1
|
Estrella LD, Manganaro JE, Sheldon L, Roland N, Snyder AD, George JW, Emanuel K, Lamberty BG, Stauch KL. Chronic glial activation and behavioral alterations induced by acute/subacute pioglitazone treatment in a mouse model of traumatic brain injury. Brain Behav Immun 2025; 123:64-80. [PMID: 39242055 DOI: 10.1016/j.bbi.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Traumatic brain injury (TBI) is a disabling neurotraumatic condition and the leading cause of injury-related deaths and disability in the United States. Attenuation of neuroinflammation early after TBI is considered an important treatment target; however, while these inflammatory responses can induce secondary brain injury, they are also involved in the repair of the nervous system. Pioglitazone, which activates peroxisome proliferator-activated receptor gamma, has been shown to decrease inflammation acutely after TBI, but the long-term consequences of its use remain unknown. For this reason, the impacts of treatment with pioglitazone during the acute/subacute phase (30 min after injury and each subsequent 24 h for 5 days) after TBI were interrogated during the chronic phase (30- and 274-days post-injury (DPI)) in mice using the controlled cortical impact model of experimental TBI. Acute/subacute pioglitazone treatment after TBI results in long-term deleterious consequences, including disruption of tau homeostasis, chronic glial cell activation, neuronal pathology, and worsened injury severity particularly at 274 DPI, with male mice being more susceptible than female mice. Further, male pioglitazone-treated TBI mice exhibited increased dominant and offensive-like behavior while having a decreased non-social exploring behavior at 274 DPI. After TBI, both sexes exhibited glial activation at 30 DPI when treated with pioglitazone; however, while injury severity was increased in females it was not impacted in male mice. This work reveals that although pioglitazone has been shown to lead to attenuated TBI outcomes acutely, sex-based differences, timing and long-term consequences of treatment with glitazones must be considered and further studied prior to their clinical use for TBI therapy.
Collapse
Affiliation(s)
- L Daniel Estrella
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Jane E Manganaro
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Lexi Sheldon
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Nashanthea Roland
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Austin D Snyder
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Joseph W George
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Katy Emanuel
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Benjamin G Lamberty
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Kelly L Stauch
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA.
| |
Collapse
|
2
|
Kursancew ACS, Faller CJ, Bortoluzzi DP, Niero LB, Brandão B, Danielski LG, Petronilho F, Generoso JS. Neuroinflammatory Response in the Traumatic Brain Injury: An Update. Neurochem Res 2024; 50:64. [PMID: 39718667 DOI: 10.1007/s11064-024-04316-4] [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: 06/18/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024]
Abstract
The central nervous system (CNS) comprises membranes and barriers that are vital to brain homeostasis. Membranes form a robust shield around neural structures, ensuring protection and structural integrity. At the same time, barriers selectively regulate the exchange of substances between blood and brain tissue, which is essential for maintaining homeostasis. Another highlight is the glymphatic system, which cleans metabolites and waste from the brain. Traumatic brain injury (TBI) represents a significant cause of disability and mortality worldwide, resulting from the application of direct mechanical force to the head that results in a primary injury. Therefore, this review aims to elucidate the mechanisms associated with the secondary injury cascade, in which there is intense activation of glial cells, dysfunction of the glymphatic system, glutamatergic neurotoxicity, additional molecular and biochemical changes that lead to a neuroinflammatory process, and oxidative stress and in which way they can be associated with cognitive damage that is capable of lasting for an extended period.
Collapse
Affiliation(s)
- Amanda C S Kursancew
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Cristiano Julio Faller
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Daniel Paulo Bortoluzzi
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Luana Budny Niero
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Beatriz Brandão
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Lucineia Gainski Danielski
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Jaqueline S Generoso
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
3
|
Allende Labastida J, Motamedi M, Wu P, Szczesny B. Protocol for inducing varying TBI severity in a mouse model using a closed-head, weight-drop, impact-induced acceleration mechanism. STAR Protoc 2024; 5:103370. [PMID: 39392749 PMCID: PMC11736045 DOI: 10.1016/j.xpro.2024.103370] [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: 04/16/2024] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/13/2024] Open
Abstract
Animal models of traumatic brain injury (TBI) are critical for understanding its complex neuropathology. Here, we present a protocol to induce varying TBI severities in mice using a closed-head, weight-drop model that includes an impact-induced acceleration mechanism. We describe steps for habituation with neurological severity score (NSS) equipment, assessing NSS baseline, performing anesthesia and TBI, assessing NSS post-injury, and analyzing data. This protocol requires no prior surgical intervention and is adaptable for rat studies. For complete details on the use and execution of this protocol, please refer to PhD Dissertation of Javier Allende Labastida1 and Tang et al.2.
Collapse
Affiliation(s)
- Javier Allende Labastida
- Departments of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
| | - Massoud Motamedi
- Departments of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
| | - Ping Wu
- Department of Neurobiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
| | - Bartosz Szczesny
- Departments of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA; Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA.
| |
Collapse
|
4
|
Wofford KL, Browne KD, Loane DJ, Meaney DF, Cullen DK. Peripheral immune cell dysregulation following diffuse traumatic brain injury in pigs. J Neuroinflammation 2024; 21:324. [PMID: 39696519 DOI: 10.1186/s12974-024-03317-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
Traumatic brain injury (TBI) is a global health problem affecting millions of individuals annually, potentially resulting in persistent neuropathology, chronic neurological deficits, and death. However, TBI not only affects neural tissue, but also affects the peripheral immune system's homeostasis and physiology. TBI disrupts the balanced signaling between the brain and the peripheral organs, resulting in immunodysregulation and increasing infection susceptibility. Indeed, secondary infections following TBI worsen neurological outcomes and are a major source of mortality and morbidity. Despite the compelling link between the damaged brain and peripheral immune functionality, little is known about how injury severity affects the peripheral immune system in closed-head diffuse TBI, the most common clinical presentation including all concussions. Therefore, we characterized peripheral blood mononuclear cells (PBMCs) and plasma changes over time and across injury severity using an established large-animal TBI model of closed-head, non-impact diffuse rotational acceleration in pigs. Across all timepoints and injury levels, we did not detect any changes to plasma cytokine concentrations. However, changes to the PBMCs were detectable and much more robust. We observed the concentration and physiology of circulating PBMCs changed in an injury severity-dependent manner, with most cellular changes occurring within the first 10 days following a high rotational velocity injury. Here, we report changes in the concentrations of myeloid and T cells, changes in PBMC composition, and changes in phagocytic clearance over time. Together, these data suggest that following a diffuse brain injury in a clinically relevant large-animal TBI model, the immune system exhibits perturbations that are detectable into the subacute timeframe. These findings invite future investigations into therapeutic interventions targeting peripheral immunity and the potential for peripheral blood cellular characterization as a diagnostic tool.
Collapse
Affiliation(s)
- Kathryn L Wofford
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105 Hayden Hall, 3320 Smith Walk, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Kevin D Browne
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105 Hayden Hall, 3320 Smith Walk, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - D Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105 Hayden Hall, 3320 Smith Walk, Philadelphia, PA, 19104, USA.
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA.
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
Joseph CR. Assessing Mild Traumatic Brain Injury-Associated Blood-Brain Barrier (BBB) Damage and Restoration Using Late-Phase Perfusion Analysis by 3D ASL MRI: Implications for Predicting Progressive Brain Injury in a Focused Review. Int J Mol Sci 2024; 25:11522. [PMID: 39519073 PMCID: PMC11547134 DOI: 10.3390/ijms252111522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Mild traumatic brain injury (mTBI) is a common occurrence around the world, associated with a variety of blunt force and torsion injuries affecting all age groups. Most never reach medical attention, and the identification of acute injury and later clearance to return to usual activities is relegated to clinical evaluation-particularly in sports injuries. Advanced structural imaging is rarely performed due to the usual absence of associated acute anatomic/hemorrhagic changes. This review targets physiologic imaging techniques available to identify subtle blood-brain barrier dysfunction and white matter tract shear injury and their association with chronic traumatic encephalopathy. These techniques provide needed objective measures to assure recovery from injury in those patients with persistent cognitive/emotional symptoms and in the face of repetitive mTBI.
Collapse
Affiliation(s)
- Charles R Joseph
- Department of Neurology and Internal Medicine, College of Osteopathic Medicine, Liberty University, Lynchburg, VA 24502, USA
| |
Collapse
|
6
|
Gotoh M, Dezawa S, Takashima I, Yamamoto S. Effects of focal cortical cooling on somatosensory evoked potentials in rats. Brain Res 2024; 1840:148995. [PMID: 38735427 DOI: 10.1016/j.brainres.2024.148995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
Although the focal brain cooling technique is widely used to examine brain function, the effects of cortical temperature at various levels on sensory information processing and neural mechanisms remain underexplored. To elucidate the mechanisms of temperature modulation in somatosensory processing, this study aimed to examine how P1 and N1 deflections of somatosensory evoked potentials (SEPs) depend on cortical temperature and how excitatory and inhibitory inputs contribute to this temperature dependency. SEPs were generated through electrical stimulation of the contralateral forepaw in anesthetized rats. The SEPs were recorded while cortical temperatures were altered between 17-38 °C either without any antagonists, with a gamma-aminobutyric acid type A (GABAA) receptor antagonist (gabazine), with an aminomethylphosphonic acid (AMPA) receptor antagonist (NBQX), or with an N-Methyl-D-aspartic acid (NMDA) receptor antagonist ([R]-CPP). The effects of different gabazine concentrations (0, 1, and 10 µM) were examined in the 35-38 °C range. The P1/N1 amplitudes and their peak-to-peak differences plotted against cortical temperature showed an inverted U relationship with a maximum at approximately 27.5 °C when no antagonists were administered. The negative correlation between these amplitudes and temperatures of ≥ 27.5 °C plateaued after gabazine administration, which occurred progressively as the gabazine concentration increased. In contrast, the correlation remained negative after the administration of NBQX and (R)-CPP. These results suggest that GABAergic inhibitory inputs contribute to the negative correlation between SEP amplitude and cortical temperature around the physiological cortical temperature.
Collapse
Affiliation(s)
- Mizuho Gotoh
- Department of Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba Japan; Department of Rehabilitation for Brain Functions, Research Institute of National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Shinnosuke Dezawa
- Department of Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba Japan; Faculty of Medical and Health Sciences, Tsukuba International University, Tsuchiura, Japan
| | - Ichiro Takashima
- Department of Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba Japan; Department of Information, Artificial Intelligence and Data Science, Daiichi Institute of Technology, Tokyo, Japan
| | - Shinya Yamamoto
- Department of Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba Japan.
| |
Collapse
|
7
|
Hiskens MI, Schneiders AG, Fenning AS. Selective COX-2 Inhibitors as Neuroprotective Agents in Traumatic Brain Injury. Biomedicines 2024; 12:1930. [PMID: 39200394 PMCID: PMC11352079 DOI: 10.3390/biomedicines12081930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/31/2024] [Accepted: 08/21/2024] [Indexed: 09/02/2024] Open
Abstract
Traumatic brain injury (TBI) is a significant contributor to mortality and morbidity in people, both young and old. There are currently no approved therapeutic interventions for TBI. Following TBI, cyclooxygenase (COX) enzymes generate prostaglandins and reactive oxygen species that perpetuate inflammation, with COX-1 and COX-2 isoforms providing differing responses. Selective COX-2 inhibitors have shown potential as neuroprotective agents. Results from animal models of TBI suggest potential treatment through the alleviation of secondary injury mechanisms involving neuroinflammation and neuronal cell death. Additionally, early clinical trials have shown that the use of celecoxib improves patient mortality and outcomes. This review aims to summarize the therapeutic effects of COX-2 inhibitors observed in TBI animal models, highlighting pertinent studies elucidating molecular pathways and expounding upon their mechanistic actions. We then investigated the current state of evidence for the utilization of COX-2 inhibitors for TBI patients.
