADRD). Despite these connections, the underlying mechanisms by which TBI induces AD-related pathology, neuronal dysfunction, and cognitive decline have yet to be elucidated. In this review, we will discuss the various in vivo and in vitro models that are being employed to provide more definite mechanistic relationships between TBI-induced mechanical injury and AD-related phenotypes. In particular, we will highlight the strengths and weaknesses of each of these model systems as it relates to advancing the understanding of the mechanisms that lead to TBI-induced AD onset and progression as well as providing platforms to evaluate potential therapies. Finally, we will discuss how emerging methods including the use of human induced pluripotent stem cell (hiPSC)-derived cultures and genome engineering technologies can be employed to generate better models of TBI-induced AD.
Collapse
Affiliation(s)
| | - David A. Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
6
|
Direct Observation of Low Strain, High Rate Deformation of Cultured Brain Tissue During Primary Blast. Ann Biomed Eng 2019; 48:1196-1206. [DOI: 10.1007/s10439-019-02437-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/08/2019] [Indexed: 10/25/2022]
|
7
|
Bugay V, Bozdemir E, Vigil FA, Chun SH, Holstein DM, Elliott WR, Sprague CJ, Cavazos JE, Zamora DO, Rule G, Shapiro MS, Lechleiter JD, Brenner R. A Mouse Model of Repetitive Blast Traumatic Brain Injury Reveals Post-Trauma Seizures and Increased Neuronal Excitability. J Neurotrauma 2019; 37:248-261. [PMID: 31025597 DOI: 10.1089/neu.2018.6333] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Repetitive blast traumatic brain injury (TBI) affects numerous soldiers on the battlefield. Mild TBI has been shown to have long-lasting effects with repeated injury. We have investigated effects on neuronal excitability after repetitive, mild TBI in a mouse model of blast-induced brain injury. We exposed mice to mild blast trauma of an average peak overpressure of 14.6 psi, repeated across three consecutive days. While a single exposure did not reveal trauma as indicated by the glial fibrillary acidic protein indicator, three repetitive blasts did show significant increases. As well, mice had an increased indicator of inflammation (Iba-1) and increased tau, tau phosphorylation, and altered cytokine levels in the spleen. Video-electroencephalographic monitoring 48 h after the final blast exposure demonstrated seizures in 50% (12/24) of the mice, most of which were non-convulsive seizures. Long-term monitoring revealed that spontaneous seizures developed in at least 46% (6/13) of the mice. Patch clamp recording of dentate gyrus hippocampus neurons 48 h post-blast TBI demonstrated a shortened latency to the first spike and hyperpolarization of action potential threshold. We also found that evoked excitatory postsynaptic current amplitudes were significantly increased. These findings indicate that mild, repetitive blast exposures cause increases in neuronal excitability and seizures and eventual epilepsy development in some animals. The non-convulsive nature of the seizures suggests that subclinical seizures may occur in individuals experiencing even mild blast events, if repeated.
Collapse
Affiliation(s)
- Vladislav Bugay
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eda Bozdemir
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Fabio A Vigil
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Sang H Chun
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Deborah M Holstein
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - William R Elliott
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Cassie J Sprague
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | - Jose E Cavazos
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Department of Neurology, University of Texas Health San Antonio, San Antonio, Texas
| | - David O Zamora
- Sensory Trauma, United States Army Institute of Surgical Research, Fort Sam Houston San Antonio, Texas
| | | | - Mark S Shapiro
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas
| | - Robert Brenner
- Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
8
|
Simultaneous electrophysiological and morphological assessment of functional damage to neural networks in vitro after 30-300 g impacts. Sci Rep 2019; 9:14994. [PMID: 31628381 PMCID: PMC6802386 DOI: 10.1038/s41598-019-51541-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/26/2019] [Indexed: 11/08/2022] Open
Abstract
An enigma of mild traumatic brain injury are observations of substantial behavior and performance deficits in the absence of bleeding or other observable structural damage. Altered behavior and performance reflect changes in action potential (AP) patterns within neuronal networks, which could result from subtle subcellular responses that affect synaptic efficacy and AP production. The aim of this study was to investigate and quantify network activity changes after simulated concussions in vitro and therewith develop a platform for simultaneous and direct observations of morphological and electrophysiological changes in neural networks. We used spontaneously active networks grown on microelectrode arrays (MEAs) to allow long-term multisite monitoring with simultaneous optical observations before and after impacts delivered by a ballistic pendulum (30 to 300 g accelerations). The monitoring of AP waveshape templates for long periods before and after impact provided an internal control for cell death or loss of cell-electrode coupling in the observed set of neurons. Network activity patterns were linked in real-time to high power phase contrast microscopy. There was no overt loss of glial or neuronal adhesion, even at high-g impacts. All recording experiments showed repeatable spike production responses: a loss of activity with recovery to near reference in 1 hr, followed by a slow activity decay to a stable, level plateau approximately 30–40% below reference. The initial recovery occurred in two steps: a rapid return of activity to an average 24% below reference, forming a level plateau lasting from 5 to 20 min, followed by a climb to within 10% of reference where a second plateau was established for 1 to 2 hrs. Cross correlation profiles revealed changes in firing hierarchy as well as in Phase 1 in spontaneous network oscillations that were reduced by as much as 20% 6–8 min post impact with only a partial recovery at 30 min. We also observed that normally stable nuclei developed irregular rotational motion after impact in 27 out of 30 networks. The evolution of network activity deficits and recovery can be linked with microscopically observable changes in the very cells that are generating the activity. The repeatable electrophysiological impact response profiles and oscillation changes can provide a quantitative basis for systematic evaluations of pharmacological intervention strategies. Future expansion to include fluorescent microscopy should allow detailed investigations of damage mechanisms on the subcellular level.
Collapse
|
9
|
Swiatkowski P, Sewell E, Sweet ES, Dickson S, Swanson RA, McEwan SA, Cuccolo N, McDonnell ME, Patel MV, Varghese N, Morrison B, Reitz AB, Meaney DF, Firestein BL. Cypin: A novel target for traumatic brain injury. Neurobiol Dis 2018; 119:13-25. [PMID: 30031156 DOI: 10.1016/j.nbd.2018.07.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/06/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022] Open
Abstract
Cytosolic PSD-95 interactor (cypin), the primary guanine deaminase in the brain, plays key roles in shaping neuronal circuits and regulating neuronal survival. Despite this pervasive role in neuronal function, the ability for cypin activity to affect recovery from acute brain injury is unknown. A key barrier in identifying the role of cypin in neurological recovery is the absence of pharmacological tools to manipulate cypin activity in vivo. Here, we use a small molecule screen to identify two activators and one inhibitor of cypin's guanine deaminase activity. The primary screen identified compounds that change the initial rate of guanine deamination using a colorimetric assay, and secondary screens included the ability of the compounds to protect neurons from NMDA-induced injury and NMDA-induced decreases in frequency and amplitude of miniature excitatory postsynaptic currents. Hippocampal neurons pretreated with activators preserved electrophysiological function and survival after NMDA-induced injury in vitro, while pretreatment with the inhibitor did not. The effects of the activators were abolished when cypin was knocked down. Administering either cypin activator directly into the brain one hour after traumatic brain injury significantly reduced fear conditioning deficits 5 days after injury, while delivering the cypin inhibitor did not improve outcome after TBI. Together, these data demonstrate that cypin activation is a novel approach for improving outcome after TBI and may provide a new pathway for reducing the deficits associated with TBI in patients.
