1
|
Al Yacoub ON, Awwad HO, Standifer KM. Recovery from Traumatic Brain Injury Is Nociceptin/Orphanin FQ Peptide Receptor Genotype-, Sex-, and Injury Severity-Dependent. J Pharmacol Exp Ther 2024; 389:136-149. [PMID: 37442620 DOI: 10.1124/jpet.123.001664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/24/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the United States, and survivors often experience mental and physical health consequences that reduce quality of life. We previously reported that blockade of the nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor reduced tissue damage markers produced by blast TBI. The goal of this study was to determine the extent to which N/OFQ and NOP receptor levels change following mild (mTBI) and moderate TBI (modTBI) and whether the absence of the NOP receptor attenuates TBI-induced sequelae. Male and female NOP receptor knockout (KO) or wild-type (WT) rats received craniotomy-only (sham) or craniotomy plus mTBI, or modTBI impact to the left cerebral hemisphere. Neurologic and vestibulomotor deficits and nociceptive hyperalgesia and allodynia found in WT male and female rats following mTBI and modTBI were greatly reduced or absent in NOP receptor KO rats. NOP receptor levels increased in brain tissue from injured males but remained unchanged in females. Neurofilament light chain (NF-L) and glial fibrillary acidic protein (GFAP) expression were reduced in NOP receptor KO rats compared with WT following TBI. Levels of N/OFQ in injured brain tissue correlated with neurobehavioral outcomes and GFAP in WT males, but not with KO male or WT and KO female rats. This study reveals a significant contribution of the N/OFQ-NOP receptor system to TBI-induced deficits and suggests that the NOP receptor should be regarded as a potential therapeutic target for TBI. SIGNIFICANCE STATEMENT: This study revealed that nociceptin/orphanin FQ peptide (NOP) receptor knockout animals experienced fewer traumatic brain injury (TBI)-induced deficits than their wild-type counterparts in a sex- and injury severity-dependent manner, suggesting that NOP receptor antagonists may be a potential therapy for TBI.
Collapse
Affiliation(s)
- Omar N Al Yacoub
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy (O.N.A., H.O.A., K.M.S.), and the Neuroscience Program (K.M.S., H.O.A.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hibah O Awwad
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy (O.N.A., H.O.A., K.M.S.), and the Neuroscience Program (K.M.S., H.O.A.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kelly M Standifer
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy (O.N.A., H.O.A., K.M.S.), and the Neuroscience Program (K.M.S., H.O.A.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
2
|
Mohammed FS, Omay SB, Sheth KN, Zhou J. Nanoparticle-based drug delivery for the treatment of traumatic brain injury. Expert Opin Drug Deliv 2023; 20:55-73. [PMID: 36420918 PMCID: PMC9983310 DOI: 10.1080/17425247.2023.2152001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/10/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Traumatic brain injuries (TBIs) impact the breadth of society and remain without any approved pharmacological treatments. Despite successful Phase II clinical trials, the failure of many Phase III clinical trials may be explained by insufficient drug targeting and retention, preventing the proper attainment of an observable dosage threshold. To address this challenge, nanoparticles can be functionalized to protect pharmacological payloads, improve targeted drug delivery to sites of injury, and can be combined with supportive scaffolding to improve secondary outcomes. AREAS COVERED This review briefly covers the pathophysiology of TBIs and their subtypes, the current pre-clinical and clinical management strategies, explores the common models of focal, diffuse, and mixed traumatic brain injury employed in experimental animals, and surveys the existing literature on nanoparticles developed to treat TBIs. EXPERT OPINION Nanoparticles are well suited to improve secondary outcomes as their multifunctionality and customizability enhance their potential for efficient targeted delivery, payload protection, increased brain penetration, low off-target toxicity, and biocompatibility in both acute and chronic timescales.