Collapse
Affiliation(s)
- Matthew I. Hiskens
- Mackay Institute of Research and Innovation, Mackay Hospital and Health Service, Mackay, QLD 4740, Australia
| | - Anthony G. Schneiders
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia (A.S.F.)
| | - Andrew S. Fenning
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia (A.S.F.)
| |
Collapse
|
8
|
Feng D, Liu T, Zhang X, Xiang T, Su W, Quan W, Jiang R. Fingolimod improves diffuse brain injury by promoting AQP4 polarization and functional recovery of the glymphatic system. CNS Neurosci Ther 2024; 30:e14669. [PMID: 38459666 PMCID: PMC10924110 DOI: 10.1111/cns.14669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/26/2024] [Accepted: 02/17/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Diffuse brain injury (DBI) models are characterized by intense global brain inflammation and edema, which characterize the most severe form of TBI. In a previous experiment, we found that fingolimod promoted recovery after controlled cortical impact injury (CCI) by modulating inflammation around brain lesions. However, it remains unclear whether fingolimod can also attenuate DBI because of its different injury mechanisms. Furthermore, whether fingolimod has additional underlying effects on repairing DBI is unknown. METHODS The impact acceleration model of DBI was established in adult Sprague-Dawley rats. Fingolimod (0.5 mg/kg) was administered 0.5, 24, and 48 h after injury for 3 consecutive days. Immunohistochemistry, immunofluorescence analysis, cytokine array, and western blotting were used to evaluate inflammatory cells, inflammatory factors, AQP4 polarization, apoptosis in brain cells, and the accumulation of APP after DBI in rats. To evaluate the function of the glymphatic system (GS), a fluorescent tracer was injected into the cistern. The neural function of rats with DBI was evaluated using various tests, including the modified neurological severity score (mNSS), horizontal ladder-crossing test, beam walking test, and tape sensing and removal test. Brain water content was also measured. RESULTS Fingolimod administration for 3 consecutive days could reduce the levels of inflammatory cytokines, neutrophil recruitment, microglia, and astrocyte activation in the brain following DBI. Moreover, fingolimod reduced apoptotic protein expression, brain cell apoptosis, brain edema, and APP accumulation. Additionally, fingolimod inhibited the loss of AQP4 polarization, improved lymphatic system function, and reduced damage to nervous system function. Notably, inhibiting the GS weakened the therapeutic effect of fingolimod on the neurological function of rats with DBI and increased the accumulation of APP in the brain. CONCLUSIONS In brief, these findings suggest that fingolimod alleviates whole-brain inflammation and GS system damage after DBI and that inhibiting the GS could weaken the positive effect of fingolimod on nerve function in rats with DBI. Thus, inhibiting inflammation and regulating the GS may be critical for the therapeutic effect of fingolimod on DBI.
Collapse
Affiliation(s)
- Dongyi Feng
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Tao Liu
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Xinjie Zhang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Tangtang Xiang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Wanqiang Su
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Wei Quan
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Rongcai Jiang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| |
Collapse
|
9
|
Bahader GA, Naghavi F, Alotaibi A, Dehghan A, Swain CC, Burkett JP, Shah ZA. Neurobehavioral and inflammatory responses following traumatic brain injury in male and female mice. Behav Brain Res 2024; 456:114711. [PMID: 37827252 PMCID: PMC10615863 DOI: 10.1016/j.bbr.2023.114711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and is associated with a high rate of functional comorbidities, including motor, cognitive, anxiety, depression, and emotional disorders. TBI pathophysiology and recovery are complicated and involve several mechanistic pathways that control neurobehavioral outcomes. In this study, male and female C57Bl/6 J mice were subjected to a controlled cortical impact model of TBI or sham injury and evaluated for different neurobehavioral and inflammatory outcomes over a month. We demonstrate that TBI mice have increased motor dysfunction at early and late time points following the injury as compared to the sham group. Anxiety-like symptoms were time- and task-dependent, with both sexes having increased anxiety-like behavior 2 weeks post-injury. Cognitive functions measured by T-maze presented greater deficits in TBI mice, while there was no sex or injury-related difference in depressive-like behaviors. Notably, a significant effect of sex was found in empathy-like behavior, with females showing more allogrooming and freezing behavior in the consoling and fear observational tests, respectively. Evaluating the impact of the injury-induced brain damage demonstrated a greater injury volume and neuronal degeneration in males compared to females one month after TBI. Moreover, male mice showed higher peripheral inflammatory responses, as represented by elevated serum levels of peripheral leukocytes and inflammatory markers. These results will have significant implications for understanding TBI's long-term consequences on neurobehavioral and inflammatory responses, which are sex-specific and can be considered for individualized therapeutic strategies in treating TBI.
Collapse
Affiliation(s)
- Ghaith A Bahader
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Farzaneh Naghavi
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Ahmed Alotaibi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Amir Dehghan
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Caroline C Swain
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - James P Burkett
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|
10
|
Freeman-Jones E, Miller WH, Work LM, Fullerton JL. Polypathologies and Animal Models of Traumatic Brain Injury. Brain Sci 2023; 13:1709. [PMID: 38137157 PMCID: PMC10741988 DOI: 10.3390/brainsci13121709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Traumatic brain injury (TBI) is an important health issue for the worldwide population, as it causes long-term pathological consequences for a diverse group of individuals. We are yet to fully elucidate the significance of TBI polypathologies, such as neuroinflammation and tau hyperphosphorylation, and their contribution to the development of chronic traumatic encephalopathy (CTE) and other neurological conditions. To advance our understanding of TBI, it is necessary to replicate TBI in preclinical models. Commonly used animal models include the weight drop model; these methods model human TBI in various ways and in different animal species. However, animal models have not demonstrated their clinical utility for identifying therapeutic interventions. Many interventions that were successful in improving outcomes for animal models did not translate into clinical benefit for patients. It is important to review current animal models and discuss their strengths and limitations within a TBI context. Modelling human TBI in animals encounters numerous challenges, yet despite these barriers, the TBI research community is working to overcome these difficulties. Developments include advances in biomarkers, standardising, and refining existing models. This progress will improve our ability to model TBI in animals and, therefore, enhance our understanding of TBI and, potentially, how to treat it.
Collapse
Affiliation(s)
- Erin Freeman-Jones
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow G12 8QQ, UK; (E.F.-J.); (W.H.M.)
| | - William H. Miller
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow G12 8QQ, UK; (E.F.-J.); (W.H.M.)
| | - Lorraine M. Work
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK;
| | - Josie L. Fullerton
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK;
| |
Collapse
|
11
|
Ge M, Wang Y, Wu T, Li H, Yang C, Chen T, Feng H, Xu D, Yao J. Serum-based Raman spectroscopic diagnosis of blast-induced brain injury in a rat model. BIOMEDICAL OPTICS EXPRESS 2023; 14:3622-3634. [PMID: 37497497 PMCID: PMC10368048 DOI: 10.1364/boe.495285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 07/28/2023]
Abstract
The diagnosis of blast-induced traumatic brain injury (bTBI) is of paramount importance for early care and clinical therapy. Therefore, the rapid diagnosis of bTBI is vital to the treatment and prognosis in clinic. In this paper, we reported a new strategy for label-free bTBI diagnosis through serum-based Raman spectroscopy. The Raman spectral characteristics of serum in rat were investigated at 3 h, 24 h, 48 h and 72 h after mild and moderate bTBIs. It has been demonstrated that both the position and intensity of Raman characteristic peaks exhibited apparent differences in the range of 800-3000cm-1 compared with control group. It could be inferred that the content, structure and interaction of biomolecules in the serum were changed after blast exposure, which might help to understand the neurological syndromes caused by bTBI. Furthermore, the control group, mild and moderate bTBIs at different times (a total of 9 groups) were automatically classified by combining principal component analysis and four machine learning algorithms (quadratic discriminant analysis, support vector machine, k-nearest neighbor, neural network). The highest classification accuracy, sensitivity and precision were up to 95.4%, 95.9% and 95.7%. It is suggested that this method has great potential for high-sensitive, rapid, and label-free diagnosis of bTBI.
Collapse
Affiliation(s)
- Meilan Ge
- School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Yuye Wang
- School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Tong Wu
- School of Marine Science and Technology, Tianjin University, Tianjin 300072, China
| | - Haibin Li
- School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Chuanyan Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tunan Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Degang Xu
- School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Jianquan Yao
- School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| |
Collapse
|
12
|
Govindarajulu M, Patel MY, Wilder DM, Krishnan J, LaValle C, Pandya JD, Shear DA, Hefeneider SH, Long JB, Arun P. Upregulation of multiple toll-like receptors in ferret brain after blast exposure: potential targets for treatment. Neurosci Lett 2023; 810:137364. [PMID: 37391063 DOI: 10.1016/j.neulet.2023.137364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Although blast-induced traumatic brain injury (bTBI) has been designated as the signature injury of recent combat operations, its precise pathological mechanism(s) has not been identified thus far. Prior preclinical studies on bTBI demonstrated acute neuroinflammatory cascades which are known to be contributing to neurodegeneration. Danger-associated chemical patterns are released from the injured cells, which activate non-specific pattern recognition receptors, such as toll-like receptors (TLRs) leading to increased expression of inflammatory genes and release of cytokines. Upregulation of specific TLRs in the brain has been described as a mechanism of injury in diverse brain injury models unrelated to blast exposure. However, the expression profile of various TLRs in bTBI has not been investigated thus far. Hence, we have evaluated the expression of transcripts for TLR1-TLR10 in the brain of a gyrencephalic animal model of bTBI. We exposed ferrets to tightly coupled repeated blasts and determined the differential expression of TLRs (TLR1-10) by quantitative RT-PCR in multiple brain regions at 4 hr, 24 hr, 7 days and 28 days post-blast injury. The results obtained indicate that multiple TLRs are upregulated in the brain at 4 hr, 24 hr, 7 days and 28 days post-blast. Specifically, upregulation of TLR2, TLR4 and TLR9 was noted in different brain regions, suggesting that multiple TLRs might play a role in the pathophysiology of bTBI and that drugs that can inhibit multiple TLRs might have enhanced efficacy to attenuate brain damage and thereby improve bTBI outcome. Taken together, these results suggest that several TLRs are upregulated in the brain after bTBI and participate in the inflammatory response and thereby provide new insights into the disease pathogenesis. Therefore, inhibition of multiple TLRs, including TLR2, 4 and 9, simultaneously might be a potential therapeutic strategy for the treatment of bTBI.
Collapse
Affiliation(s)
- Manoj Govindarajulu
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Mital Y Patel
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Donna M Wilder
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jishnu Krishnan
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Christina LaValle
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jignesh D Pandya
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Deborah A Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | - Joseph B Long
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Peethambaran Arun
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
13
|
Deshetty UM, Periyasamy P. Potential Biomarkers in Experimental Animal Models for Traumatic Brain Injury. J Clin Med 2023; 12:3923. [PMID: 37373618 DOI: 10.3390/jcm12123923] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a complex and multifaceted disorder that has become a significant public health concern worldwide due to its contribution to mortality and morbidity. This condition encompasses a spectrum of injuries, including axonal damage, contusions, edema, and hemorrhage. Unfortunately, specific effective therapeutic interventions to improve patient outcomes following TBI are currently lacking. Various experimental animal models have been developed to mimic TBI and evaluate potential therapeutic agents to address this issue. These models are designed to recapitulate different biomarkers and mechanisms involved in TBI. However, due to the heterogeneous nature of clinical TBI, no single experimental animal model can effectively mimic all aspects of human TBI. Accurate emulation of clinical TBI mechanisms is also tricky due to ethical considerations. Therefore, the continued study of TBI mechanisms and biomarkers, of the duration and severity of brain injury, treatment strategies, and animal model optimization is necessary. This review focuses on the pathophysiology of TBI, available experimental TBI animal models, and the range of biomarkers and detection methods for TBI. Overall, this review highlights the need for further research to improve patient outcomes and reduce the global burden of TBI.
Collapse
Affiliation(s)
- Uma Maheswari Deshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
14
|
Burns JM, Kalinosky BT, Sloan MA, Cerna CZ, Fines DA, Valdez CM, Voorhees WB. Dilation of the superior sagittal sinus detected in rat model of mild traumatic brain injury using 1 T magnetic resonance imaging. Front Neurol 2023; 14:1045695. [PMID: 37181576 PMCID: PMC10169716 DOI: 10.3389/fneur.2023.1045695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 03/23/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Mild traumatic brain injury (mTBI) is a common injury that can lead to temporary and, in some cases, life-long disability. Magnetic resonance imaging (MRI) is widely used to diagnose and study brain injuries and diseases, yet mTBI remains notoriously difficult to detect in structural MRI. mTBI is thought to be caused by microstructural or physiological changes in the function of the brain that cannot be adequately captured in structural imaging of the gray and white matter. However, structural MRIs may be useful in detecting significant changes in the cerebral vascular system (e.g., the blood-brain barrier (BBB), major blood vessels, and sinuses) and the ventricular system, and these changes may even be detectable in images taken by low magnetic field strength MRI scanners (<1.5T). Methods In this study, we induced a model of mTBI in the anesthetized rat animal model using a commonly used linear acceleration drop-weight technique. Using a 1T MRI scanner, the brain of the rat was imaged, without and with contrast, before and after mTBI on post-injury days 1, 2, 7, and 14 (i.e., P1, P2, P7, and P14). Results Voxel-based analyses of MRIs showed time-dependent, statistically significant T2-weighted signal hypointensities in the superior sagittal sinus (SSS) and hyperintensities of the gadolinium-enhanced T1-weighted signal in the superior subarachnoid space (SA) and blood vessels near the dorsal third ventricle. These results showed a widening, or vasodilation, of the SSS on P1 and of the SA on P1-2 on the dorsal surface of the cortex near the site of the drop-weight impact. The results also showed vasodilation of vasculature near the dorsal third ventricle and basal forebrain on P1-7. Discussion Vasodilation of the SSS and SA near the site of impact could be explained by the direct mechanical injury resulting in local changes in tissue function, oxygenation, inflammation, and blood flow dynamics. Our results agreed with literature and show that the 1T MRI scanner performs at a level comparable to higher field strength scanners for this type of research.