Collapse
Affiliation(s)
- Przemyslaw Swiatkowski
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Molecular Biosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Emily Sewell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Eric S Sweet
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Samantha Dickson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Rachel A Swanson
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Sara A McEwan
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Nicholas Cuccolo
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Mark E McDonnell
- Fox Chase Chemical Diversity Center, Inc., Doylestown, PA 18902, USA
| | - Mihir V Patel
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Nevin Varghese
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Allen B Reitz
- Fox Chase Chemical Diversity Center, Inc., Doylestown, PA 18902, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA.
| |
Collapse
|
10
|
Campos-Pires R, Koziakova M, Yonis A, Pau A, Macdonald W, Harris K, Edge CJ, Franks NP, Mahoney PF, Dickinson R. Xenon Protects against Blast-Induced Traumatic Brain Injury in an In Vitro Model. J Neurotrauma 2018; 35:1037-1044. [PMID: 29285980 PMCID: PMC5899289 DOI: 10.1089/neu.2017.5360] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The aim of this study was to evaluate the neuroprotective efficacy of the inert gas xenon as a treatment for patients with blast-induced traumatic brain injury in an in vitro laboratory model. We developed a novel blast traumatic brain injury model using C57BL/6N mouse organotypic hippocampal brain-slice cultures exposed to a single shockwave, with the resulting injury quantified using propidium iodide fluorescence. A shock tube blast generator was used to simulate open field explosive blast shockwaves, modeled by the Friedlander waveform. Exposure to blast shockwave resulted in significant (p < 0.01) injury that increased with peak-overpressure and impulse of the shockwave, and which exhibited a secondary injury development up to 72 h after trauma. Blast-induced propidium iodide fluorescence overlapped with cleaved caspase-3 immunofluorescence, indicating that shock-wave–induced cell death involves apoptosis. Xenon (50% atm) applied 1 h after blast exposure reduced injury 24 h (p < 0.01), 48 h (p < 0.05), and 72 h (p < 0.001) later, compared with untreated control injury. Xenon-treated injured slices were not significantly different from uninjured sham slices at 24 h and 72 h. We demonstrate for the first time that xenon treatment after blast traumatic brain injury reduces initial injury and prevents subsequent injury development in vitro. Our findings support the idea that xenon may be a potential first-line treatment for those with blast-induced traumatic brain injury.
Collapse
Affiliation(s)
- Rita Campos-Pires
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Mariia Koziakova
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Amina Yonis
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Ashni Pau
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Warren Macdonald
- 2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom .,3 Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Katie Harris
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Christopher J Edge
- 4 Department of Life Sciences, Imperial College London , London, United Kingdom .,5 Department of Anaesthetics, Royal Berkshire Hospital NHS Foundation Trust , Reading, United Kingdom
| | - Nicholas P Franks
- 4 Department of Life Sciences, Imperial College London , London, United Kingdom
| | - Peter F Mahoney
- 6 Royal Centre for Defence Medicine , Medical Directorate Joint Force Command, ICT Centre, Birmingham, United Kingdom
| | - Robert Dickinson
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| |
Collapse
|
11
|
Zander NE, Piehler T, Hogberg H, Pamies D. Explosive Blast Loading on Human 3D Aggregate Minibrains. Cell Mol Neurobiol 2017; 37:1331-1334. [PMID: 28110483 PMCID: PMC11482198 DOI: 10.1007/s10571-017-0463-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/09/2017] [Indexed: 11/26/2022]
Abstract
The effects of primary explosive blast on brain tissue still remain mostly unknown. There are few in vitro models that use real explosives to probe the mechanisms of injury at the cellular level. In this work, 3D aggregates of human brain cells or brain microphysiological system were exposed to military explosives at two different pressures (50 and 100 psi). Results indicate that membrane damage and oxidative stress increased with blast pressure, but cell death remained minimal.
Collapse
Affiliation(s)
- Nicole E Zander
- Department of the Army, US Army Research Laboratory, Weapons and Materials Research Directorate, RDRL-WMM-G, Building 4600, Aberdeen Proving Ground, MD, 21005, USA.
| | - Thuvan Piehler
- Department of the Army, US Army Research Laboratory, Weapons and Materials Research Directorate, RDRL-WMM-G, Building 4600, Aberdeen Proving Ground, MD, 21005, USA
| | - Helena Hogberg
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615N. Wolfe Street, Baltimore, MD, 21205, USA
| | - David Pamies
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615N. Wolfe Street, Baltimore, MD, 21205, USA
| |
Collapse
|
12
|
Sawyer TW, Ritzel DV, Wang Y, Josey T, Villanueva M, Nelson P, Song Y, Shei Y, Hennes G, Vair C, Parks S, Fan C, McLaws L. Primary Blast Causes Delayed Effects without Cell Death in Shell-Encased Brain Cell Aggregates. J Neurotrauma 2017; 35:174-186. [PMID: 28726571 DOI: 10.1089/neu.2016.4961] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Previous work in this laboratory used underwater explosive exposures to isolate the effects of shock-induced principle stress without shear on rat brain aggregate cultures. The current study has utilized simulated air blast to expose aggregates in suspension and enclosed within a spherical shell, enabling the examination of a much more complex biomechanical insult. Culture medium-filled spheres were exposed to single pulse overpressures of 15-30 psi (∼6-7 msec duration) and measurements within the sphere at defined sites showed complex and spatially dependent pressure changes. When brain aggregates were exposed to similar conditions, no cell death was observed and no changes in several commonly used biomarkers of traumatic brain injury (TBI) were noted. However, similarly to underwater blast, immediate and transient increases in the protein kinase B signaling pathway were observed at early time-points (3 days). In contrast, the oligodendrocyte marker 2',3'-cyclic nucleotide 3'-phosphodiesterase, as well as vascular endothelial growth factor, both displayed markedly delayed (14-28 days) and pressure-dependent responses. The imposition of a spherical shell between the single pulse shock wave and the target brain tissue introduces greatly increased complexity to the insult. This work shows that brain tissue can not only discriminate the nature of the pressure changes it experiences, but that a portion of its response is significantly delayed. These results have mechanistic implications for the study of primary blast-induced TBI and also highlight the importance of rigorously characterizing the actual pressure variations experienced by target tissue in primary blast studies.
Collapse
Affiliation(s)
- Thomas W Sawyer
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | | | - Yushan Wang
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Tyson Josey
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Mercy Villanueva
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Peggy Nelson
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Yanfeng Song
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Yimin Shei
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Grant Hennes
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Cory Vair
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | | | - Changyang Fan
- 4 Canada West Biosciences , Camrose, Alberta, Canada
| | - Lori McLaws
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| |
Collapse
|
13
|
Irvine KA, Clark JD. Chronic Pain After Traumatic Brain Injury: Pathophysiology and Pain Mechanisms. PAIN MEDICINE 2017; 19:1315-1333. [DOI: 10.1093/pm/pnx153] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Karen-Amanda Irvine
- Veterans Affairs Palo Alto Health Care System, Anesthesiology Service, Palo Alto, California
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - J David Clark
- Veterans Affairs Palo Alto Health Care System, Anesthesiology Service, Palo Alto, California
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
14
|
Wang Y, Sawyer TW, Tse YC, Fan C, Hennes G, Barnes J, Josey T, Weiss T, Nelson P, Wong TP. Primary Blast-Induced Changes in Akt and GSK 3β Phosphorylation in Rat Hippocampus. Front Neurol 2017; 8:413. [PMID: 28868045 PMCID: PMC5563325 DOI: 10.3389/fneur.2017.00413] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/31/2017] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) due to blast from improvised explosive devices has been a leading cause of morbidity and mortality in recent conflicts in Iraq and Afghanistan. However, the mechanisms of primary blast-induced TBI are not well understood. The Akt signal transduction pathway has been implicated in various brain pathologies including TBI. In the present study, the effects of simulated primary blast waves on the phosphorylation status of Akt and its downstream effector kinase, glycogen synthase kinase 3β (GSK3β), in rat hippocampus, were investigated. Male Sprague-Dawley (SD) rats (350–400 g) were exposed to a single pulse shock wave (25 psi; ~7 ms duration) and sacrificed 1 day, 1 week, or 6 weeks after exposure. Total and phosphorylated Akt, as well as phosphorylation of its downstream effector kinase GSK3β (at serine 9), were detected with western blot analysis and immunohistochemistry. Results showed that Akt phosphorylation at both serine 473 and threonine 308 was increased 1 day after blast on the ipsilateral side of the hippocampus, and this elevation persisted until at least 6 weeks postexposure. Similarly, phosphorylation of GSK3β at serine 9, which inhibits GSK3β activity, was also increased starting at 1 day and persisted until at least 6 weeks after primary blast on the ipsilateral side. In contrast, p-Akt was increased at 1 and 6 weeks on the contralateral side, while p-GSK3β was increased 1 day and 1 week after primary blast exposure. No significant changes in total protein levels of Akt and GSK were observed on either side of the hippocampus at any time points. Immunohistochemical results showed that increased p-Akt was mainly of neuronal origin in the CA1 region of the hippocampus and once phosphorylated, the majority was translocated to the dendritic and plasma membranes. Finally, electrophysiological data showed that evoked synaptic N-methyl-d-aspartate (NMDA) receptor activity was significantly increased 6 weeks after primary blast, suggesting that increased Akt phosphorylation may enhance synaptic NMDA receptor activation, or that enhanced synaptic NMDA receptor activation may increase Akt phosphorylation.