Collapse
Affiliation(s)
- Farrah S. Mohammed
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Sacit Bulent Omay
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
| | - Kevin N. Sheth
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
- Department of Neurology, Yale University, New Haven, Connecticut, USA
| | - Jiangbing Zhou
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
3
|
Cralley AL, Moore EE, Fox CJ, Kissau D, DeBot M, Schaid TR, Mitra S, Hom P, Fragoso M, Ghasabyan A, Erickson C, D'Alessandro A, Hansen KC, Cohen MJ, Silliman CC, Sauaia A. Zone 1 REBOA in a combat DCBI swine model does not worsen brain injury. Surgery 2022; 172:751-758. [PMID: 35690490 PMCID: PMC9675949 DOI: 10.1016/j.surg.2022.04.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/29/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Zone 1 resuscitative endovascular balloon occlusion of the aorta has been recommended for refractory shock after a dismounted complex blast injury for the austere combat scenario. While resuscitative endovascular balloon occlusion of the aorta should enhance coronary perfusion, there is a potential risk of secondary brain injury due to loss of cerebral autoregulation. We developed a combat casualty relevant dismounted complex blast injury swine model to evaluate the effects of resuscitative endovascular balloon occlusion of the aorta zone I on intracranial pressure and cerebral edema. We hypothesized that zone 1 aortic occlusion with resuscitative endovascular balloon occlusion of the aorta would increase mean arterial pressure transmitted in excessive intracranial pressure, thereby worsening brain injury. METHODS 50 kg male Yorkshire swine were subjected to a combination dismounted complex blast injury model consisting of blast traumatic brain injury (50 psi, ARA Mobile Shock Laboratory), tissue injury (bilateral femur fractures), and hemorrhagic shock (controlled bleeding to a base deficit goal of 10 mEq/L). During the shock phase, pigs were randomized to no aortic occlusion (n = 8) or to 30 minutes of zone 1 resuscitative endovascular balloon occlusion of the aorta (zone 1 aortic occlusion group, n = 6). After shock, pigs in both groups received a modified Tactical Combat Casualty Care-based resuscitation and were monitored for an additional 240 minutes until euthanasia/death for a total of 6 hours. Intracranial pressure was monitored throughout, and brains were harvested for water content. Linear mixed models for repeated measures were used to compare mean arterial pressure and intracranial pressure between zone 1 aortic occlusion and no aortic occlusion groups. RESULTS After dismounted complex blast injury, the zone 1 group had a significantly higher mean arterial pressure during hemorrhagic shock compared to the control group (41.2 mm Hg vs 16.7 mm Hg, P = .002). During balloon occlusion, intracranial pressure was not significantly elevated in the zone 1 aortic occlusion group vs control, but intracranial pressure was significantly lower in the zone 1 group at the end of the observation period. In addition, the zone 1 aortic occlusion group did not have increased brain water content (zone 1 aortic occlusion: 3.95 ± 0.1g vs no aortic occlusion: 3.95 ± 0.3 g, P = .87). Troponin levels significantly increased in the no aortic occlusion group but did not in the zone 1 aortic occlusion group. CONCLUSION Zone 1 aortic occlusion using resuscitative endovascular balloon occlusion of the aorta in a large animal dismounted complex blast injury model improved proximal mean arterial pressure while not significantly increasing intracranial pressure during balloon inflation. Observation up to 240 minutes postresuscitation did not show clinical signs of worsening brain injury or cardiac injury. These data suggest that in a dismounted complex blast injury swine model, resuscitative endovascular balloon occlusion of the aorta in zone 1 may provide neuro- and cardioprotection in the setting of blast traumatic brain injury. However, longer monitoring periods may be needed to confirm that the neuroprotection is lasting.