Collapse
Affiliation(s)
- Jennie M. Burns
- General Dynamics Information Technology, Defense Division, JBSA Fort SamHouston, TX, United States
| | - Benjamin T. Kalinosky
- General Dynamics Information Technology, Defense Division, JBSA Fort SamHouston, TX, United States
| | - Mark A. Sloan
- General Dynamics Information Technology, Defense Division, JBSA Fort SamHouston, TX, United States
| | - Cesario Z. Cerna
- General Dynamics Information Technology, Defense Division, JBSA Fort SamHouston, TX, United States
| | - David A. Fines
- General Dynamics Information Technology, Defense Division, JBSA Fort SamHouston, TX, United States
| | - Christopher M. Valdez
- Radio Frequency Bioeffects Branch, Bioeffects Division, Airman Systems Directorate, 711th Human Performance Wing, Air Force Research Laboratory, JBSA FortSam Houston, TX, United States
| | - William B. Voorhees
- Radio Frequency Bioeffects Branch, Bioeffects Division, Airman Systems Directorate, 711th Human Performance Wing, Air Force Research Laboratory, JBSA FortSam Houston, TX, United States
| |
Collapse
|
15
|
Barker S, Paul BD, Pieper AA. Increased Risk of Aging-Related Neurodegenerative Disease after Traumatic Brain Injury. Biomedicines 2023; 11:1154. [PMID: 37189772 PMCID: PMC10135798 DOI: 10.3390/biomedicines11041154] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/30/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Traumatic brain injury (TBI) survivors frequently suffer from chronically progressive complications, including significantly increased risk of developing aging-related neurodegenerative disease. As advances in neurocritical care increase the number of TBI survivors, the impact and awareness of this problem are growing. The mechanisms by which TBI increases the risk of developing aging-related neurodegenerative disease, however, are not completely understood. As a result, there are no protective treatments for patients. Here, we review the current literature surrounding the epidemiology and potential mechanistic relationships between brain injury and aging-related neurodegenerative disease. In addition to increasing the risk for developing all forms of dementia, the most prominent aging-related neurodegenerative conditions that are accelerated by TBI are amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Parkinson's disease (PD), and Alzheimer's disease (AD), with ALS and FTD being the least well-established. Mechanistic links between TBI and all forms of dementia that are reviewed include oxidative stress, dysregulated proteostasis, and neuroinflammation. Disease-specific mechanistic links with TBI that are reviewed include TAR DNA binding protein 43 and motor cortex lesions in ALS and FTD; alpha-synuclein, dopaminergic cell death, and synergistic toxin exposure in PD; and brain insulin resistance, amyloid beta pathology, and tau pathology in AD. While compelling mechanistic links have been identified, significantly expanded investigation in the field is needed to develop therapies to protect TBI survivors from the increased risk of aging-related neurodegenerative disease.
Collapse
Affiliation(s)
- Sarah Barker
- Center for Brain Health Medicines, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Bindu D. Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21211, USA;
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21211, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21211, USA
- Lieber Institute for Brain Development, Baltimore, MD 21205, USA
| | - Andrew A. Pieper
- Center for Brain Health Medicines, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Translational Therapeutics Core, Cleveland Alzheimer’s Disease Research Center, Cleveland, OH 44106, USA
| |
Collapse
|
16
|
Traumatic Brain Injury Induces Microglial and Caspase3 Activation in the Retina. Int J Mol Sci 2023; 24:ijms24054451. [PMID: 36901880 PMCID: PMC10003323 DOI: 10.3390/ijms24054451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Traumatic brain injury (TBI) is among the main causes of sudden death after head trauma. These injuries can result in severe degeneration and neuronal cell death in the CNS, including the retina, which is a crucial part of the brain responsible for perceiving and transmitting visual information. The long-term effects of mild-repetitive TBI (rmTBI) are far less studied thus far, even though damage induced by repetitive injuries occurring in the brain is more common, especially amongst athletes. rmTBI can also have a detrimental effect on the retina and the pathophysiology of these injuries is likely to differ from severe TBI (sTBI) retinal injury. Here, we show how rmTBI and sTBI can differentially affect the retina. Our results indicate an increase in the number of activated microglial cells and Caspase3-positive cells in the retina in both traumatic models, suggesting a rise in the level of inflammation and cell death after TBI. The pattern of microglial activation appears distributed and widespread but differs amongst the various retinal layers. sTBI induced microglial activation in both the superficial and deep retinal layers. In contrast to sTBI, no significant change occurred following the repetitive mild injury in the superficial layer, only the deep layer (spanning from the inner nuclear layer to the outer plexiform layer) shows microglial activation. This difference suggests that alternate response mechanisms play a role in the case of the different TBI incidents. The Caspase3 activation pattern showed a uniform increase in both the superficial and deep layers of the retina. This suggests a different action in the course of the disease in sTBI and rmTBI models and points to the need for new diagnostic procedures. Our present results suggest that the retina might serve as such a model of head injuries since the retinal tissue reacts to both forms of TBI and is the most accessible part of the human brain.
Collapse
|
17
|
Donaldson RI, Buchanan OJ, Graham TL, Ross JD. Development of a Novel Epidural Hemorrhage Model in Swine. Mil Med 2023; 188:20-26. [PMID: 34676417 DOI: 10.1093/milmed/usab427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/20/2021] [Accepted: 10/07/2021] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Traumatic brain injury is a major public health concern. Among patients with severe traumatic brain injury, epidural hemorrhage is known to swiftly lead to brain herniation and death unless there is emergent neurosurgical intervention. However, immediate neurosurgeon availability is frequently a problem outside of level I trauma centers. In this context, the authors desired to test a novel device for the emergent management of life-threatening epidural hemorrhage. A review of existing animal models determined that all were inadequate for this purpose, as they were found to be either inappropriate or obsolete. Here, we describe the development of a new epidural hemorrhage model in swine (Sus scrofa, 18-26 kg) ideal for translational device testing. MATERIALS AND METHODS Vascular access was achieved using an ultrasound-guided percutaneous Seldinger catheter-over-wire technique with 5 Fr catheters placed in the bilateral carotid arteries, for continuous blood pressure and to allow for withdrawal of blood for creation of epidural hemorrhage. To simulate an actively bleeding and life-threatening epidural hemorrhage, unadulterated autologous blood was infused from the vascular access point into the epidural space. To be useful for this application and clinical scenario, brain death needed to occur after the planned intervention time but before the end of the protocol period (if no intervention took place). An iterative approach to model development determined that this could be achieved with an initial infusion rate of 1.0 mL/min, slowed to 0.5 mL/min after the first 10 minutes, paired with an intervention time at 15 minutes. All experiments were performed at Oregon Health & Science University, an Association for Assessment and Accreditation of Laboratory Animal Care accredited facility. Oregon Health & Science University's Institutional Animal Care and Use Committee, as well as the United States Army Animal Care and Use Review Office, reviewed and approved this protocol before the initiation of experiments (respectively, protocol numbers IP00002901 and 18116010.e001). RESULTS The final developed model allows for the infusion of a known volume of autologous, unadulterated blood directly into the epidural space, without the use of a balloon or other restricting membranes, and is rapidly fatal in the absence of intervention. CONCLUSIONS This animal model is the first to mirror the expected clinical course of epidural hemorrhage in a physiologically relevant manner, while allowing translational testing of emergency devices. This model successfully allowed the initial testing of a novel interventional device for the emergent management of epidural hemorrhage that was designed for use in the absence of traditional neurosurgical capabilities.
Collapse
Affiliation(s)
- Ross I Donaldson
- Critical Innovations LLC, Los Angeles, CA 90302, USA.,Department of Emergency Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.,Department of Emergency Medicine, Harbor-UCLA Medical Center, Torrance, CA 90509, USA.,Department of Epidemiology, UCLA-Fielding School of Public Health, Los Angeles, CA 90095, USA
| | | | - Todd L Graham
- Military & Health Research Foundation, Laurel, MD 20723, USA.,Charles T Dotter Department of Interventional Radiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - James D Ross
- Military & Health Research Foundation, Laurel, MD 20723, USA.,Charles T Dotter Department of Interventional Radiology, Oregon Health & Science University, Portland, OR 97239, USA.,Center for Regenerative Medicine, Oregon Health & Science University School of Medicine, Portland, OR 97239, USA
| |
Collapse
|
18
|
Development of a Weight Drop Injury Device Suitable for Blunt, Closed-Head Injury Using a Rodent Model. BIOMEDICAL ENGINEERING ADVANCES 2022. [DOI: 10.1016/j.bea.2022.100067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
19
|
Song H, McEwan PP, Ameen-Ali KE, Tomasevich A, Kennedy-Dietrich C, Palma A, Arroyo EJ, Dolle JP, Johnson VE, Stewart W, Smith DH. Concussion leads to widespread axonal sodium channel loss and disruption of the node of Ranvier. Acta Neuropathol 2022; 144:967-985. [PMID: 36107227 PMCID: PMC9547928 DOI: 10.1007/s00401-022-02498-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 01/26/2023]
Abstract
Despite being a major health concern, little is known about the pathophysiological changes that underly concussion. Nonetheless, emerging evidence suggests that selective damage to white matter axons, or diffuse axonal injury (DAI), disrupts brain network connectivity and function. While voltage-gated sodium channels (NaChs) and their anchoring proteins at the nodes of Ranvier (NOR) on axons are key elements of the brain's network signaling machinery, changes in their integrity have not been studied in context with DAI. Here, we utilized a clinically relevant swine model of concussion that induces evolving axonal pathology, demonstrated by accumulation of amyloid precursor protein (APP) across the white matter. Over a two-week follow-up post-concussion with this model, we found widespread loss of NaCh isoform 1.6 (Nav1.6), progressive increases in NOR length, the appearance of void and heminodes and loss of βIV-spectrin, ankyrin G, and neurofascin 186 or their collective diffusion into the paranode. Notably, these changes were in close proximity, yet distinct from APP-immunoreactive swollen axonal profiles, potentially representing a unique, newfound phenotype of axonal pathology in DAI. Since concussion in humans is non-fatal, the clinical relevance of these findings was determined through examination of post-mortem brain tissue from humans with higher levels of acute traumatic brain injury. Here, a similar loss of Nav1.6 and changes in NOR structures in brain white matter were observed as found in the swine model of concussion. Collectively, this widespread and progressive disruption of NaChs and NOR appears to be a form of sodium channelopathy, which may represent an important substrate underlying brain network dysfunction after concussion.