Collapse
Affiliation(s)
- Yushan Wang
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Thomas W Sawyer
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Yiu Chung Tse
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Changyang Fan
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Grant Hennes
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Julia Barnes
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Tyson Josey
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Tracy Weiss
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Peggy Nelson
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Tak Pan Wong
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Vogel EW, Morales FN, Meaney DF, Bass CR, Morrison B. Phosphodiesterase-4 inhibition restored hippocampal long term potentiation after primary blast. Exp Neurol 2017; 293:91-100. [PMID: 28366471 PMCID: PMC6016024 DOI: 10.1016/j.expneurol.2017.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/08/2017] [Accepted: 03/30/2017] [Indexed: 01/03/2023]
Abstract
Due to recent military conflicts and terrorist attacks, blast-induced traumatic brain injury (bTBI) presents a health concern for military and civilian personnel alike. Although secondary blast (penetrating injury) and tertiary blast (inertia-driven brain deformation) are known to be injurious, the effects of primary blast caused by the supersonic shock wave interacting with the skull and brain remain debated. Our group previously reported that in vitro primary blast exposure reduced long-term potentiation (LTP), the electrophysiological correlate of learning and memory, in rat organotypic hippocampal slice cultures (OHSCs) and that primary blast affects key proteins governing LTP. Recent studies have investigated phosphodiesterase-4 (PDE4) inhibition as a therapeutic strategy for reducing LTP deficits following inertia-driven TBI. We investigated the therapeutic potential of PDE4 inhibitors, specifically roflumilast, to ameliorate primary blast-induced deficits in LTP. We found that roflumilast at concentrations of 1nM or greater prevented deficits in neuronal plasticity measured 24h post-injury. We also observed a therapeutic window of at least 6h, but <23h. Additionally, we investigated molecular mechanisms that could elucidate this therapeutic effect. Roflumilast treatment (1nM delivered 6h post-injury) significantly increased total AMPA glutamate receptor 1 (GluR1) subunit expression, phosphorylation of the GluR1 subunit at the serine-831 site, and phosphorylation of stargazin at the serine-239/240 site upon LTP induction, measured 24h following injury. Roflumilast treatment significantly increased PSD-95 regardless of LTP induction. These findings indicate that further investigation into the translation of PDE4 inhibition as a therapy following bTBI is warranted.
Collapse
Affiliation(s)
- Edward W Vogel
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Fatima N Morales
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cameron R Bass
- Department of Biomedical Engineering, Duke University, Durham, NC 27705, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
16
|
Boothe DL, Yu AB, Kudela P, Anderson WS, Vettel JM, Franaszczuk PJ. Impact of Neuronal Membrane Damage on the Local Field Potential in a Large-Scale Simulation of Cerebral Cortex. Front Neurol 2017. [PMID: 28638364 PMCID: PMC5461262 DOI: 10.3389/fneur.2017.00236] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Within multiscale brain dynamics, the structure–function relationship between cellular changes at a lower scale and coordinated oscillations at a higher scale is not well understood. This relationship may be particularly relevant for understanding functional impairments after a mild traumatic brain injury (mTBI) when current neuroimaging methods do not reveal morphological changes to the brain common in moderate to severe TBI such as diffuse axonal injury or gray matter lesions. Here, we created a physiology-based model of cerebral cortex using a publicly released modeling framework (GEneral NEural SImulation System) to explore the possibility that performance deficits characteristic of blast-induced mTBI may reflect dysfunctional, local network activity influenced by microscale neuronal damage at the cellular level. We operationalized microscale damage to neurons as the formation of pores on the neuronal membrane based on research using blast paradigms, and in our model, pores were simulated by a change in membrane conductance. We then tracked changes in simulated electrical activity. Our model contained 585 simulated neurons, comprised of 14 types of cortical and thalamic neurons each with its own compartmental morphology and electrophysiological properties. Comparing the functional activity of neurons before and after simulated damage, we found that simulated pores in the membrane reduced both action potential generation and local field potential (LFP) power in the 1–40 Hz range of the power spectrum. Furthermore, the location of damage modulated the strength of these effects: pore formation on simulated axons reduced LFP power more strongly than did pore formation on the soma and the dendrites. These results indicate that even small amounts of cellular damage can negatively impact functional activity of larger scale oscillations, and our findings suggest that multiscale modeling provides a promising avenue to elucidate these relationships.
Collapse
Affiliation(s)
- David L Boothe
- U.S. Army Research Laboratory, Aberdeen Proving Ground, Aberdeen, MD, United States.,Altus Engineering, Churchville, MD, United States
| | - Alfred B Yu
- U.S. Army Research Laboratory, Aberdeen Proving Ground, Aberdeen, MD, United States
| | - Pawel Kudela
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,The Johns Hopkins Institute for Clinical and Translational Research, Baltimore, MD, United States
| | - William S Anderson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jean M Vettel
- U.S. Army Research Laboratory, Aberdeen Proving Ground, Aberdeen, MD, United States.,Psychological & Brain Sciences, University of California, Santa Barbara, CA, United States.,Department of Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Piotr J Franaszczuk
- U.S. Army Research Laboratory, Aberdeen Proving Ground, Aberdeen, MD, United States.,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
17
|
Lucke-Wold BP, Turner RC, Logsdon AF, Rosen CL, Qaiser R. Blast Scaling Parameters: Transitioning from Lung to Skull Base Metrics. JOURNAL OF SURGERY AND EMERGENCY MEDICINE 2017; 1. [PMID: 28386605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 09/28/2022]
Abstract
Neurotrauma from blast exposure is one of the single most characteristic injuries of modern warfare. Understanding blast traumatic brain injury is critical for developing new treatment options for warfighters and civilians exposed to improvised explosive devices. Unfortunately, the pre-clinical models that are widely utilized to investigate blast exposure are based on archaic lung based parameters developed in the early 20th century. Improvised explosive devices produce a different type of injury paradigm than the typical mortar explosion. Protective equipment for the chest cavity has also improved over the past 100 years. In order to improve treatments, it is imperative to develop models that are based more on skull-based parameters. In this mini-review, we discuss the important anatomical and biochemical features necessary to develop a skull-based model.