Collapse
Affiliation(s)
| | - Ernest E Moore
- Department of Surgery, University of Colorado, Aurora, CO; Ernest E. Moore Shock Trauma Center at Denver Health, CO
| | - Charles J Fox
- Department of Vascular Surgery, University of Maryland Vascular Surgery Baltimore, MD
| | - Daniel Kissau
- Department of Surgery, University of Colorado, Aurora, CO
| | - Margot DeBot
- Department of Surgery, University of Colorado, Aurora, CO
| | - Terry R Schaid
- Department of Surgery, University of Colorado, Aurora, CO
| | | | - Patrick Hom
- Department of Surgery, University of Colorado, Aurora, CO
| | - Miguel Fragoso
- Department of Surgery, University of Colorado, Aurora, CO
| | | | - Christopher Erickson
- Department of Vascular Surgery, University of Maryland Vascular Surgery Baltimore, MD
| | - Angelo D'Alessandro
- Department of Proteomics and Metabolomics, University of Colorado, Aurora, CO
| | - Kirk C Hansen
- Department of Vascular Surgery, University of Maryland Vascular Surgery Baltimore, MD; Department of Proteomics and Metabolomics, University of Colorado, Aurora, CO
| | | | - Christopher C Silliman
- Department of Pediatrics, University of Colorado, Aurora, CO; Vitalant Research Institute, Denver, CO
| | - Angela Sauaia
- Department of Health Systems, Management and Policy, School of Public Health, University of Colorado Denver, Aurora, CO
| |
Collapse
|
4
|
Abstract
BACKGROUND Improvised explosive devices have resulted in a unique polytrauma injury pattern termed dismounted complex blast injury (DCBI), which is frequent in the modern military theater. Dismounted complex blast injury is characterized by extremity amputations, junctional vascular injury, and blast traumatic brain injury (bTBI). We developed a combat casualty relevant DCBI swine model, which combines hemorrhagic shock (HS) and tissue injury (TI) with a bTBI, to study interventions in this unique and devastating military injury pattern. METHODS A 50-kg male Yorkshire swine were randomized to the DCBI or SHAM group (instrumentation only). Those in the DCBI group were subjected to HS, TI, and bTBI. The blast injury was applied using a 55-psi shock tube wave. Tissue injury was created with bilateral open femur fractures. Hemorrhagic shock was induced by bleeding from femoral arteries to target pressure. A resuscitation protocol modified from the Tactical Combat Casualty Care guidelines simulated battlefield resuscitation for 240 minutes. RESULTS Eight swine underwent the DCBI model and five were allocated to the SHAM group. In the DCBI model the mean base excess achieved at the end of the HS shock was -8.57 ± 5.13 mmol·L -1 . A significant coagulopathy was detected in the DCBI model as measured by prothrombin time (15.8 seconds DCBI vs. 12.86 seconds SHAM; p = 0.02) and thromboelastography maximum amplitude (68.5 mm DCBI vs. 78.3 mm in SHAM; p = 0.0003). For the DCBI models, intracranial pressure (ICP) increased by a mean of 13 mm Hg, reaching a final ICP of 24 ± 7.7 mm Hg. CONCLUSION We created a reproducible large animal model to study the combined effects of severe HS, TI, and bTBI on coagulation and ICP in the setting of DCBI, with significant translational applications for the care of military warfighters. Within the 4-hour observational period, the swine developed a consistent coagulopathy with a concurrent brain injury evidenced by increasing ICP.
Collapse
|
5
|
Menichetti A, Bartsoen L, Depreitere B, Vander Sloten J, Famaey N. A Machine Learning Approach to Investigate the Uncertainty of Tissue-Level Injury Metrics for Cerebral Contusion. Front Bioeng Biotechnol 2021; 9:714128. [PMID: 34692652 PMCID: PMC8531645 DOI: 10.3389/fbioe.2021.714128] [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: 05/24/2021] [Accepted: 08/10/2021] [Indexed: 11/13/2022] Open
Abstract
Controlled cortical impact (CCI) on porcine brain is often utilized to investigate the pathophysiology and functional outcome of focal traumatic brain injury (TBI), such as cerebral contusion (CC). Using a finite element (FE) model of the porcine brain, the localized brain strain and strain rate resulting from CCI can be computed and compared to the experimentally assessed cortical lesion. This way, tissue-level injury metrics and corresponding thresholds specific for CC can be established. However, the variability and uncertainty associated with the CCI experimental parameters contribute to the uncertainty of the provoked cortical lesion and, in turn, of the predicted injury metrics. Uncertainty quantification via probabilistic methods (Monte Carlo simulation, MCS) requires a large number of FE simulations, which results in a time-consuming process. Following the recent success of machine learning (ML) in TBI biomechanical modeling, we developed an artificial neural network as surrogate of the FE porcine brain model to predict the brain strain and the strain rate in a computationally efficient way. We assessed the effect of several experimental and modeling parameters on four FE-derived CC injury metrics (maximum principal strain, maximum principal strain rate, product of maximum principal strain and strain rate, and maximum shear strain). Next, we compared the in silico brain mechanical response with cortical damage data from in vivo CCI experiments on pig brains to evaluate the predictive performance of the CC injury metrics. Our ML surrogate was capable of rapidly predicting the outcome of the FE porcine brain undergoing CCI. The now computationally efficient MCS showed that depth and velocity of indentation were the most influential parameters for the strain and the strain rate-based injury metrics, respectively. The sensitivity analysis and comparison with the cortical damage experimental data indicate a better performance of maximum principal strain and maximum shear strain as tissue-level injury metrics for CC. These results provide guidelines to optimize the design of CCI tests and bring new insights to the understanding of the mechanical response of brain tissue to focal traumatic brain injury. Our findings also highlight the potential of using ML for computationally efficient TBI biomechanics investigations.