Collapse
Affiliation(s)
- Hailong Song
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Przemyslaw P McEwan
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Kamar E Ameen-Ali
- School of Neuroscience and Psychology, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Alexandra Tomasevich
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | | | - Alexander Palma
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Edgardo J Arroyo
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Jean-Pierre Dolle
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Victoria E Johnson
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - William Stewart
- School of Neuroscience and Psychology, University of Glasgow, Glasgow, G12 8QQ, UK
- Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, G51 4TF, UK
| | - Douglas H Smith
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA.
| |
Collapse
|
20
|
Kaya D, Micili SC, Kizmazoglu C, Mucuoglu AO, Buyukcoban S, Ersoy N, Yilmaz O, Isik AT. Allopurinol attenuates repeated traumatic brain injury in old rats: A preliminary report. Exp Neurol 2022; 357:114196. [PMID: 35931122 DOI: 10.1016/j.expneurol.2022.114196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/13/2022] [Accepted: 07/31/2022] [Indexed: 12/01/2022]
Abstract
Traumatic brain injury (TBI) is an overlooked cause of morbidity, which was shown to accelerate inflammation, oxidative stress, and neuronal cell loss and is associated with spatial learning and memory impairments and some psychiatric disturbances in older adults. However, there is no effective treatment in order to offer a favorable outcome encompassing a good recovery after TBI in older adults. Hence, the present study aimed to investigate the histological and neurobehavioral effects of Allopurinol (ALL) in older rats that received repeated TBI (rTBI). For this purpose, a weight-drop rTBI model was used on old male Wistar rats. Rats received 5 repeated TBI/sham injuries 24 h apart and were treated with saline or Allopurinol 100 mg/kg, i.p. each time. They were randomly assigned to three groups: control group (no injury); rTBI group (received 5 rTBI and treated with saline); rTBI+ALL group (received 5 rTBI and treated with Allopurinol). Then, half of the animals from each group were sacrificed on day 6 and the remaining animals were assessed with Open field, Elevated plus maze and Morris Water Maze test. Basic neurological tasks were evaluated with neurological assessment protocol every other day until after the 19th day from the last injury. Brain sections were processed for neuronal cell count in the hippocampus (CA1), dentate gyrus (DG), and prefrontal cortex (PC). Also, an immunohistochemical assay was performed to determine NeuN, iNOS, and TNFα levels in the brain regions. The number of neurons was markedly reduced in CA1, GD, and PC in rats receiving saline compared to those receiving allopurinol treatment. Immunohistochemical analysis showed marked induction of iNOS and TNFα expression in the brain tissues which were reduced after allopurinol at 6 and 19 days post-injury. Also, ALL-treated rats demonstrated a remarkable induce in NeuN expression, indicating a reduction in rTBI-induced neuronal cell death. In neurobehavioral analyses, time spent in closed arms, in the corner of the open field, swimming latency, and distance were impaired in injured rats; however, all of them were significantly improved by allopurinol therapy. To sum up, this study demonstrated that ALL may mitigate rTBI-induced damage in aged rats, which suggests ALL as a potential therapeutic strategy for the treatment of recurrent TBI.
Collapse
Affiliation(s)
- Derya Kaya
- Dokuz Eylul University Faculty of Medicine, Department of Geriatric Medicine, Unit for Brain Aging and Dementia, Izmir, Turkey; Geriatric Science Association, Izmir, Turkey.
| | - Serap Cilaker Micili
- Dokuz Eylul University Faculty of Medicine, Department of Histology and Embryology, Izmir, Turkey
| | - Ceren Kizmazoglu
- Dokuz Eylul University Faculty of Medicine, Department of Neurosurgery, Izmir, Turkey
| | - Ali Osman Mucuoglu
- Dokuz Eylul University Faculty of Medicine, Department of Neurosurgery, Izmir, Turkey
| | - Sibel Buyukcoban
- Dokuz Eylul University Faculty of Medicine, Department of Anaesthesiology and Reanimation, İzmir, Turkey
| | - Nevin Ersoy
- Dokuz Eylul University Faculty of Medicine, Department of Histology and Embryology, Izmir, Turkey
| | - Osman Yilmaz
- Dokuz Eylul University Health Sciences Institute, Department of Laboratory Animal Science, Izmir, Turkey
| | - Ahmet Turan Isik
- Dokuz Eylul University Faculty of Medicine, Department of Geriatric Medicine, Unit for Brain Aging and Dementia, Izmir, Turkey; Geriatric Science Association, Izmir, Turkey
| |
Collapse
|
21
|
Swanson RL, Acharya NK, Cifu DX. Cerebral Microvascular Pathology Is a Common Endophenotype Between Traumatic Brain Injury, Cardiovascular Disease, and Dementia: A Hypothesis and Review. Cureus 2022; 14:e25318. [PMID: 35774720 PMCID: PMC9236636 DOI: 10.7759/cureus.25318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/05/2022] Open
Abstract
Traumatic brain injury (TBI) exposure has been associated with an increased risk of age-related cognitive decline or dementia in multiple epidemiological studies. Current therapeutic interventions in the field of Brain Injury Medicine focus largely on episodic symptom management during the chronic phase of TBI recovery, rather than targeting specific underlying pathological processes. This approach may be especially problematic for secondary age-related cognitive decline or dementia following TBI exposure. Although there are likely multiple pathophysiological mechanisms involved, a growing body of literature demonstrates that cerebral microvascular pathology is a common endophenotype across the spectrum of TBI severity. Similarly, a combination of pre-clinical and clinical research over the past two decades has implicated cerebral microvascular pathology in the initiation and progression of multiple neurodegenerative diseases, including Alzheimer’s disease and other dementias. We hypothesize that cerebral microvascular pathology is a common endophenotype between TBI, cardiovascular disease (CVD), and dementia, which can be targeted through modifiable cardiovascular risk factor reductions during the chronic phase of TBI recovery. We posit that our hypothesis is supported by the currently available scientific literature, as detailed in our review.
Collapse
|
22
|
Nidhi, Singh G, Valecha R, Shukla G, Kaushik D, Rahman MA, Gautam RK, Madan K, Mittal V, Singla RK. Neurobehavioral and Biochemical Evidences in Support of Protective Effect of Marrubiin (Furan Labdane Diterpene) from Marrubium vulgare Linn. and Its Extracts after Traumatic Brain Injury in Experimental Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:4457973. [PMID: 35656476 PMCID: PMC9155918 DOI: 10.1155/2022/4457973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 02/05/2023]
Abstract
Traumatic brain injuries due to sudden accidents cause major physical and mental health problems and are one of the main reasons behind the mortality and disability of patients. Research on alternate natural sources could be a boon for the rehabilitation of poor TBI patients. The literature indicates the Marrubium vulgare Linn. and its secondary metabolite marrubiin (furan labdane diterpene) possess various pharmacological properties such as vasorelaxant, calcium channel blocker, antioxidant, and antiedematogenic activities. Hence, in the present research, both marrubiin and hydroalcoholic extracts of the plant were evaluated for their neuroprotective effect after TBI. The neurological severity score and oxidative stress parameters are significantly altered by the test samples. Moreover, the neurotransmitter analysis indicated a significant change in GABA and glutamate. The histopathological study also supported the observed results. The improved neuroprotective potential of the extract could be attributed to the presence of a large number of secondary metabolites including marrubiin.
Collapse
Affiliation(s)
- Nidhi
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Rekha Valecha
- Department of Pharmacy, Delhi Skill and Entrepreneurship University, New Delhi 110065, India
| | - Govind Shukla
- University College of Ayurveda, Dr. S. R. Rajasthan Ayurveda University, Jodhpur 342304, India
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Mohammad Akhlaquer Rahman
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Rupesh K. Gautam
- Department of Pharmacology, MM School of Pharmacy, MM University, Sadopur-Ambala 134007, India
| | - Kumud Madan
- Lloyd Institute of Management and Technology (Pharm.), Greater Noida, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- iGlobal Research and Publishing Foundation, New Delhi 110059, India
| |
Collapse
|
23
|
Nwafor DC, Brichacek AL, Foster CH, Lucke-Wold BP, Ali A, Colantonio MA, Brown CM, Qaiser R. Pediatric Traumatic Brain Injury: An Update on Preclinical Models, Clinical Biomarkers, and the Implications of Cerebrovascular Dysfunction. J Cent Nerv Syst Dis 2022; 14:11795735221098125. [PMID: 35620529 PMCID: PMC9127876 DOI: 10.1177/11795735221098125] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 04/14/2022] [Indexed: 11/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of pediatric morbidity and mortality. Recent studies suggest that children and adolescents have worse post-TBI outcomes and take longer to recover than adults. However, the pathophysiology and progression of TBI in the pediatric population are studied to a far lesser extent compared to the adult population. Common causes of TBI in children are falls, sports/recreation-related injuries, non-accidental trauma, and motor vehicle-related injuries. A fundamental understanding of TBI pathophysiology is crucial in preventing long-term brain injury sequelae. Animal models of TBI have played an essential role in addressing the knowledge gaps relating to pTBI pathophysiology. Moreover, a better understanding of clinical biomarkers is crucial to diagnose pTBI and accurately predict long-term outcomes. This review examines the current preclinical models of pTBI, the implications of pTBI on the brain’s vasculature, and clinical pTBI biomarkers. Finally, we conclude the review by speculating on the emerging role of the gut-brain axis in pTBI pathophysiology.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- West Virginia University School of Medicine, Morgantown, WV, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Allison L. Brichacek
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Chase H. Foster
- Department of Neurosurgery, George Washington University Hospital, Washington D.C., USA
| | | | - Ahsan Ali
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | | | - Candice M. Brown
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Rabia Qaiser
- Department of Neurosurgery, Baylor Scott and White, Temple, TX, USA
| |
Collapse
|
24
|
Vinh To X, Mohamed AZ, Cumming P, Nasrallah FA. Subacute cytokine changes after a traumatic brain injury predict chronic brain microstructural alterations on advanced diffusion imaging in the male rat. Brain Behav Immun 2022; 102:137-150. [PMID: 35183698 DOI: 10.1016/j.bbi.2022.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The process of neuroinflammation occurring after traumatic brain injury (TBI) has received significant attention as a potential prognostic indicator and interventional target to improve patients' outcomes. Indeed, many of the secondary consequences of TBI have been attributed to neuroinflammation and peripheral inflammatory changes. However, inflammatory biomarkers in blood have not yet emerged as a clinical tool for diagnosis of TBI and predicting outcome. The controlled cortical impact model of TBI in the rodent gives reliable readouts of the dynamics of post-TBI neuroinflammation. We now extend this model to include a panel of plasma cytokine biomarkers measured at different time points post-injury, to test the hypothesis that these markers can predict brain microstructural outcome as quantified by advanced diffusion-weighted magnetic resonance imaging (MRI). METHODS Fourteen 8-10-week-old male rats were randomly assigned to sham surgery (n = 6) and TBI (n = 8) treatment with a single moderate-severe controlled cortical impact. We collected blood samples for cytokine analysis at days 1, 3, 7, and 60 post-surgery, and carried out standard structural and advanced diffusion-weighted MRI at day 60. We then utilized principal component regression to build an equation predicting different aspects of microstructural changes from the plasma inflammatory marker concentrations measured at different time points. RESULTS The TBI group had elevated plasma levels of IL-1β and several neuroprotective cytokines and chemokines (IL-7, CCL3, and GM-CSF) compared to the sham group from days 3 to 60 post-injury. The plasma marker panels obtained at day 7 were significantly associated with the outcome at day 60 of the trans-hemispheric cortical map transfer process that is a frequent finding in unilateral TBI models. DISCUSSION These results confirm and extend prior studies showing that day 7 post-injury is a critical temporal window for the reorganisation process following TBI. High plasma level of IL-1β and low plasma levels of the neuroprotective IL-7, CCL3, and GM-CSF of TBI animals at day 60 were associated with greater TBI pathology.
Collapse
Affiliation(s)
- Xuan Vinh To
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Abdalla Z Mohamed
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia; Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland; School of Psychology and Counselling, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Fatima A Nasrallah
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia; The Centre for Advanced Imaging, The University of Queensland, Queensland, Australia.
| |
Collapse
|
25
|
Jacotte-Simancas A, Middleton JW, Stielper ZF, Edwards S, Molina PE, Gilpin NW. Brain Injury Effects on Neuronal Activation and Synaptic Transmission in the Basolateral Amygdala of Adult Male and Female Wistar Rats. J Neurotrauma 2022; 39:544-559. [PMID: 35081744 PMCID: PMC8978566 DOI: 10.1089/neu.2021.0270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Traumatic brain injury (TBI) is defined as brain damage produced by an external mechanical force that leads to behavioral, cognitive, and psychiatric sequelae. The basolateral amygdala (BLA) is involved in emotional regulation, and its function and morphology are altered following TBI. Little is known about potential sex-specific effects of TBI on BLA neuronal function, but it is critical for the field to identify potential sex differences in TBI effects on brain and behavior. Here, we hypothesized that TBI would produce sex-specific acute (1 h) effects on BLA neuronal activation, excitability, and synaptic transmission in adult male and female rats. Forty-nine Wistar rats (n = 23 males and 26 females) were randomized to TBI (using lateral fluid percussion) or Sham groups in two separate studies. Study 1 used in situ hybridization (i.e., RNAscope) to measure BLA expression of c-fos (a marker of cell activation), vGlut, and vGat (markers of glutamatergic and GABAergic neurons, respectively) messenger RNA (mRNA). Study 2 used slice electrophysiology to measure intrinsic excitability and excitatory/inhibitory synaptic transmission in putative pyramidal neurons in the BLA. Physiological measures of injury severity were collected from all animals. Our results show that females exhibit increased apnea duration and reduced respiratory rate post-TBI relative to males. In male and female rats, TBI increased c-fos expression in BLA glutamatergic cells but not in BLA GABAergic cells, and TBI increased firing rate in BLA pyramidal neurons. Further, TBI increased spontaneous excitatory and inhibitory postsynaptic current (sEPSC and sIPSC) amplitude in BLA neurons of females relative to all other groups. TBI increased sEPSC frequency in BLA neurons of females relative to males but did not alter sIPSC frequency. In summary, lateral fluid percussion produced different physiological responses in male and female rats, as well as sex-specific alterations in BLA neuronal activation, excitability, and synaptic transmission 1 h after injury.