Collapse
Affiliation(s)
| | - Ryan C Turner
- Department of Neurosurgery, West Virginia University, Morgantown, WV, USA
| | | | - Charles L Rosen
- Department of Neurosurgery, West Virginia University, Morgantown, WV, USA
| | - Rabia Qaiser
- Department of Neurosurgery, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
18
|
Miller AP, Shah AS, Aperi BV, Kurpad SN, Stemper BD, Glavaski-Joksimovic A. Acute death of astrocytes in blast-exposed rat organotypic hippocampal slice cultures. PLoS One 2017; 12:e0173167. [PMID: 28264063 PMCID: PMC5338800 DOI: 10.1371/journal.pone.0173167] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 02/16/2017] [Indexed: 01/06/2023] Open
Abstract
Blast traumatic brain injury (bTBI) affects civilians, soldiers, and veterans worldwide and presents significant health concerns. The mechanisms of neurodegeneration following bTBI remain elusive and current therapies are largely ineffective. It is important to better characterize blast-evoked cellular changes and underlying mechanisms in order to develop more effective therapies. In the present study, our group utilized rat organotypic hippocampal slice cultures (OHCs) as an in vitro system to model bTBI. OHCs were exposed to either 138 ± 22 kPa (low) or 273 ± 23 kPa (high) overpressures using an open-ended helium-driven shock tube, or were assigned to sham control group. At 2 hours (h) following injury, we have characterized the astrocytic response to a blast overpressure. Immunostaining against the astrocytic marker glial fibrillary acidic protein (GFAP) revealed acute shearing and morphological changes in astrocytes, including clasmatodendrosis. Moreover, overlap of GFAP immunostaining and propidium iodide (PI) indicated astrocytic death. Quantification of the number of dead astrocytes per counting area in the hippocampal cornu Ammonis 1 region (CA1), demonstrated a significant increase in dead astrocytes in the low- and high-blast, compared to sham control OHCs. However only a small number of GFAP-expressing astrocytes were co-labeled with the apoptotic marker Annexin V, suggesting necrosis as the primary type of cell death in the acute phase following blast exposure. Moreover, western blot analyses revealed calpain mediated breakdown of GFAP. The dextran exclusion additionally indicated membrane disruption as a potential mechanism of acute astrocytic death. Furthermore, although blast exposure did not evoke significant changes in glutamate transporter 1 (GLT-1) expression, loss of GLT-1-expressing astrocytes suggests dysregulation of glutamate uptake following injury. Our data illustrate the profound effect of blast overpressure on astrocytes in OHCs at 2 h following injury and suggest increased calpain activity and membrane disruption as potential underlying mechanisms.
Collapse
Affiliation(s)
- Anna P. Miller
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Alok S. Shah
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Brandy V. Aperi
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Shekar N. Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Brian D. Stemper
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| | - Aleksandra Glavaski-Joksimovic
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
19
|
Effgen GB, Morrison B. Electrophysiological and Pathological Characterization of the Period of Heightened Vulnerability to Repetitive Injury in an in Vitro Stretch Model. J Neurotrauma 2017; 34:914-924. [DOI: 10.1089/neu.2016.4477] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Gwen B. Effgen
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, New York
| |
Collapse
|
20
|
Sawyer TW, Lee JJ, Villanueva M, Wang Y, Nelson P, Song Y, Fan C, Barnes J, McLaws L. The Effect of Underwater Blast on Aggregating Brain Cell Cultures. J Neurotrauma 2017; 34:517-528. [PMID: 27163293 DOI: 10.1089/neu.2016.4430] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Although the deleterious effects of primary blast on gas-filled organs are well accepted, the effect of blast-induced shock waves on the brain is less clear because of factors that complicate the interpretation of clinical and experimental data. Brain cell aggregate cultures are comprised of multiple differentiated brain cell types and were used to examine the effects of underwater blast. Suspensions of these cultures encased in dialysis tubing were exposed to explosive-generated underwater blasts of low (∼300 kPa), medium (∼2,700 kPa), or high (∼14,000 kPa) intensities and harvested at 1-28 days post-exposure. No changes in gross morphology were noted immediately or weeks after blast wave exposure, and no increases in either apoptotic (caspase-3) or necrotic (lactate dehydrogenase) cell death were observed. Changes in neuronal (neurofilament H, acetylcholinesterase, and choline acetyltransferase) and glial (glial fibrillary acidic protein, glutamine synthetase) endpoints did not occur. However, significant time- and pressure-related increases in Akt (protein kinase B) phosphorylation were noted, as well as declines in vascular endothelial growth factor levels, implicating pathways involved in cellular survival mechanisms. The free-floating nature of the aggregates during blast wave exposure, coupled with their highly hydrolyzed dialysis tubing containment, results in minimized boundary effects, thus enabling accurate assessment of brain cell response to a simplified shock-induced stress wave. This work shows that, at its simplest, blast-induced shock waves produce subtle changes in brain tissue. This study has mechanistic implications for the study of primary blast-induced traumatic brain injury and supports the thesis that underwater blast may cause subtle changes in the brains of submerged individuals.
Collapse
Affiliation(s)
- Thomas W Sawyer
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Julian J Lee
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Mercy Villanueva
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Yushan Wang
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Peggy Nelson
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Yanfeng Song
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| | - Chengyang Fan
- 2 Canada West Biosciences , Calgary, Alberta, Canada
| | - Julia Barnes
- 3 Hyland Quality Systems , Medicine Hat, Alberta, Canada
| | - Lori McLaws
- 1 Defence Research and Development Canada, Suffield Research Center , Medicine Hat, Alberta, Canada
| |
Collapse
|
21
|
Vogel EW, Rwema SH, Meaney DF, Bass CRD, Morrison B. Primary Blast Injury Depressed Hippocampal Long-Term Potentiation through Disruption of Synaptic Proteins. J Neurotrauma 2016; 34:1063-1073. [PMID: 27573357 DOI: 10.1089/neu.2016.4578] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Blast-induced traumatic brain injury (bTBI) is a major threat to United States service members in military conflicts worldwide. The effects of primary blast, caused by the supersonic shockwave interacting with the skull and brain, remain unclear. Our group has previously reported that in vitro primary blast exposure can reduce long-term potentiation (LTP), the electrophysiological correlate of learning and memory, in rat organotypic hippocampal slice cultures (OHSCs) without significant changes to cell viability or basal, evoked neuronal function. We investigated the time course of primary blast-induced deficits in LTP and the molecular mechanisms that could underlie these deficits. We found that pure primary blast exposure induced LTP deficits in a delayed manner, requiring longer than 1 hour to develop, and that these deficits spontaneously recovered by 10 days following exposure depending on blast intensity. Additionally, we observed that primary blast exposure reduced total α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptor 1 (GluR1) subunit expression and phosphorylation of the GluR1 subunit at the serine-831 site. Blast also reduced the expression of postsynaptic density protein-95 (PSD-95) and phosphorylation of stargazin protein at the serine-239/240 site. Finally, we found that modulation of the cyclic adenosine monophosphate (cAMP) pathway ameliorated electrophysiological and protein-expression changes caused by blast. These findings could inform the development of novel therapies to treat blast-induced loss of neuronal function.