Collapse
Affiliation(s)
- Andrea Menichetti
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Laura Bartsoen
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | | | - Jos Vander Sloten
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Nele Famaey
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
De Kegel D, Musigazi GU, Menichetti A, Hellings PW, Sciot R, Demaerel P, Famaey N, Vander Sloten J, Depreitere B. Investigation of tissue level tolerance for cerebral contusion in a controlled cortical impact porcine model. TRAFFIC INJURY PREVENTION 2021; 22:616-622. [PMID: 34477471 DOI: 10.1080/15389588.2021.1957856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE Cerebral contusions (CC) represent a frequent lesion in traumatic brain injury, with potential morbidity from mass effect and tissue loss. Better understanding of the mechanical etiology will help to improve head protection. The goal of this study is to investigate the threshold for mechanical impact parameters to induce CC in an in vivo porcine controlled cortical impact model. METHODS Thirty-four adult male pigs underwent craniotomy and controlled cortical impact with a hemispherical tip on intact dura under general anesthesia. Peak impact depth varied between 1.1 and 12.6 mm, and impact velocity between 0.4 and 2.2 m/s while the dwell time was kept at 200 ms. Two days following impact, the animals underwent magnetic resonance (MR) imaging of the brain, and were subsequently sacrificed for brain extraction. CC damage was investigated by magnetic resonance imaging and histology. RESULTS All animals recovered from the impact without overt neurological deficit. Provoked injuries were histologically confirmed to be CC. Decreasing probability of cortical damage and white matter edema volume was observed with decreasing impact depth and velocity. No CC could be demonstrated below a product of impact depth and velocity of 0.8 mm*m/s, whereas the probability for CC was one third below 15 mm*m/s. The threshold for CC development as estimated from the current series of experiments, was situated at an impact depth of 2.0 mm and impact velocity of 0.4 m/s. CONCLUSION Mechanical thresholds for CC development could be explored in the current porcine controlled cortical impact model. Findings will be used to further refine a cerebral contusion porcine model with volumetric histology data in light of future finite element cerebral contusion validation studies.