Collapse
Affiliation(s)
- Alejandra Jacotte-Simancas
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Jason W. Middleton
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Zachary F. Stielper
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Scott Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Patricia E. Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Nicholas W. Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Southeast Louisiana VA Healthcare System, New Orleans, Louisiana, USA
| |
Collapse
|
26
|
Hutchinson EB, Romero-Lozano A, Johnson HR, Knutsen AK, Bosomtwi A, Korotcov A, Shunmugavel A, King SG, Schwerin SC, Juliano SL, Dardzinski BJ, Pierpaoli C. Translationally Relevant Magnetic Resonance Imaging Markers in a Ferret Model of Closed Head Injury. Front Neurosci 2022; 15:779533. [PMID: 35280340 PMCID: PMC8904401 DOI: 10.3389/fnins.2021.779533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 12/17/2021] [Indexed: 11/13/2022] Open
Abstract
Pre-clinical models of traumatic brain injury (TBI) have been the primary experimental tool for understanding the potential mechanisms and cellular alterations that follow brain injury, but the human relevance and translational value of these models are often called into question. Efforts to better recapitulate injury biomechanics and the use of non-rodent species with neuroanatomical similarities to humans may address these concerns and promise to advance experimental studies toward clinical impact. In addition to improving translational aspects of animal models, it is also advantageous to establish pre-clinical outcomes that can be directly compared with the same outcomes in humans. Non-invasive imaging and particularly MRI is promising for this purpose given that MRI is a primary tool for clinical diagnosis and at the same time increasingly available at the pre-clinical level. The objective of this study was to identify which commonly used radiologic markers of TBI outcomes can be found also in a translationally relevant pre-clinical model of TBI. The ferret was selected as a human relevant species for this study with folded cortical geometry and relatively high white matter content and the closed head injury model of engineered rotation and acceleration (CHIMERA) TBI model was selected for biomechanical similarities to human injury. A comprehensive battery of MRI protocols based on common data elements (CDEs) for human TBI was collected longitudinally for the identification of MRI markers and voxelwise analysis of T2, contrast enhancement and diffusion tensor MRI values. The most prominent MRI findings were consistent with focal hemorrhage and edema in the brain stem region following high severity injury as well as vascular and meningeal injury evident by contrast enhancement. While conventional MRI outcomes were not highly conspicuous in less severe cases, quantitative voxelwise analysis indicated diffusivity and anisotropy alterations in the acute and chronic periods after TBI. The main conclusions of this study support the translational relevance of closed head TBI models in intermediate species and identify brain stem and meningeal vulnerability. Additionally, the MRI findings highlight a subset of CDEs with promise to bridge pre-clinical studies with human TBI outcomes.
Collapse
Affiliation(s)
- Elizabeth B. Hutchinson
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
- *Correspondence: Elizabeth B. Hutchinson,
| | | | - Hannah R. Johnson
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | - Andrew K. Knutsen
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Asamoah Bosomtwi
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Alexandru Korotcov
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Anandakumar Shunmugavel
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- National Institutes of Health, National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD, United States
| | - Sarah G. King
- National Institutes of Health, National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD, United States
| | - Susan C. Schwerin
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sharon L. Juliano
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Bernard J. Dardzinski
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Carlo Pierpaoli
- National Institutes of Health, National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD, United States
| |
Collapse
|
27
|
Williams HC, Carlson SW, Saatman KE. A role for insulin-like growth factor-1 in hippocampal plasticity following traumatic brain injury. VITAMINS AND HORMONES 2022; 118:423-455. [PMID: 35180936 DOI: 10.1016/bs.vh.2021.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Traumatic brain injury (TBI) initiates a constellation of secondary injury cascades, leading to neuronal damage and dysfunction that is often beyond the scope of endogenous repair mechanisms. Cognitive deficits are among the most persistent morbidities resulting from TBI, necessitating a greater understanding of mechanisms of posttraumatic hippocampal damage and neuroplasticity and identification of therapies that improve recovery by enhancing repair pathways. Focusing here on hippocampal neuropathology associated with contusion-type TBIs, the impact of brain trauma on synaptic structure and function and the process of adult neurogenesis is discussed, reviewing initial patterns of damage as well as evidence for spontaneous recovery. A case is made that insulin-like growth factor-1 (IGF-1), a growth-promoting peptide synthesized in both the brain and the periphery, is well suited to augment neuroplasticity in the injured brain. Essential during brain development, multiple lines of evidence delineate roles in the adult brain for IGF-1 in the maintenance of synapses, regulation of neurotransmission, and modulation of forms of synaptic plasticity such as long-term potentiation. Further, IGF-1 enhances adult hippocampal neurogenesis though effects on proliferation and neuronal differentiation of neural progenitor cells and on dendritic growth of newly born neurons. Post-injury administration of IGF-1 has been effective in rodent models of TBI in improving learning and memory, attenuating death of mature hippocampal neurons and promoting neurogenesis, providing critical proof-of-concept data. More studies are needed to explore the effects of IGF-1-based therapies on synaptogenesis and synaptic plasticity following TBI and to optimize strategies in order to stimulate only appropriate, functional neuroplasticity.
Collapse
Affiliation(s)
- Hannah C Williams
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Shaun W Carlson
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kathryn E Saatman
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
28
|
Tucker LB, McCabe JT. Measuring Anxiety-Like Behaviors in Rodent Models of Traumatic Brain Injury. Front Behav Neurosci 2021; 15:682935. [PMID: 34776887 PMCID: PMC8586518 DOI: 10.3389/fnbeh.2021.682935] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 10/06/2021] [Indexed: 12/31/2022] Open
Abstract
Anxiety is a common complaint following acquired traumatic brain injury (TBI). However, the measurement of dysfunctional anxiety behavioral states following experimental TBI in rodents is complex. Some studies report increased anxiety after TBI, whereas others find a decreased anxiety-like state, often described as increased risk-taking behavior or impulsivity. These inconsistencies may reflect a lack of standardization of experimental injury models or of behavioral testing techniques. Here, we review the most commonly employed unconditioned tests of anxiety and discuss them in a context of experimental TBI. Special attention is given to the effects of repeated testing, and consideration of potential sensory and motor confounds in injured rodents. The use of multiple tests and alternative data analysis methods are discussed, as well as the potential for the application of common data elements (CDEs) as a means of providing a format for documentation of experimental details and procedures of each published research report. CDEs may improve the rigor, reproducibility, as well as endpoint for better relating findings with clinical TBI phenotypes and the final goal of translation. While this may not resolve all incongruities in findings across laboratories, it is seen as a way forward for standardized and universal data collection for improvement of data quality and sharing, and advance therapies for neuropsychiatric symptoms that often present for decades following TBI.
Collapse
Affiliation(s)
- Laura B Tucker
- Preclinical Behavior and Models Core, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Joseph T McCabe
- Preclinical Behavior and Models Core, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
29
|
Traumatic brain injury augurs ill for prolonged deficits in the brain's structural and functional integrity following controlled cortical impact injury. Sci Rep 2021; 11:21559. [PMID: 34732737 PMCID: PMC8566513 DOI: 10.1038/s41598-021-00660-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/06/2021] [Indexed: 12/02/2022] Open
Abstract
Previous neuroimaging studies in rodents investigated effects of the controlled cortical impact (CCI) model of traumatic brain injury (TBI) within one-month post-TBI. This study extends this temporal window to monitor the structural–functional alterations from two hours to six months post-injury. Thirty-seven male Sprague–Dawley rats were randomly assigned to TBI and sham groups, which were scanned at two hours, 1, 3, 7, 14, 30, 60 days, and six months following CCI or sham surgery. Structural MRI, diffusion tensor imaging, and resting-state functional magnetic resonance imaging were acquired to assess the dynamic structural, microstructural, and functional connectivity alterations post-TBI. There was a progressive increase in lesion size associated with brain volume loss post-TBI. Furthermore, we observed reduced fractional anisotropy within 24 h and persisted to six months post-TBI, associated with acutely reduced axial diffusivity, and chronic increases in radial diffusivity post-TBI. Moreover, a time-dependent pattern of altered functional connectivity evolved over the six months’ follow-up post-TBI. This study extends the current understanding of the CCI model by confirming the long-term persistence of the altered microstructure and functional connectivity, which may hold a strong translational potential for understanding the long-term sequelae of TBI in humans.
Collapse
|
30
|
Priemer DS, Perl DP. A Commentary on "Delayed-Onset Neuropathological Complications From a Foramen Magnum & Occipital Crest Focused Traumatic Brain Injury of the Vietnam War" Parts I, II, and III. Mil Med 2021; 187:938-940. [PMID: 34632520 DOI: 10.1093/milmed/usab376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/28/2021] [Indexed: 11/12/2022] Open
Affiliation(s)
- David S Priemer
- Department of Pathology, Uniformed Services University F. Edward Hébert School of Medicine, Bethesda, MD 20814, USA.,Department of Defense, Uniformed Services University Brain Tissue Repository, Bethesda, MD 20817, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Daniel P Perl
- Department of Pathology, Uniformed Services University F. Edward Hébert School of Medicine, Bethesda, MD 20814, USA.,Department of Defense, Uniformed Services University Brain Tissue Repository, Bethesda, MD 20817, USA
| |
Collapse
|
31
|
Smith DH, Kochanek PM, Rosi S, Meyer R, Ferland-Beckham C, Prager EM, Ahlers ST, Crawford F. Roadmap for Advancing Pre-Clinical Science in Traumatic Brain Injury. J Neurotrauma 2021; 38:3204-3221. [PMID: 34210174 PMCID: PMC8820284 DOI: 10.1089/neu.2021.0094] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pre-clinical models of disease have long played important roles in the advancement of new treatments. However, in traumatic brain injury (TBI), despite the availability of numerous model systems, translation from bench to bedside remains elusive. Integrating clinical relevance into pre-clinical model development is a critical step toward advancing therapies for TBI patients across the spectrum of injury severity. Pre-clinical models include in vivo and ex vivo animal work-both small and large-and in vitro modeling. The wide range of pre-clinical models reflect substantial attempts to replicate multiple aspects of TBI sequelae in humans. Although these models reveal multiple putative mechanisms underlying TBI pathophysiology, failures to translate these findings into successful clinical trials call into question the clinical relevance and applicability of the models. Here, we address the promises and pitfalls of pre-clinical models with the goal of evolving frameworks that will advance translational TBI research across models, injury types, and the heterogenous etiology of pathology.
Collapse
Affiliation(s)
- Douglas H Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine; Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Rangos Research Center, Pittsburgh, Pennsylvania, USA
| | - Susanna Rosi
- Departments of Physical Therapy Rehabilitation Science, Neurological Surgery, Weill Institute for Neuroscience, University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Retsina Meyer
- Cohen Veterans Bioscience, New York, New York, USA.,Delix Therapeutics, Inc, Boston, Massachusetts, USA
| | | | | | - Stephen T Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate Naval Medical Research Center, Silver Spring, Maryland, USA
| | | |
Collapse
|
32
|
Animal models of traumatic brain injury: a review of pathophysiology to biomarkers and treatments. Exp Brain Res 2021; 239:2939-2950. [PMID: 34324019 DOI: 10.1007/s00221-021-06178-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
Traumatic brain injury (TBI) is one of the main causes of death and disability in both civilian and military population. TBI may occur via a variety of etiologies, all of which involve trauma to the head. However, the neuroprotective drugs which were found to be very effective in animal TBI models failed in phase II or phase III clinical trials, emphasizing a compelling need to review the current status of animal TBI models and therapeutic strategies. No single animal model can adequately mimic all aspects of human TBI owing to the heterogeneity of clinical TBI. However, due to the ethical limitations, it is difficult to precisely emulate the TBI mechanisms that occur in humans. Therefore, many animal models with varying severity and mechanisms of brain injury have been developed, and each model has its own pros and cons in its implementation for TBI research. These challenges pose a need for study of continued TBI mechanisms, brain injury severity, duration, treatment strategies, and optimization of animal models across the neurotrauma research community. The aim of this review is to discuss (1) causes of TBI, (2) its prevalence in military and civilian population, (3) classification and pathophysiology of TBI, (4) biomarkers and detection methods, (5) animal models of TBI, and (6) the advantages and disadvantages of each model and the species used, as well as possible treatments.