Collapse
Affiliation(s)
- Edward W Vogel
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Steve H Rwema
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - David F Meaney
- 2 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Cameron R Dale Bass
- 3 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Barclay Morrison
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
22
|
Smith M, Piehler T, Benjamin R, Farizatto KL, Pait MC, Almeida MF, Ghukasyan VV, Bahr BA. Blast waves from detonated military explosive reduce GluR1 and synaptophysin levels in hippocampal slice cultures. Exp Neurol 2016; 286:107-115. [PMID: 27720798 DOI: 10.1016/j.expneurol.2016.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 12/15/2022]
Abstract
Explosives create shockwaves that cause blast-induced neurotrauma, one of the most common types of traumatic brain injury (TBI) linked to military service. Blast-induced TBIs are often associated with reduced cognitive and behavioral functions due to a variety of factors. To study the direct effects of military explosive blasts on brain tissue, we removed systemic factors by utilizing rat hippocampal slice cultures. The long-term slice cultures were briefly sealed air-tight in serum-free medium, lowered into a 37°C water-filled tank, and small 1.7-gram assemblies of cyclotrimethylene trinitramine (RDX) were detonated 15cm outside the tank, creating a distinct shockwave recorded at the culture plate position. Compared to control mock-treated groups of slices that received equal submerge time, 1-3 blast impacts caused a dose-dependent reduction in the AMPA receptor subunit GluR1. While only a small reduction was found in hippocampal slices exposed to a single RDX blast and harvested 1-2days later, slices that received two consecutive RDX blasts 4min apart exhibited a 26-40% reduction in GluR1, and the receptor subunit was further reduced by 64-72% after three consecutive blasts. Such loss correlated with increased levels of HDAC2, a histone deacetylase implicated in stress-induced reduction of glutamatergic transmission. No evidence of synaptic marker recovery was found at 72h post-blast. The presynaptic marker synaptophysin was found to have similar susceptibility as GluR1 to the multiple explosive detonations. In contrast to the synaptic protein reductions, actin levels were unchanged, spectrin breakdown was not detected, and Fluoro-Jade B staining found no indication of degenerating neurons in slices exposed to three RDX blasts, suggesting that small, sub-lethal explosives are capable of producing selective alterations to synaptic integrity. Together, these results indicate that blast waves from military explosive cause signs of synaptic compromise without producing severe neurodegeneration, perhaps explaining the cognitive and behavioral changes in those blast-induced TBI sufferers that have no detectable neuropathology.
Collapse
Affiliation(s)
- Marquitta Smith
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Thuvan Piehler
- U.S. Army Research Laboratory, Aberdeen Proving Ground, MD 21005, USA
| | - Richard Benjamin
- U.S. Army Research Laboratory, Aberdeen Proving Ground, MD 21005, USA
| | - Karen L Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Morgan C Pait
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Vladimir V Ghukasyan
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA.
| |
Collapse
|
23
|
Zander NE, Piehler T, Banton R, Benjamin R. Effects of repetitive low-pressure explosive blast on primary neurons and mixed cultures. J Neurosci Res 2016; 94:827-36. [PMID: 27317559 DOI: 10.1002/jnr.23786] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/28/2016] [Accepted: 05/23/2016] [Indexed: 02/05/2023]
Abstract
Repetitive mild traumatic brain injury represents a considerable health concern, particularly for athletes and military personnel. For blast-induced brain injury, threshold shock-impulse levels required to induce such injuries and cumulative effects with single and/or multiple exposures are not well characterized. Currently, there is no established in vitro experimental model with blast pressure waves generated by live explosives. This study presents results of primary neurons and mixed cultures subjected to our unique in vitro indoor experimental platform that uses real military explosive charges to probe the effects of primary explosive blast at the cellular level. The effects of the blast on membrane permeability, generation of reactive oxygen species (ROS), uptake of sodium ions, intracellular calcium, and release of glutamate were probed 2 and 24 hr postblast. Significant changes in membrane permeability and sodium uptake among the sham, single-blast-injured, and triple-blast-injured samples were observed. A significant increase in ROS and glutamate release was observed for the triple-blast-injured samples compared with the sham. Changes in intracellular calcium were not significant. These results suggest that blast exposure disrupts the integrity of the plasma membrane, leading to the upset of ion homeostasis, formation of ROS, and glutamate release. Published 2016. †This article is a U.S. Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- Nicole E Zander
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Thuvan Piehler
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Rohan Banton
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Richard Benjamin
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| |
Collapse
|
24
|
Beamer M, Tummala SR, Gullotti D, Kopil C, Gorka S, Bass CRD, Morrison B, Cohen AS, Meaney DF. Primary blast injury causes cognitive impairments and hippocampal circuit alterations. Exp Neurol 2016; 283:16-28. [PMID: 27246999 DOI: 10.1016/j.expneurol.2016.05.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/14/2016] [Accepted: 05/20/2016] [Indexed: 11/17/2022]
Abstract
Blast-induced traumatic brain injury (bTBI) and its long term consequences are a major health concern among veterans. Despite recent work enhancing our knowledge about bTBI, very little is known about the contribution of the blast wave alone to the observed sequelae. Herein, we isolated its contribution in a mouse model by constraining the animals' heads during exposure to a shockwave (primary blast). Our results show that exposure to primary blast alone results in changes in hippocampus-dependent behaviors that correspond with electrophysiological changes in area CA1 and are accompanied by reactive gliosis. Specifically, five days after exposure, behavior in an open field and performance in a spatial object recognition (SOR) task were significantly different from sham. Network electrophysiology, also performed five days after injury, demonstrated a significant decrease in excitability and increase in inhibitory tone. Immunohistochemistry for GFAP and Iba1 performed ten days after injury showed a significant increase in staining. Interestingly, a threefold increase in the impulse of the primary blast wave did not exacerbate these measures. However, we observed a significant reduction in the contribution of the NMDA receptors to the field EPSP at the highest blast exposure level. Our results emphasize the need to account for the effects of primary blast loading when studying the sequelae of bTBI.
Collapse
Affiliation(s)
- Matthew Beamer
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Shanti R Tummala
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - David Gullotti
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine Kopil
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Gorka
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA
| | | | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Akiva S Cohen
- Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David F Meaney
- Department of Bioengineering(1), University of Pennsylvania, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Zander NE, Piehler T, Banton R, Boggs M. The effect of explosive blast loading on human neuroblastoma cells. Anal Biochem 2016; 504:4-6. [PMID: 27033003 DOI: 10.1016/j.ab.2016.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/10/2016] [Accepted: 03/20/2016] [Indexed: 10/22/2022]
Abstract
Diagnosis of mild to moderate traumatic brain injury is challenging because brain tissue damage progresses slowly and is not readily detectable by conventional imaging techniques. We have developed a novel in vitro model to study primary blast loading on dissociated neurons using nitroamine explosives such as those used on the battlefield. Human neuroblastoma cells were exposed to single and triple 50-psi explosive blasts and single 100-psi blasts. Changes in membrane permeability and oxidative stress showed a significant increase for the single and triple 100-psi blast conditions compared with single 50-psi blast and controls.
Collapse
Affiliation(s)
- Nicole E Zander
- U.S. Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, MD 21005, USA.
| | - Thuvan Piehler
- U.S. Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, MD 21005, USA
| | - Rohan Banton
- U.S. Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, MD 21005, USA
| | - Mary Boggs
- Department of Biology, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
26
|
Effgen GB, Ong T, Nammalwar S, Ortuño AI, Meaney DF, 'Dale' Bass CR, Morrison B. Primary Blast Exposure Increases Hippocampal Vulnerability to Subsequent Exposure: Reducing Long-Term Potentiation. J Neurotrauma 2016; 33:1901-1912. [PMID: 26699926 DOI: 10.1089/neu.2015.4327] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Up to 80% of injuries sustained by U.S. soldiers in Operation Enduring Freedom and Operation Iraqi Freedom were the result of blast exposure from improvised explosive devices. Some soldiers experience multiple blasts while on duty, and it has been suggested that symptoms of repetitive blast are similar to those that follow multiple non-blast concussions, such as sport-related concussion. Despite the interest in the effects of repetitive blast exposure, it remains unknown whether an initial blast renders the brain more vulnerable to subsequent exposure, resulting in a synergistic injury response. To investigate the effect of multiple primary blasts on the brain, organotypic hippocampal slice cultures were exposed to single or repetitive (two or three total) primary blasts of varying intensities. Long-term potentiation was significantly reduced following two Level 2 (92.7 kPa, 1.4 msec, 38.5 kPa·msec) blasts delivered 24 h apart without altering basal evoked response. This deficit persisted when the interval between injuries was increased to 72 h but not when the interval was extended to 144 h. The repeated blast exposure with a 24 h interval increased microglia staining and activation significantly but did not significantly increase cell death or damage axons, dendrites, or principal cell layers. Lack of overt structural damage and change in basal stimulated neuron response suggest that injury from repetitive primary blast exposure may specifically affect long-term potentiation. Our studies suggest repetitive primary blasts can exacerbate injury dependent on the injury severity and interval between exposures.