Collapse
Affiliation(s)
| | | | | | | | - Raf Sciot
- Translation Cell and Tissue Research, KULeuven, Leuven, Belgium
| | | | - Nele Famaey
- Biomechanics section, KULeuven, Heverlee, Belgium
| | | | - Bart Depreitere
- Experimental Neurosurgery and Neuroanatomy, KULeuven, Leuven, Belgium
| |
Collapse
|
7
|
Tatara Y, Shimada R, Kibayashi K. Effects of Preexisting Diabetes Mellitus on the Severity of Traumatic Brain Injury. J Neurotrauma 2020; 38:886-902. [PMID: 32998635 DOI: 10.1089/neu.2020.7118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Falls and traffic accidents can cause traumatic brain injury (TBI). Assessment of the injury severity is essential to determine the prognosis or the cause of death. Diabetes mellitus (DM) is a common preexisting disease in elderly adults. We hypothesized that preexisting DM exacerbates TBI secondary to prolonged inflammation. In this study, we investigated TBI-induced changes in nerve function and inflammatory cell migration to the injury site, and the extent of brain contusion in KK-Ay (DM) and C57BL/6J (non-DM) mice. A controlled cortical impact device was used to induce TBI in each mouse. The brain contusion volume was measured using magnetic resonance imaging. Nerve function changes were assessed using the following animal behavior tasks: neurological severity score (NSS), Morris water maze, forced swim test, and beam walking. Immunohistochemical examinations of brain sections were performed to assess the infiltration of neutrophils, astrocytes, microglia, and macrophages, and to detect apoptosis. These experiments were performed on post-injury days 1-90 (over five experiments/time-points in each group). Compared with non-DM mice, DM mice showed significantly greater brain contusion volume, greater deterioration in the NSS, and a higher number of neutrophils, macrophages, and apoptotic cells in the brain tissue specimens. This study indicates that the prognosis of normal mice and DM mice differs, even if they acquire a TBI of the same severity. Therefore, it is important to evaluate patients with TBI for DM and other preexisting diseases in order to provide adequate treatment or to determine the correct cause of death.
Collapse
Affiliation(s)
- Yuki Tatara
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Ryo Shimada
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuhiko Kibayashi
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
8
|
Falnikar A, Stratton J, Lin R, Andrews CE, Tyburski A, Trovillion VA, Gottschalk C, Ghosh B, Iacovitti L, Elliott MB, Lepore AC. Differential Response in Novel Stem Cell Niches of the Brain after Cervical Spinal Cord Injury and Traumatic Brain Injury. J Neurotrauma 2018; 35:2195-2207. [PMID: 29471717 DOI: 10.1089/neu.2017.5497] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Populations of neural stem cells (NSCs) reside in a number of defined niches in the adult central nervous system (CNS) where they continually give rise to mature cell types throughout life, including newly born neurons. In addition to the prototypical niches of the subventricular zone (SVZ) and subgranular zone (SGZ) of the hippocampal dentate gyrus, novel stem cell niches that are also neurogenic have recently been identified in multiple midline structures, including circumventricular organs (CVOs) of the brain. These resident NSCs serve as a homeostatic source of new neurons and glial cells under intact physiological conditions. Importantly, they may also have the potential for reparative processes in pathological states such as traumatic spinal cord injury (SCI) and traumatic brain injury (TBI). As the response in these novel CVO stem cell niches has been characterized after stroke but not following SCI or TBI, we quantitatively assessed cell proliferation and the neuronal and glial lineage fate of resident NSCs in three CVO nuclei-area postrema (AP), median eminence (ME), and subfornical organ (SFO) -in rat models of cervical contusion-type SCI and controlled cortical impact (CCI)-induced TBI. Using bromodeoxyuridine (BrdU) labeling of proliferating cells, we find that TBI significantly enhanced proliferation in AP, ME, and SFO, whereas cervical SCI had no effects at early or chronic time-points post-injury. In addition, SCI did not alter NSC differentiation profile into doublecortin-positive neuroblasts, GFAP-expressing astrocytes, or Olig2-labeled cells of the oligodendrocyte lineage within AP, ME, or SFO at both time-points. In contrast, CCI induced a pronounced increase in Sox2- and doublecortin-labeled cells in the AP and Iba1-labeled microglia in the SFO. Lastly, plasma derived from CCI animals significantly increased NSC expansion in an in vitro neurosphere assay, whereas plasma from SCI animals did not exert such an effect, suggesting that signaling factors present in blood may be relevant to stimulating CVO niches after CNS injury and may explain the differential in vivo effects of SCI and TBI on the novel stem cell niches.