Collapse
|
33
|
Whitney K, Nikulina E, Rahman SN, Alexis A, Bergold PJ. Delayed dosing of minocycline plus N-acetylcysteine reduces neurodegeneration in distal brain regions and restores spatial memory after experimental traumatic brain injury. Exp Neurol 2021; 345:113816. [PMID: 34310944 DOI: 10.1016/j.expneurol.2021.113816] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022]
Abstract
Multiple drugs to treat traumatic brain injury (TBI) have failed clinical trials. Most drugs lose efficacy as the time interval increases between injury and treatment onset. Insufficient therapeutic time window is a major reason underlying failure in clinical trials. Few drugs have been developed with therapeutic time windows sufficiently long enough to treat TBI because little is known about which brain functions can be targeted if therapy is delayed hours to days after injury. We identified multiple injury parameters that are improved by first initiating treatment with the drug combination minocycline (MINO) plus N-acetylcysteine (NAC) at 72 h after injury (MN72) in a mouse closed head injury (CHI) experimental TBI model. CHI produces spatial memory deficits resulting in impaired performance on Barnes maze, hippocampal neuronal loss, and bilateral damage to hippocampal neurons, dendrites, spines and synapses. MN72 treatment restores Barnes maze acquisition and retention, protects against hippocampal neuronal loss, limits damage to dendrites, spines and synapses, and accelerates recovery of microtubule associated protein 2 (MAP2) expression, a key protein in maintaining proper dendritic architecture and synapse density. These data show that in addition to the structural integrity of the dendritic arbor, spine and synapse density can be successfully targeted with drugs first dosed days after injury. Retention of substantial drug efficacy even when first dosed 72 h after injury makes MINO plus NAC a promising candidate to treat clinical TBI.
Collapse
Affiliation(s)
- Kristen Whitney
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America; Program in Neural and Behavioral Science, School of Graduate Studies, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Elena Nikulina
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Syed N Rahman
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Alisia Alexis
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Peter J Bergold
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America; Program in Neural and Behavioral Science, School of Graduate Studies, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America.
| |
Collapse
|
34
|
Mester JR, Bazzigaluppi P, Dorr A, Beckett T, Burke M, McLaurin J, Sled JG, Stefanovic B. Attenuation of tonic inhibition prevents chronic neurovascular impairments in a Thy1-ChR2 mouse model of repeated, mild traumatic brain injury. Am J Cancer Res 2021; 11:7685-7699. [PMID: 34335958 PMCID: PMC8315057 DOI: 10.7150/thno.60190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: Mild traumatic brain injury (mTBI), the most common type of brain trauma, frequently leads to chronic cognitive and neurobehavioral deficits. Intervening effectively is impeded by our poor understanding of its pathophysiological sequelae. Methods: To elucidate the long-term neurovascular sequelae of mTBI, we combined optogenetics, two-photon fluorescence microscopy, and intracortical electrophysiological recordings in mice to selectively stimulate peri-contusional neurons weeks following repeated closed-head injury and probe individual vessel's function and local neuronal reactivity. Results: Compared to sham-operated animals, mTBI mice showed doubled cortical venular speeds (115 ± 25%) and strongly elevated cortical venular reactivity (53 ± 17%). Concomitantly, the pericontusional neurons exhibited attenuated spontaneous activity (-57 ± 79%) and decreased reactivity (-47 ± 28%). Post-mortem immunofluorescence revealed signs of peri-contusional senescence and DNA damage, in the absence of neuronal loss or gliosis. Alteration of neuronal and vascular functioning was largely prevented by chronic, low dose, systemic administration of a GABA-A receptor inverse agonist (L-655,708), commencing 3 days following the third impact. Conclusions: Our findings indicate that repeated mTBI leads to dramatic changes in the neurovascular unit function and that attenuation of tonic inhibition can prevent these alterations. The sustained disruption of the neurovascular function may underlie the concussed brain's long-term susceptibility to injury, and calls for development of better functional assays as well as of neurovascularly targeted interventions.
Collapse
|
35
|
Mayer AR, Ling JM, Dodd AB, Rannou-Latella JG, Stephenson DD, Dodd RJ, Mehos CJ, Patton DA, Cullen DK, Johnson VE, Pabbathi Reddy S, Robertson-Benta CR, Gigliotti AP, Meier TB, Vermillion MS, Smith DH, Kinsler R. Reproducibility and Characterization of Head Kinematics During a Large Animal Acceleration Model of Traumatic Brain Injury. Front Neurol 2021; 12:658461. [PMID: 34177763 PMCID: PMC8219951 DOI: 10.3389/fneur.2021.658461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/03/2021] [Indexed: 11/13/2022] Open
Abstract
Acceleration parameters have been utilized for the last six decades to investigate pathology in both human and animal models of traumatic brain injury (TBI), design safety equipment, and develop injury thresholds. Previous large animal models have quantified acceleration from impulsive loading forces (i.e., machine/object kinematics) rather than directly measuring head kinematics. No study has evaluated the reproducibility of head kinematics in large animal models. Nine (five males) sexually mature Yucatan swine were exposed to head rotation at a targeted peak angular velocity of 250 rad/s in the coronal plane. The results indicated that the measured peak angular velocity of the skull was 51% of the impulsive load, was experienced over 91% longer duration, and was multi- rather than uni-planar. These findings were replicated in a second experiment with a smaller cohort (N = 4). The reproducibility of skull kinematics data was mostly within acceptable ranges based on published industry standards, although the coefficients of variation (8.9% for peak angular velocity or 12.3% for duration) were higher than the impulsive loading parameters produced by the machine (1.1 vs. 2.5%, respectively). Immunohistochemical markers of diffuse axonal injury and blood-brain barrier breach were not associated with variation in either skull or machine kinematics, suggesting that the observed levels of variance in skull kinematics may not be biologically meaningful with the current sample sizes. The findings highlight the reproducibility of a large animal acceleration model of TBI and the importance of direct measurements of skull kinematics to determine the magnitude of angular velocity, refine injury criteria, and determine critical thresholds.
Collapse
Affiliation(s)
- Andrew R. Mayer
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
- Neurology Department, University of New Mexico School of Medicine, Albuquerque, NM, United States
- Psychiatry Department, University of New Mexico School of Medicine, Albuquerque, NM, United States
- Psychology Department, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Josef M. Ling
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Andrew B. Dodd
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Julie G. Rannou-Latella
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - David D. Stephenson
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Rebecca J. Dodd
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Carissa J. Mehos
- Neurosciences Department, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Declan A. Patton
- Center for Injury Research and Prevention, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - D. Kacy Cullen
- Department of Neurosurgery and Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Victoria E. Johnson
- Department of Neurosurgery and Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sharvani Pabbathi Reddy
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | | | - Andrew P. Gigliotti
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Timothy B. Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Meghan S. Vermillion
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Douglas H. Smith
- Department of Neurosurgery and Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rachel Kinsler
- Enroute Care Group, U.S. Army Aeromedical Research Laboratory, Fort Rucker, AL, United States
| |
Collapse
|
36
|
Bodnar CN, Watson JB, Higgins EK, Quan N, Bachstetter AD. Inflammatory Regulation of CNS Barriers After Traumatic Brain Injury: A Tale Directed by Interleukin-1. Front Immunol 2021; 12:688254. [PMID: 34093593 PMCID: PMC8176952 DOI: 10.3389/fimmu.2021.688254] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/05/2021] [Indexed: 01/13/2023] Open
Abstract
Several barriers separate the central nervous system (CNS) from the rest of the body. These barriers are essential for regulating the movement of fluid, ions, molecules, and immune cells into and out of the brain parenchyma. Each CNS barrier is unique and highly dynamic. Endothelial cells, epithelial cells, pericytes, astrocytes, and other cellular constituents each have intricate functions that are essential to sustain the brain's health. Along with damaging neurons, a traumatic brain injury (TBI) also directly insults the CNS barrier-forming cells. Disruption to the barriers first occurs by physical damage to the cells, called the primary injury. Subsequently, during the secondary injury cascade, a further array of molecular and biochemical changes occurs at the barriers. These changes are focused on rebuilding and remodeling, as well as movement of immune cells and waste into and out of the brain. Secondary injury cascades further damage the CNS barriers. Inflammation is central to healthy remodeling of CNS barriers. However, inflammation, as a secondary pathology, also plays a role in the chronic disruption of the barriers' functions after TBI. The goal of this paper is to review the different barriers of the brain, including (1) the blood-brain barrier, (2) the blood-cerebrospinal fluid barrier, (3) the meningeal barrier, (4) the blood-retina barrier, and (5) the brain-lesion border. We then detail the changes at these barriers due to both primary and secondary injury following TBI and indicate areas open for future research and discoveries. Finally, we describe the unique function of the pro-inflammatory cytokine interleukin-1 as a central actor in the inflammatory regulation of CNS barrier function and dysfunction after a TBI.
Collapse
Affiliation(s)
- Colleen N. Bodnar
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - James B. Watson
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Emma K. Higgins
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| | - Adam D. Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
37
|
Chen B, Shi QX, Nie C, Zhao ZP, Luo L, Zhao QJ, Si SY, Xu BX, Wang T, Gao LY, Gu JW. Establishment and Evaluation of a Novel High-Efficiency Model of Graded Traumatic Brain Injury in Mice. World Neurosurg 2021; 154:e7-e18. [PMID: 33992827 DOI: 10.1016/j.wneu.2021.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Although previous studies have made significant contributions to establishing animal traumatic brain injury (TBI) models for simulation of human TBI, the accuracy, controllability, and modeling efficiency of animal TBI models need to be further improved. This study established a novel high-efficiency graded mouse TBI model induced by shock wave. METHODS A total of 125 mice were randomly divided into sham, 0.7 mm, 0.6 mm, and 0.5 mm groups according to the depth of the cross groove of the aluminum sheets. The stability and repeatability of apparatus were evaluated, and the integrity of the blood-brain barrier, cerebral edema, neuropathologic immunohistochemistry, apoptosis-related protein, and behavioral tests of neurologic function were used to validate this new model. RESULTS The results showed that 4 mice were injured simultaneously in 1 experiment. They received the same intensity of shock waves. Moreover, the mortality rates caused by 3 different aluminum sheets were consistent with the mortality rates of mild TBI, moderate TBI, and severe TBI. Compared with the sham group, mice in different injured groups significantly increased brain water content, blood-brain barrier permeability, and neuronal apoptosis. And the mice in all injured groups showed poor motor ability, balancing, spatial learning, and memory abilities. CONCLUSIONS The novel TBI apparatus has advantages in its small size, simple operation, high repeatability, high efficiency, and graded severity. Our TBI apparatus provides a novel tool to investigate the neuropathologic changes and underlying mechanisms of TBI with various levels of severities.
Collapse
Affiliation(s)
- Bing Chen
- Savaid Medical College, University of Chinese Academy of Sciences, Hangzhou, China
| | - Quan-Xing Shi
- People's Liberation Army Strategic Support Force Medical Center, Hangzhou, China
| | - Chuang Nie
- People's Liberation Army Strategic Support Force Medical Center, Hangzhou, China
| | - Zhi-Ping Zhao
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Ling Luo
- People's Liberation Army Strategic Support Force Medical Center, Hangzhou, China
| | - Quan-Jun Zhao
- People's Liberation Army Strategic Support Force Medical Center, Hangzhou, China
| | - Shao-Yan Si
- People's Liberation Army Strategic Support Force Medical Center, Hangzhou, China
| | - Bing-Xin Xu
- People's Liberation Army Strategic Support Force Medical Center, Hangzhou, China
| | - Tao Wang
- People's Liberation Army Strategic Support Force Medical Center, Hangzhou, China
| | - Ling-Yu Gao
- Zhejiang University School of Medicine, Hangzhou, China
| | - Jian-Wen Gu
- People's Liberation Army Strategic Support Force Medical Center, Hangzhou, China.
| |
Collapse
|
38
|
Hegdekar N, Lipinski MM, Sarkar C. N-Acetyl-L-leucine improves functional recovery and attenuates cortical cell death and neuroinflammation after traumatic brain injury in mice. Sci Rep 2021; 11:9249. [PMID: 33927281 PMCID: PMC8084982 DOI: 10.1038/s41598-021-88693-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/15/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and long-term disability around the world. Even mild to moderate TBI can lead to lifelong neurological impairment due to acute and progressive neurodegeneration and neuroinflammation induced by the injury. Thus, the discovery of novel treatments which can be used as early therapeutic interventions following TBI is essential to restrict neuronal cell death and neuroinflammation. We demonstrate that orally administered N-acetyl-l-leucine (NALL) significantly improved motor and cognitive outcomes in the injured mice, led to the attenuation of cell death, and reduced the expression of neuroinflammatory markers after controlled cortical impact (CCI) induced experimental TBI in mice. Our data indicate that partial restoration of autophagy flux mediated by NALL may account for the positive effect of treatment in the injured mouse brain. Taken together, our study indicates that treatment with NALL would be expected to improve neurological function after injury by restricting cortical cell death and neuroinflammation. Therefore, NALL is a promising novel, neuroprotective drug candidate for the treatment of TBI.