Collapse
Affiliation(s)
- Gwen B Effgen
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Tiffany Ong
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Shruthi Nammalwar
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Andrea I Ortuño
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - David F Meaney
- 2 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | | | - Barclay Morrison
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
27
|
Stemper BD, Shah AS, Budde MD, Olsen CM, Glavaski-Joksimovic A, Kurpad SN, McCrea M, Pintar FA. Behavioral Outcomes Differ between Rotational Acceleration and Blast Mechanisms of Mild Traumatic Brain Injury. Front Neurol 2016; 7:31. [PMID: 27014184 PMCID: PMC4789366 DOI: 10.3389/fneur.2016.00031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/29/2016] [Indexed: 11/20/2022] Open
Abstract
Mild traumatic brain injury (mTBI) can result from a number of mechanisms, including blunt impact, head rotational acceleration, exposure to blast, and penetration of projectiles. Mechanism is likely to influence the type, severity, and chronicity of outcomes. The objective of this study was to determine differences in the severity and time course of behavioral outcomes following blast and rotational mTBI. The Medical College of Wisconsin (MCW) Rotational Injury model and a shock tube model of primary blast injury were used to induce mTBI in rats and behavioral assessments were conducted within the first week, as well as 30 and 60 days following injury. Acute recovery time demonstrated similar increases over protocol-matched shams, indicating acute injury severity equivalence between the two mechanisms. Post-injury behavior in the elevated plus maze demonstrated differing trends, with rotationally injured rats acutely demonstrating greater activity, whereas blast-injured rats had decreased activity that developed at chronic time points. Similarly, blast-injured rats demonstrated trends associated with cognitive deficits that were not apparent following rotational injuries. These findings demonstrate that rotational and blast injury result in behavioral changes with different qualitative and temporal manifestations. Whereas rotational injury was characterized by a rapidly emerging phenotype consistent with behavioral disinhibition, blast injury was associated with emotional and cognitive differences that were not evident acutely, but developed later, with an anxiety-like phenotype still present in injured animals at our most chronic measurements.
Collapse
Affiliation(s)
- Brian D. Stemper
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alok S. Shah
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Matthew D. Budde
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher M. Olsen
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Shekar N. Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Frank A. Pintar
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| |
Collapse
|
28
|
Przekwas A, Somayaji MR, Gupta RK. Synaptic Mechanisms of Blast-Induced Brain Injury. Front Neurol 2016; 7:2. [PMID: 26834697 PMCID: PMC4720734 DOI: 10.3389/fneur.2016.00002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 01/04/2016] [Indexed: 01/08/2023] Open
Abstract
Blast wave-induced traumatic brain injury (TBI) is one of the most common injuries to military personnel. Brain tissue compression/tension due to blast-induced cranial deformations and shear waves due to head rotation may generate diffuse micro-damage to neuro-axonal structures and trigger a cascade of neurobiological events culminating in cognitive and neurodegenerative disorders. Although diffuse axonal injury is regarded as a signature wound of mild TBI (mTBI), blast loads may also cause synaptic injury wherein neuronal synapses are stretched and sheared. This synaptic injury may result in temporary disconnect of the neural circuitry and transient loss in neuronal communication. We hypothesize that mTBI symptoms such as loss of consciousness or dizziness, which start immediately after the insult, could be attributed to synaptic injury. Although empirical evidence is beginning to emerge; the detailed mechanisms underlying synaptic injury are still elusive. Coordinated in vitro-in vivo experiments and mathematical modeling studies can shed light into the synaptic injury mechanisms and their role in the potentiation of mTBI symptoms.
Collapse
Affiliation(s)
- Andrzej Przekwas
- Computational Medicine and Biology Division, CFD Research Corporation, Huntsville, AL, USA
| | | | - Raj K. Gupta
- Department of Defense Blast Injury Research Program Coordinating Office, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| |
Collapse
|
29
|
Vogel EW, Effgen GB, Patel TP, Meaney DF, Bass CRD, Morrison B. Isolated Primary Blast Inhibits Long-Term Potentiation in Organotypic Hippocampal Slice Cultures. J Neurotrauma 2015; 33:652-61. [PMID: 26414012 DOI: 10.1089/neu.2015.4045] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Over the last 13 years, traumatic brain injury (TBI) has affected over 230,000 U.S. service members through the conflicts in Iraq and Afghanistan, mostly as a result of exposure to blast events. Blast-induced TBI (bTBI) is multi-phasic, with the penetrating and inertia-driven phases having been extensively studied. The effects of primary blast injury, caused by the shockwave interacting with the brain, remain unclear. Earlier in vivo studies in mice and rats have reported mixed results for primary blast effects on behavior and memory. Using a previously developed shock tube and in vitro sample receiver, we investigated the effect of isolated primary blast on the electrophysiological function of rat organotypic hippocampal slice cultures (OHSC). We found that pure primary blast exposure inhibited long-term potentiation (LTP), the electrophysiological correlate of memory, with a threshold between 9 and 39 kPa·ms impulse. This deficit occurred well below a previously identified threshold for cell death (184 kPa·ms), supporting our previously published finding that primary blast can cause changes in brain function in the absence of cell death. Other functional measures such as spontaneous activity, network synchronization, stimulus-response curves, and paired-pulse ratios (PPRs) were less affected by primary blast exposure, as compared with LTP. This is the first study to identify a tissue-level tolerance threshold for electrophysiological changes in neuronal function to isolated primary blast.
Collapse
Affiliation(s)
- Edward W Vogel
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Gwen B Effgen
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Tapan P Patel
- 2 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - David F Meaney
- 2 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Cameron R Dale Bass
- 3 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Barclay Morrison
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
30
|
Miyazaki H, Miyawaki H, Satoh Y, Saiki T, Kawauchi S, Sato S, Saitoh D. Thoracic shock wave injury causes behavioral abnormalities in mice. Acta Neurochir (Wien) 2015; 157:2111-20; discussion 2120. [PMID: 26489739 DOI: 10.1007/s00701-015-2613-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/09/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Mild traumatic brain injury (mTBI) is caused by complex mechanisms of systemic, local and cerebral responses to blast exposure. However, the molecular mechanisms of cognitive impairment after exposure to blast waves are not clearly known. We tested the hypothesis that thoracic injury induced functional and morphological impairment in the brain, leading to behavioral abnormalities. METHODS Mice were exposed to laser-induced shock waves (LISWs) impacting the thorax and assessed for behavioral outcome at 7 and 28 days post injury. Hippocampus and lung were collected for histopathological analysis and gene expression profiling after injury. RESULTS Thoracic injury transiently decreased the heart rate, blood pressure, peripheral oxyhemoglobin saturation and cerebral blood flow immediately after LISW exposure. Although LISWs exposure caused pulmonary contusions, hemorrhage was not apparent in the brain. At 7 and 28 days after, the injured mice exhibited impaired short-term memory and depression-like behavior compared with controls. Histological assessments showed an increase in neuronal cell death after shock wave exposure, especially in the CA3 region of the hippocampus. Moreover, shock wave exposure altered the expression of functionally relevant genes in the hippocampus at 1 h and 1 day post injury. CONCLUSIONS Our findings indicate that the LISW-induced thoracic injury with no direct impact on the brain affected the hippocampal gene expression and led to morphological alterations, resulting in behavioral abnormalities. Therefore, body protection may be extremely important in the effective prevention against blast-induced alterations in brain function.