Collapse
Affiliation(s)
- Nivedita Hegdekar
- Department of Anesthesiology, Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Marta M Lipinski
- Department of Anesthesiology, Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Chinmoy Sarkar
- Department of Anesthesiology, Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
39
|
Mayer AR, Dodd AB, Ling JM, Stephenson DD, Rannou-Latella JG, Vermillion MS, Mehos CJ, Johnson VE, Gigliotti AP, Dodd RJ, Chaudry IH, Meier TB, Smith DH, Bragin DE, Lai C, Wagner CL, Guedes VA, Gill JM, Kinsler R. Survival Rates and Biomarkers in a Large Animal Model of Traumatic Brain Injury Combined With Two Different Levels of Blood Loss. Shock 2021; 55:554-562. [PMID: 32881755 PMCID: PMC8112147 DOI: 10.1097/shk.0000000000001653] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION The pathology resulting from concurrent traumatic brain injury (TBI) and hemorrhagic shock (HS; TBI+HS) are leading causes of mortality and morbidity worldwide following trauma. However, the majority of large animal models of TBI+HS have utilized focal/contusional injuries rather than incorporating the types of brain trauma (closed-head injury caused by dynamic acceleration) that typify human injury. OBJECTIVE To examine survival rates and effects on biomarkers from rotational TBI with two levels of HS. METHODS Twenty-two sexually mature Yucatan swine (30.39 ± 2.25 kg; 11 females) therefore underwent either Sham trauma procedures (n = 6) or a dynamic acceleration TBI combined with either 55% (n = 8) or 40% (n = 8) blood loss in this serial study. RESULTS Survival rates were significantly higher for the TBI+40% (87.5%) relative to TBI+55% (12.5%) cohort, with the majority of TBI+55% animals expiring within 2 h post-trauma from apnea. Blood-based neural biomarkers and immunohistochemistry indicated evidence of diffuse axonal injury (increased NFL/Aβ42), blood-brain barrier breach (increased immunoglobulin G) and inflammation (increased glial fibrillary acidic protein/ionized calcium-binding adaptor molecule 1) in the injured cohorts relative to Shams. Invasive hemodynamic measurements indicated increased shock index and decreased pulse pressure in both injury cohorts, with evidence of partial recovery for invasive hemodynamic measurements in the TBI+40% cohort. Similarly, although both injury groups demonstrated ionic and blood gas abnormalities immediately postinjury, metabolic acidosis continued to increase in the TBI+55% group ∼85 min postinjury. Somewhat surprisingly, both neural and physiological biomarkers showed significant changes within the Sham cohort across the multi-hour experimental procedure, most likely associated with prolonged anesthesia. CONCLUSION Current results suggest the TBI+55% model may be more appropriate for severe trauma requiring immediate medical attention/standard fluid resuscitation protocols whereas the TBI+40% model may be useful for studies of prolonged field care.
Collapse
Affiliation(s)
- Andrew R. Mayer
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, New Mexico
- Neurology Department, University of New Mexico School of Medicine, Albuquerque, New Mexico
- Psychiatry Department, University of New Mexico School of Medicine, Albuquerque, New Mexico
- Psychology Department, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Andrew B. Dodd
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, New Mexico
| | - Josef M. Ling
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, New Mexico
| | - David D. Stephenson
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, New Mexico
| | | | - Meghan S. Vermillion
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, New Mexico
| | - Carissa J. Mehos
- Neurosciences Department, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Victoria E. Johnson
- Department of Neurosurgery and Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew P. Gigliotti
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, New Mexico
| | - Rebecca J. Dodd
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, New Mexico
| | - Irshad H. Chaudry
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Timothy B. Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Douglas H. Smith
- Department of Neurosurgery and Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Denis E. Bragin
- The Mind Research Network/Lovelace Biomedical Research Institute, Albuquerque, New Mexico
- Neurosurgery Department, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Chen Lai
- National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland
| | - Chelsea L. Wagner
- National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland
| | - Vivian A. Guedes
- National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland
| | - Jessica M. Gill
- National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland
| | - Rachel Kinsler
- Enroute Care Group, U.S. Army Aeromedical Research Laboratory, Fort Rucker, Alabama
| |
Collapse
|
40
|
Wiegand TLT, Sollmann N, Bonke EM, Umeasalugo KE, Sobolewski KR, Plesnila N, Shenton ME, Lin AP, Koerte IK. Translational neuroimaging in mild traumatic brain injury. J Neurosci Res 2021; 100:1201-1217. [PMID: 33789358 DOI: 10.1002/jnr.24840] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/09/2021] [Accepted: 03/17/2021] [Indexed: 01/26/2023]
Abstract
Traumatic brain injuries (TBIs) are common with an estimated 27.1 million cases per year. Approximately 80% of TBIs are categorized as mild TBI (mTBI) based on initial symptom presentation. While in most individuals, symptoms resolve within days to weeks, in some, symptoms become chronic. Advanced neuroimaging has the potential to characterize brain morphometric, microstructural, biochemical, and metabolic abnormalities following mTBI. However, translational studies are needed for the interpretation of neuroimaging findings in humans with respect to the underlying pathophysiological processes, and, ultimately, for developing novel and more targeted treatment options. In this review, we introduce the most commonly used animal models for the study of mTBI. We then summarize the neuroimaging findings in humans and animals after mTBI and, wherever applicable, the translational aspects of studies available today. Finally, we highlight the importance of translational approaches and outline future perspectives in the field of translational neuroimaging in mTBI.
Collapse
Affiliation(s)
- Tim L T Wiegand
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nico Sollmann
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| | - Elena M Bonke
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
| | - Kosisochukwu E Umeasalugo
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Kristen R Sobolewski
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| | - Martha E Shenton
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander P Lin
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Inga K Koerte
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
Nakata M, Shimoda M, Yamamoto S. UV-Induced Neuronal Degeneration in the Rat Cerebral Cortex. Cereb Cortex Commun 2021; 2:tgab006. [PMID: 34296154 PMCID: PMC8152860 DOI: 10.1093/texcom/tgab006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/01/2022] Open
Abstract
Irradiation with ultraviolet (UV) light on the cortical surface can induce a focal brain lesion (UV lesion) in rodents. In the present study, we investigated the process of establishing a UV lesion. Rats underwent UV irradiation (365-nm wavelength, 2.0 mWh) over the dura, and time-dependent changes in the cortical tissue were analyzed histologically. We found that the majority of neurons in the lesion started to degenerate within 24 h and the rest disappeared within 5 days after irradiation. UV-induced neuronal degeneration progressed in a layer-dependent manner. Moreover, UV-induced terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) positivity and heme oxygenase-1 (HO-1) immunoreactivity were also detected. These findings suggest that UV irradiation in the brain can induce gradual neural degeneration and oxidative stress. Importantly, UV vulnerability may vary among cortical layers. UV-induced cell death may be due to apoptosis; however, there remains a possibility that UV-irradiated cells were degenerated via processes other than apoptosis. The UV lesion technique will not only assist in investigating brain function at a targeted site but may also serve as a pathophysiological model of focal brain injury and/or neurodegenerative disorders.
Collapse
Affiliation(s)
- Mariko Nakata
- Integrative Neuroscience Research Group, Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8568, Japan
| | - Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shinya Yamamoto
- Integrative Neuroscience Research Group, Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8568, Japan
| |
Collapse
|
42
|
Kaur A, Jaiswal G, Brar J, Kumar P. Neuroprotective effect of nerolidol in traumatic brain injury associated behavioural comorbidities in rats. Toxicol Res (Camb) 2021; 10:40-50. [PMID: 33613971 PMCID: PMC7885190 DOI: 10.1093/toxres/tfaa100] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/20/2020] [Accepted: 11/25/2020] [Indexed: 11/14/2022] Open
Abstract
Traumatic brain injury (TBI) is an insult to the brain from an external mechanical force, leading to temporary/permanent secondary injuries, i.e. impairment of cognitive, physical, and psycho-social functions with altered consciousness. The leading mechanism responsible for neuronal damage following TBI is an increase in oxidative reactions initiated by free radicals generated by the injury along with various other mechanisms. Nerolidol is reported to have potent antioxidant and anti-neuroinflammatory properties. The present study was designed to explore the neuroprotective effect of nerolidol in weight-drop-induced TBI in rats. Animals were injured on the 1st day by dropping a free-falling weight of 200 gm from a height of 1 m through a guide pipe onto the exposed skull. After 14 days of injury, nerolidol (25, 50, and 100 mg/kg, i.p.) treatment was given for the next 14 days. Locomotor activity and motor coordination were evaluated using an actophotometer and rotarod, respectively. Cognitive impairment was observed through the Morris Water Maze and Object Recognition Test. On the 29th day, animals were sacrificed, and their brains were collected for the biochemical estimation. The weight drop model significantly decreased locomotor activity, motor coordination, increased Acetylcholinesterase (AChE) activity, oxidative stress, and induced cognitive deficits in TBI rats. Nerolidol significantly improved locomotor activity, reversed motor incoordination and cognitive impairment, and reduced the AChE activity and oxidative/nitrosative stress. The present study demonstrates the promising neuroprotective effects of nerolidol, which might improve the quality of life of TBI patients.
Collapse
Affiliation(s)
- Amandeep Kaur
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda-151001 (Punjab), India
| | - Gagandeep Jaiswal
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda-151001 (Punjab), India
| | - Jasdeep Brar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda-151001 (Punjab), India
| | - Puneet Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda-151001 (Punjab), India
- Department of Pharmacology, Central University of Punjab, Bathinda-151001 (Punjab), India
| |
Collapse
|
43
|
Albayram O, Albayram S, Mannix R. Chronic traumatic encephalopathy-a blueprint for the bridge between neurological and psychiatric disorders. Transl Psychiatry 2020; 10:424. [PMID: 33293571 PMCID: PMC7723988 DOI: 10.1038/s41398-020-01111-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 10/21/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a perplexing condition characterized by a broad and diverse range of neuropathology and psychopathology. While there are no agreed upon or validated clinical criteria for CTE, case series of CTE have described a wide range of neuropsychiatric symptoms that have been attributed to repetitive traumatic brain injuries (rTBI). However, the direct links between the psychopathology of psychiatric and neurological conditions from rTBI to CTE remains poorly understood. Prior studies suggest that repetitive cerebral injuries are associated with damage to neural circuitry involved in emotional and memory processes, but these studies do not offer longitudinal assessments that prove causation. More recent studies on novel targets, such as transmission of misfolded proteins, as well as newly advanced non-invasive imaging techniques may offer more direct evidence of the pathogenesis of CTE by tracing the progression of pathology and display of related behavioral impairments. Understanding this interface in the context of rTBI can play an important role in future approaches to the definition, assessment, prevention, and treatment of CTE and mental illnesses.
Collapse
Affiliation(s)
- Onder Albayram
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Sait Albayram
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Rebekkah Mannix
- Division of Emergency Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
44
|
Keating CE, Cullen DK. Mechanosensation in traumatic brain injury. Neurobiol Dis 2020; 148:105210. [PMID: 33259894 DOI: 10.1016/j.nbd.2020.105210] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is distinct from other neurological disorders because it is induced by a discrete event that applies extreme mechanical forces to the brain. This review describes how the brain senses, integrates, and responds to forces under both normal conditions and during injury. The response to forces is influenced by the unique mechanical properties of brain tissue, which differ by region, cell type, and sub-cellular structure. Elements such as the extracellular matrix, plasma membrane, transmembrane receptors, and cytoskeleton influence its properties. These same components also act as force-sensors, allowing neurons and glia to respond to their physical environment and maintain homeostasis. However, when applied forces become too large, as in TBI, these components may respond in an aberrant manner or structurally fail, resulting in unique pathological sequelae. This so-called "pathological mechanosensation" represents a spectrum of cellular responses, which vary depending on the overall biomechanical parameters of the injury and may be compounded by repetitive injuries. Such aberrant physical responses and/or damage to cells along with the resulting secondary injury cascades can ultimately lead to long-term cellular dysfunction and degeneration, often resulting in persistent deficits. Indeed, pathological mechanosensation not only directly initiates secondary injury cascades, but this post-physical damage environment provides the context in which these cascades unfold. Collectively, these points underscore the need to use experimental models that accurately replicate the biomechanics of TBI in humans. Understanding cellular responses in context with injury biomechanics may uncover therapeutic targets addressing various facets of trauma-specific sequelae.