Collapse
Affiliation(s)
- Hiromi Miyazaki
- Division of Traumatology, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Hiroki Miyawaki
- Department of Traumatology and Critical Care Medicine, National Defense Medical College Hospital, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yasushi Satoh
- Department of Anesthesiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takami Saiki
- Division of Traumatology, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Satoko Kawauchi
- Division of Biomedical Information Sciences, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Shunichi Sato
- Division of Biomedical Information Sciences, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Daizoh Saitoh
- Division of Traumatology, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| |
Collapse
|
31
|
Lucke-Wold BP, Logsdon AF, Smith KE, Turner RC, Alkon DL, Tan Z, Naser ZJ, Knotts CM, Huber JD, Rosen CL. Bryostatin-1 Restores Blood Brain Barrier Integrity following Blast-Induced Traumatic Brain Injury. Mol Neurobiol 2015; 52:1119-1134. [PMID: 25301233 PMCID: PMC5000781 DOI: 10.1007/s12035-014-8902-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/24/2014] [Indexed: 02/08/2023]
Abstract
Recent wars in Iraq and Afghanistan have accounted for an estimated 270,000 blast exposures among military personnel. Blast traumatic brain injury (TBI) is the 'signature injury' of modern warfare. Blood brain barrier (BBB) disruption following blast TBI can lead to long-term and diffuse neuroinflammation. In this study, we investigate for the first time the role of bryostatin-1, a specific protein kinase C (PKC) modulator, in ameliorating BBB breakdown. Thirty seven Sprague-Dawley rats were used for this study. We utilized a clinically relevant and validated blast model to expose animals to moderate blast exposure. Groups included: control, single blast exposure, and single blast exposure + bryostatin-1. Bryostatin-1 was administered i.p. 2.5 mg/kg after blast exposure. Evan's blue, immunohistochemistry, and western blot analysis were performed to assess injury. Evan's blue binds to albumin and is a marker for BBB disruption. The single blast exposure caused an increase in permeability compared to control (t = 4.808, p < 0.05), and a reduction back toward control levels when bryostatin-1 was administered (t = 5.113, p < 0.01). Three important PKC isozymes, PKCα, PKCδ, and PKCε, were co-localized primarily with endothelial cells but not astrocytes. Bryostatin-1 administration reduced toxic PKCα levels back toward control levels (t = 4.559, p < 0.01) and increased the neuroprotective isozyme PKCε (t = 6.102, p < 0.01). Bryostatin-1 caused a significant increase in the tight junction proteins VE-cadherin, ZO-1, and occludin through modulation of PKC activity. Bryostatin-1 ultimately decreased BBB breakdown potentially due to modulation of PKC isozymes. Future work will examine the role of bryostatin-1 in preventing chronic neurodegeneration following repetitive neurotrauma.
Collapse
Affiliation(s)
- Brandon P Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Aric F Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Kelly E Smith
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Ryan C Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Daniel L Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, 26506, USA
| | - Zhenjun Tan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Zachary J Naser
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Office of Professional Studies in Health Sciences, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Chelsea M Knotts
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Jason D Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Charles L Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Department of Neurosurgery, West Virginia University School of Medicine, One Medical Center Drive, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV, 26506-9183, USA.
| |
Collapse
|
32
|
Hue CD, Cho FS, Cao S, Nicholls RE, Vogel Iii EW, Sibindi C, Arancio O, Dale Bass CR, Meaney DF, Morrison Iii B. Time Course and Size of Blood-Brain Barrier Opening in a Mouse Model of Blast-Induced Traumatic Brain Injury. J Neurotrauma 2015; 33:1202-11. [PMID: 26414212 DOI: 10.1089/neu.2015.4067] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
An increasing number of studies have reported blood-brain barrier (BBB) dysfunction after blast-induced traumatic brain injury (bTBI). Despite this evidence, there is limited quantitative understanding of the extent of BBB opening and the time course of damage after blast injury. In addition, many studies do not report kinematic parameters of head motion, making it difficult to separate contributions of primary and tertiary blast-loading. Detailed characterization of blast-induced BBB damage may hold important implications for serum constituents that may potentially cross the compromised barrier and contribute to neurotoxicity, neuroinflammation, and persistent neurologic deficits. Using an in vivo bTBI model, systemic administration of sodium fluorescein (NaFl; 376 Da), Evans blue (EB; 69 kDa when bound to serum albumin), and dextrans (3-500 kDa) was used to estimate the pore size of BBB opening and the time required for recovery. Exposure to blast with 272 ± 6 kPa peak overpressure, 0.69 ± 0.01 ms duration, and 65 ± 1 kPa*ms impulse resulted in significant acute extravasation of NaFl, 3 kDa dextran, and EB. However, there was no significant acute extravasation of 70 kDa or 500 kDa dextrans, and minimal to no extravasation of NaFl, dextrans, or EB 1 day after exposure. This study presents a detailed analysis of the time course and pore size of BBB opening after bTBI, supported by a characterization of kinematic parameters associated with blast-induced head motion.
Collapse
Affiliation(s)
- Christopher D Hue
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Frances S Cho
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Siqi Cao
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Russell E Nicholls
- 2 Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University , New York, New York
| | - Edward W Vogel Iii
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Cosmas Sibindi
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Ottavio Arancio
- 2 Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University , New York, New York
| | - Cameron R Dale Bass
- 3 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - David F Meaney
- 4 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Barclay Morrison Iii
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
33
|
Zander NE, Piehler T, Boggs ME, Banton R, Benjamin R. In vitro studies of primary explosive blast loading on neurons. J Neurosci Res 2015; 93:1353-63. [PMID: 25914380 DOI: 10.1002/jnr.23594] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 03/18/2015] [Accepted: 03/30/2015] [Indexed: 12/28/2022]
Abstract
In a military setting, traumatic brain injury (TBI) is frequently caused by blast waves that can trigger a series of neuronal biochemical changes. Although many animal models have been used to study the effects of primary blast waves, elucidating the mechanisms of damage in a whole-animal model is extremely complex. In vitro models of primary blast, which allow for the deconvolution of mechanisms, are relatively scarce. It is largely unknown how structural damage at the cellular level impacts the functional activity at variable time scales after the TBI event. A novel in vitro system was developed to probe the effects of explosive blast (ranging from ∼25 to 40 psi) on dissociated neurons. PC12 neurons were cultured on laminin-coated substrates, submerged underwater, and subjected to single and multiple blasts in a controlled environment. Changes in cell membrane permeability, viability, and cell morphology were evaluated. Significant increases in axonal beading were observed in the injured cells. In addition, although cell death was minimal after a single insult, cell viability decreased significantly following repeated blast exposure.
Collapse
Affiliation(s)
- Nicole E Zander
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Thuvan Piehler
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Mary E Boggs
- Department of Biology, University of Delaware, Newark, Delaware
| | - Rohan Banton
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| | - Richard Benjamin
- United States Army Research Laboratory, Weapons and Materials Research Directorate, Aberdeen Proving Ground, Aberdeen, Maryland
| |
Collapse
|
34
|
Miller AP, Shah AS, Aperi BV, Budde MD, Pintar FA, Tarima S, Kurpad SN, Stemper BD, Glavaski-Joksimovic A. Effects of blast overpressure on neurons and glial cells in rat organotypic hippocampal slice cultures. Front Neurol 2015; 6:20. [PMID: 25729377 PMCID: PMC4325926 DOI: 10.3389/fneur.2015.00020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 01/25/2015] [Indexed: 11/13/2022] Open
Abstract
Due to recent involvement in military conflicts, and an increase in the use of explosives, there has been an escalation in the incidence of blast-induced traumatic brain injury (bTBI) among US military personnel. Having a better understanding of the cellular and molecular cascade of events in bTBI is prerequisite for the development of an effective therapy that currently is unavailable. The present study utilized organotypic hippocampal slice cultures (OHCs) exposed to blast overpressures of 150 kPa (low) and 280 kPa (high) as an in vitro bTBI model. Using this model, we further characterized the cellular effects of the blast injury. Blast-evoked cell death was visualized by a propidium iodide (PI) uptake assay as early as 2 h post-injury. Quantification of PI staining in the cornu Ammonis 1 and 3 (CA1 and CA3) and the dentate gyrus regions of the hippocampus at 2, 24, 48, and 72 h following blast exposure revealed significant time dependent effects. OHCs exposed to 150 kPa demonstrated a slow increase in cell death plateauing between 24 and 48 h, while OHCs from the high-blast group exhibited a rapid increase in cell death already at 2 h, peaking at ~24 h post-injury. Measurements of lactate dehydrogenase release into the culture medium also revealed a significant increase in cell lysis in both low- and high-blast groups compared to sham controls. OHCs were fixed at 72 h post-injury and immunostained for markers against neurons, astrocytes, and microglia. Labeling OHCs with PI, neuronal, and glial markers revealed that the blast-evoked extensive neuronal death and to a lesser extent loss of glial cells. Furthermore, our data demonstrated activation of astrocytes and microglial cells in low- and high-blasted OHCs, which reached a statistically significant difference in the high-blast group. These data confirmed that our in vitro bTBI model is a useful tool for studying cellular and molecular changes after blast exposure.
Collapse
Affiliation(s)
- Anna P Miller
- Department of Neurosurgery, Medical College of Wisconsin , Milwaukee, WI , USA ; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin , Milwaukee, WI , USA ; Clement J. Zablocki Veterans Affairs Medical Center , Milwaukee, WI , USA
| | - Alok S Shah
- Department of Neurosurgery, Medical College of Wisconsin , Milwaukee, WI , USA ; Clement J. Zablocki Veterans Affairs Medical Center , Milwaukee, WI , USA
| | - Brandy V Aperi
- Department of Neurosurgery, Medical College of Wisconsin , Milwaukee, WI , USA ; Clement J. Zablocki Veterans Affairs Medical Center , Milwaukee, WI , USA
| | - Matthew D Budde
- Department of Neurosurgery, Medical College of Wisconsin , Milwaukee, WI , USA ; Clement J. Zablocki Veterans Affairs Medical Center , Milwaukee, WI , USA
| | - Frank A Pintar
- Department of Neurosurgery, Medical College of Wisconsin , Milwaukee, WI , USA ; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin , Milwaukee, WI , USA ; Clement J. Zablocki Veterans Affairs Medical Center , Milwaukee, WI , USA
| | - Sergey Tarima
- Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin , Milwaukee, WI , USA
| | - Shekar N Kurpad
- Department of Neurosurgery, Medical College of Wisconsin , Milwaukee, WI , USA ; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin , Milwaukee, WI , USA ; Clement J. Zablocki Veterans Affairs Medical Center , Milwaukee, WI , USA
| | - Brian D Stemper
- Department of Neurosurgery, Medical College of Wisconsin , Milwaukee, WI , USA ; Clement J. Zablocki Veterans Affairs Medical Center , Milwaukee, WI , USA
| | - Aleksandra Glavaski-Joksimovic
- Department of Neurosurgery, Medical College of Wisconsin , Milwaukee, WI , USA ; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin , Milwaukee, WI , USA ; Clement J. Zablocki Veterans Affairs Medical Center , Milwaukee, WI , USA
| |
Collapse
|
35
|
Heinzelmann M, Reddy SY, French LM, Wang D, Lee H, Barr T, Baxter T, Mysliwiec V, Gill J. Military personnel with chronic symptoms following blast traumatic brain injury have differential expression of neuronal recovery and epidermal growth factor receptor genes. Front Neurol 2014; 5:198. [PMID: 25346719 PMCID: PMC4191187 DOI: 10.3389/fneur.2014.00198] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/18/2014] [Indexed: 01/23/2023] Open
Abstract
Objective: Approximately one-quarter of military personnel who deployed to combat stations sustained one or more blast-related, closed-head injuries. Blast injuries result from the detonation of an explosive device. The mechanisms associated with blast exposure that give rise to traumatic brain injury (TBI), and place military personnel at high risk for chronic symptoms of post-concussive disorder (PCD), post-traumatic stress disorder (PTSD), and depression are not elucidated. Methods: To investigate the mechanisms of persistent blast-related symptoms, we examined expression profiles of transcripts across the genome to determine the role of gene activity in chronic symptoms following blast-TBI. Active duty military personnel with (1) a medical record of a blast-TBI that occurred during deployment (n = 19) were compared to control participants without TBI (n = 17). Controls were matched to cases on demographic factors including age, gender, and race, and also in diagnoses of sleep disturbance, and symptoms of PTSD and depression. Due to the high number of PCD symptoms in the TBI+ group, we did not match on this variable. Using expression profiles of transcripts in microarray platform in peripheral samples of whole blood, significantly differentially expressed gene lists were generated. Statistical threshold is based on criteria of 1.5 magnitude fold-change (up or down) and p-values with multiple test correction (false discovery rate <0.05). Results: There were 34 transcripts in 29 genes that were differentially regulated in blast-TBI participants compared to controls. Up-regulated genes included epithelial cell transforming sequence and zinc finger proteins, which are necessary for astrocyte differentiation following injury. Tensin-1, which has been implicated in neuronal recovery in pre-clinical TBI models, was down-regulated in blast-TBI participants. Protein ubiquitination genes, such as epidermal growth factor receptor, were also down-regulated and identified as the central regulators in the gene network determined by interaction pathway analysis. Conclusion: In this study, we identified a gene-expression pathway of delayed neuronal recovery in military personnel a blast-TBI and chronic symptoms. Future work is needed to determine if therapeutic agents that regulate these pathways may provide novel treatments for chronic blast-TBI-related symptoms.
Collapse
Affiliation(s)
- Morgan Heinzelmann
- National Institute of Nursing Research, National Institutes of Health , Bethesda, MD , USA
| | - Swarnalatha Y Reddy
- National Institute of Nursing Research, National Institutes of Health , Bethesda, MD , USA
| | - Louis M French
- Center for Neuroscience and Regenerative Medicine , Bethesda, MD , USA ; Defense and Veterans Brain Injury Center, Walter Reed National Military Medical Center , Bethesda, MD , USA
| | - Dan Wang
- National Institute of Nursing Research, National Institutes of Health , Bethesda, MD , USA
| | - Hyunhwa Lee
- National Institute of Nursing Research, National Institutes of Health , Bethesda, MD , USA
| | - Taura Barr
- West Virginia University Health Sciences Center , Morgantown, WV , USA
| | - Tristin Baxter
- Sleep Medicine Clinic, Madigan Army Medical Center , Tacoma, WA , USA
| | - Vincent Mysliwiec
- Sleep Medicine Clinic, Madigan Army Medical Center , Tacoma, WA , USA
| | - Jessica Gill
- National Institute of Nursing Research, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|