Collapse
Affiliation(s)
- Carolyn E Keating
- Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz VA Medical Center, USA
| | - D Kacy Cullen
- Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz VA Medical Center, USA.
| |
Collapse
|
45
|
Santiago-Castañeda C, Segovia-Oropeza M, Concha L, Orozco-Suárez SA, Rocha L. Propylparaben Reduces the Long-Term Consequences in Hippocampus Induced by Traumatic Brain Injury in Rats: Its Implications as Therapeutic Strategy to Prevent Neurodegenerative Diseases. J Alzheimers Dis 2020; 82:S215-S226. [PMID: 33185606 DOI: 10.3233/jad-200914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Severe traumatic brain injury (TBI), an important risk factor for Alzheimer's disease, induces long-term hippocampal damage and hyperexcitability. On the other hand, studies support that propylparaben (PPB) induces hippocampal neuroprotection in neurodegenerative diseases. OBJECTIVE Experiments were designed to evaluate the effects of subchronic treatment with PPB on TBI-induced changes in the hippocampus of rats. METHODS Severe TBI was induced using the lateral fluid percussion model. Subsequently, rats received subchronic administration with PPB (178 mg/kg, TBI+PPB) or vehicle (TBI+PEG) daily for 5 days. The following changes were examined during the experimental procedure: sensorimotor dysfunction, changes in hippocampal excitability, as well as neuronal damage and volume. RESULTS TBI+PEG group showed sensorimotor dysfunction (p < 0.001), hyperexcitability (64.2%, p < 0.001), and low neuronal preservation ipsi- and contralateral to the trauma. Magnetic resonance imaging (MRI) analysis revealed lower volume (17.2%; p < 0.01) and great damage to the ipsilateral hippocampus. TBI+PPB group showed sensorimotor dysfunction that was partially reversed 30 days after trauma. This group showed hippocampal excitability and neuronal preservation similar to the control group. However, MRI analysis revealed lower hippocampal volume (p < 0.05) when compared with the control group. CONCLUSION The present study confirms that post-TBI subchronic administration with PPB reduces the long-term consequences of trauma in the hippocampus. Implications of PPB as a neuroprotective strategy to prevent the development of Alzheimer's disease as consequence of TBI are discussed.
Collapse
Affiliation(s)
- Cindy Santiago-Castañeda
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| | - Marysol Segovia-Oropeza
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| | - Luis Concha
- Institute of Neurobiology, National Autonomous University of Mexico, Campus Juriquilla, Queretaro, Mexico
| | - Sandra Adela Orozco-Suárez
- Unit for Medical Research in Neurological Diseases, Specialties Hospital, National Medical Center SXXI (CMN-SXXI), Mexico City, Mexico
| | - Luisa Rocha
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
46
|
Acute damage to the blood–brain barrier and perineuronal net integrity in a clinically-relevant rat model of traumatic brain injury. Neuroreport 2020; 31:1167-1174. [PMID: 32991524 DOI: 10.1097/wnr.0000000000001531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Closed-head, frontal impacts in which the brain undergoes both lateral and rotational acceleration comprise the majority of human traumatic brain injury (TBI). Here, we utilize a clinically relevant model to examine the effects of a single concussion on aspects of brain integrity: the blood-brain barrier, the perineuronal nets (PNNs), and diffuse axonal injury. Adult, male Sprague-Dawley rats received either a frontal, closed-head concussive TBI, or no injury and were evaluated at 1- or 7-day post-injury. Using immunolabeling for albumin, we observed a significant increase in the permeability of the blood-brain barrier at 1-, but not 7-day post-injury. Breakdown of the PNN, as measured by the binding of wisteria floribunda, was observed at 1-day post-injury in the dorsal, lateral, and ventral cortices. This difference was resolved at 7-day. Finally, axonal injury was identified at both 1- and 7-day post-injury. This preclinical model of closed-head, frontal TBI presents a useful tool with which to understand better the acute pathophysiology of a single, frontal TBI.
Collapse
|
47
|
Fuentes del Toro S, Santos-Cuadros S, Olmeda E, San Román JL. Study of the Emergency Braking Test with an Autonomous Bus and the sEMG Neck Response by Means of a Low-Cost System. MICROMACHINES 2020; 11:mi11100931. [PMID: 33066252 PMCID: PMC7602115 DOI: 10.3390/mi11100931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/05/2023]
Abstract
Nowadays, due to the advances and the increasing implementation of the autonomous braking systems in vehicles, the non-collision accident is expected to become more common than a crash when a sudden stop happens. The most common injury in this kind of accident is whiplash or cervical injury since the neck has high sensitivity to sharp deceleration. To date, biomechanical research has usually been developed inside laboratories and does not entirely represent real conditions (e.g., restraint systems or surroundings of the experiment). With the aim of knowing the possible neck effects and consequences of an automatic emergency braking inside an autonomous bus, a surface electromyography (sEMG) system built by low-cost elements and developed by us, in tandem with other devices, such as accelerometers or cameras, were used. Moreover, thanks to the collaboration of 18 participants, it was possible to study the non-collision effects in two different scenarios (braking test in which the passenger is seated and looking ahead while talking with somebody in front of him (BT1) and, a second braking test where the passenger used a smartphone (BT2) and nobody is seated in front of him talking to him). The aim was to assess the sEMG neck response in the most common situations when somebody uses some kind of transport in order to conclude which environments are riskier regarding a possible cervical injury.
Collapse
Affiliation(s)
- Sergio Fuentes del Toro
- Mechanical Engineering Department, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganés, Spain; (S.S.-C.); (E.O.); (J.L.S.R.)
- Institute for Automotive Vehicle Safety (ISVA), Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganés, Spain
- Correspondence: ; Tel.: +34-916-624-8840
| | - Silvia Santos-Cuadros
- Mechanical Engineering Department, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganés, Spain; (S.S.-C.); (E.O.); (J.L.S.R.)
- Institute for Automotive Vehicle Safety (ISVA), Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganés, Spain
| | - Ester Olmeda
- Mechanical Engineering Department, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganés, Spain; (S.S.-C.); (E.O.); (J.L.S.R.)
- Institute for Automotive Vehicle Safety (ISVA), Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganés, Spain
| | - José Luis San Román
- Mechanical Engineering Department, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganés, Spain; (S.S.-C.); (E.O.); (J.L.S.R.)
- Institute for Automotive Vehicle Safety (ISVA), Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganés, Spain
| |
Collapse
|
48
|
McCabe JT, Tucker LB. Sex as a Biological Variable in Preclinical Modeling of Blast-Related Traumatic Brain Injury. Front Neurol 2020; 11:541050. [PMID: 33101170 PMCID: PMC7554632 DOI: 10.3389/fneur.2020.541050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022] Open
Abstract
Approaches to furthering our understanding of the bioeffects, behavioral changes, and treatment options following exposure to blast are a worldwide priority. Of particular need is a more concerted effort to employ animal models to determine possible sex differences, which have been reported in the clinical literature. In this review, clinical and preclinical reports concerning blast injury effects are summarized in relation to sex as a biological variable (SABV). The review outlines approaches that explore the pertinent role of sex chromosomes and gonadal steroids for delineating sex as a biological independent variable. Next, underlying biological factors that need exploration for blast effects in light of SABV are outlined, including pituitary, autonomic, vascular, and inflammation factors that all have evidence as having important SABV relevance. A major second consideration for the study of SABV and preclinical blast effects is the notable lack of consistent model design—a wide range of devices have been employed with questionable relevance to real-life scenarios—as well as poor standardization for reporting of blast parameters. Hence, the review also provides current views regarding optimal design of shock tubes for approaching the problem of primary blast effects and sex differences and outlines a plan for the regularization of reporting. Standardization and clear description of blast parameters will provide greater comparability across models, as well as unify consensus for important sex difference bioeffects.
Collapse
Affiliation(s)
- Joseph T McCabe
- Pre-clinical Studies Core, Center for Neuroscience and Regenerative Medicine, Bethesda, IL, United States.,Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Laura B Tucker
- Pre-clinical Studies Core, Center for Neuroscience and Regenerative Medicine, Bethesda, IL, United States.,Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
49
|
Taguchi D, Ehara A, Seo Y, Ueda S. Microhemorrhage in a Rat Model of Neonatal Shaking Brain Injury: Correlation between MRI and Iron Histochemistry. Acta Histochem Cytochem 2020; 53:83-91. [PMID: 32873992 PMCID: PMC7450178 DOI: 10.1267/ahc.20007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/07/2020] [Indexed: 11/22/2022] Open
Abstract
Previous studies have shown that neonatal shaking brain injury (SBI) causes transient microhemorrhages (MHs) in the gray matter of the cerebral cortex and hippocampus. Iron deposits and iron-uptake cells are observed surrounding MHs in this SBI model, suggesting local hypoxic-ischemic conditions. However, whether the shaken pups suffered systemic hypoxic-ischemic conditions has remained uncertain. Further, histopathological correlations of MHs on magnetic resonance imaging (MRI) are still unclear. The present study examined MHs after neonatal SBI using a combination of histochemical and susceptibility-weighted imaging (SWI) analyses. Systemic oxygen saturation analyses indicated no significant difference between shaken and non-shaken pups. MHs on postnatal day 4 (P4) pups showed decreased signal intensity on SWI. Iron histochemistry revealed that these hypointense areas almost completely comprised red blood cells (RBCs). MHs that appeared on P4 gradually disappeared by P7-12 on SWI. These resolved areas contained small numbers of RBCs, numerous iron-positive cells, and punctate regions with iron reaction products. Perivascular iron products were evident after P12. These changes progressed faster in the hippocampus than in cortical areas. These changes in MHs following neonatal SBI may provide new insights into microvascular pathologies and impacts on brain functions as adults.
Collapse
Affiliation(s)
- Daisuke Taguchi
- Department of Judo Therapy, Faculty of Medical Technology, Teikyo University
- Department Histology and Neurobiology, Dokkyo Medical University, School of Medicine
| | - Ayuka Ehara
- Department Histology and Neurobiology, Dokkyo Medical University, School of Medicine
| | - Yoshiteru Seo
- Department of Regulatory Physiology, Dokkyo Medical University, School of Medicine
| | - Shuichi Ueda
- Department Histology and Neurobiology, Dokkyo Medical University, School of Medicine
| |
Collapse
|
50
|
Nasution RA, Islam AA, Hatta M, Prihantono, Kaelan C, Poniman J, Wangi H. Modification of the Marmarou model in developing countries. Ann Med Surg (Lond) 2020; 57:109-113. [PMID: 32742649 PMCID: PMC7385274 DOI: 10.1016/j.amsu.2020.07.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 01/14/2023] Open
Abstract
Introduction Head injury is an injury or wound of the brain tissue due to external forces; it can cause a decrease or change in the status of consciousness. Many head injury models have used mice as experimental animals; the Marmarou model is the most famous and the most widely-used diffuse brain injury model. In this study, we slightly modified the Marmarou model. The purpose of this study is to help researchers examining head injuries in mice, especially those in developing countries who have limited facilities and infrastructure. Methods This experimental research uses animals models (Rattus novergicus, strain Sprague Dawley) that fit several criteria, including male, aged 10–12 weeks, and body weight of 200–300 g. This study involves a slight modification on the tube used, with a 20 cm-long weight of 20 g. The blood samples for the following assays of ELISA and brain tissue samples were collected at 24 h and 4, 5, 6, and 7 days post-trauma. Results A significant effect on the brain was seen with the Marmarou model modification, at a mass weight of 20 g and height of 20 cm, with 0.04 J energy produced. Changes were also seen in the histological features of brain tissue and the serum levels of AQP-4, F2 IsoPs, MPO, and VEGF from 24 h until 7 days after trauma. Conclusion This report describes the development of an experimental head injury approach modifying the Marmarou model that is able to produce a diffuse brain injury model in mice. Head injury is an injury of the brain tissue due to external forces. The Marmarou model is the most famous and widely-used diffuse brain injury model in mice. A significant effect on the brain was seen with our modified Marmarou trauma model.
Collapse
Affiliation(s)
- Rizha Anshori Nasution
- Department of Neurosurgery, Pelamonia Hospital, Makassar, Indonesia.,Doctoral Program of Biomedical Sciences, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Andi Assadul Islam
- Department of Neurosurgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Mochammad Hatta
- Clinical Microbiologist Program, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Prihantono
- Department of Surgery Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Cahyono Kaelan
- Department of Pathological Anatomy, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Jeni Poniman
- Department of Pathological Anatomy, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Harakati Wangi
- Department of Internal Medicine, Pelamonia Hospital, Makassar, Indonesia
| |
Collapse
|