Collapse
Affiliation(s)
- Aditi Falnikar
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Jarred Stratton
- 2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ruihe Lin
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Carrie E Andrews
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ashley Tyburski
- 2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Victoria A Trovillion
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Chelsea Gottschalk
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Biswarup Ghosh
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Lorraine Iacovitti
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Melanie B Elliott
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania.,2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Angelo C Lepore
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
9
|
Liang J, Yokell ZA, Nakmaili DU, Gan RZ, Lu H. The effect of blast overpressure on the mechanical properties of a chinchilla tympanic membrane. Hear Res 2017; 354:48-55. [DOI: 10.1016/j.heares.2017.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 07/30/2017] [Accepted: 08/15/2017] [Indexed: 10/19/2022]
|
10
|
Lafuente JV, Sharma A, Muresanu DF, Ozkizilcik A, Tian ZR, Patnaik R, Sharma HS. Repeated Forced Swim Exacerbates Methamphetamine-Induced Neurotoxicity: Neuroprotective Effects of Nanowired Delivery of 5-HT3-Receptor Antagonist Ondansetron. Mol Neurobiol 2017; 55:322-334. [DOI: 10.1007/s12035-017-0744-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
11
|
Semple BD, Sadjadi R, Carlson J, Chen Y, Xu D, Ferriero DM, Noble-Haeusslein LJ. Long-Term Anesthetic-Dependent Hypoactivity after Repetitive Mild Traumatic Brain Injuries in Adolescent Mice. Dev Neurosci 2016; 38:220-238. [PMID: 27548472 DOI: 10.1159/000448089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/28/2016] [Indexed: 11/19/2022] Open
Abstract
Recent evidence supports the hypothesis that repetitive mild traumatic brain injuries (rmTBIs) culminate in neurological impairments and chronic neurodegeneration, which have wide-ranging implications for patient management and return-to-play decisions for athletes. Adolescents show a high prevalence of sports-related head injuries and may be particularly vulnerable to rmTBIs due to ongoing brain maturation. However, it remains unclear whether rmTBIs, below the threshold for acute neuronal injury or symptomology, influence long-term outcomes. To address this issue, we first defined a very mild injury in adolescent mice (postnatal day 35) as evidenced by an increase in Iba-1- labeled microglia in white matter in the acutely injured brain, in the absence of indices of cell death, axonal injury, and vasogenic edema. Using this level of injury severity and Avertin (2,2,2-tribromoethanol) as the anesthetic, we compared mice subjected to either a single mTBI or 2 rmTBIs, each separated by 48 h. Neurobehavioral assessments were conducted at 1 week and at 1 and 3 months postimpact. Mice subjected to rmTBIs showed transient anxiety and persistent and pronounced hypoactivity compared to sham control mice, alongside normal sensorimotor, cognitive, social, and emotional function. As isoflurane is more commonly used than Avertin in animal models of TBI, we next examined long-term outcomes after rmTBIs in mice that were anesthetized with this agent. However, there was no evidence of abnormal behaviors even with the addition of a third rmTBI. To determine whether isoflurane may be neuroprotective, we compared the acute pathology after a single mTBI in mice anesthetized with either Avertin or isoflurane. Pathological findings were more pronounced in the group exposed to Avertin compared to the isoflurane group. These collective findings reveal distinct behavioral phenotypes (transient anxiety and prolonged hypoactivity) that emerge in response to rmTBIs. Our findings further suggest that selected anesthetics may confer early neuroprotection after rmTBIs, and as such mask long-term abnormal phenotypes that may otherwise emerge as a consequence of acute pathogenesis.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neurological Surgery, University of California San Francisco, San Francisco, Calif., USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Yousuf MA, Tan C, Torres-Altoro MI, Lu FM, Plautz E, Zhang S, Takahashi M, Hernandez A, Kernie SG, Plattner F, Bibb JA. Involvement of aberrant cyclin-dependent kinase 5/p25 activity in experimental traumatic brain injury. J Neurochem 2016; 138:317-27. [PMID: 26998748 DOI: 10.1111/jnc.13620] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/02/2016] [Accepted: 03/14/2016] [Indexed: 11/27/2022]
Abstract
Traumatic brain injury (TBI) is associated with adverse effects on brain functions, including sensation, language, emotions and/or cognition. Therapies for improving outcomes following TBI are limited. A better understanding of the pathophysiological mechanisms of TBI may suggest novel treatment strategies to facilitate recovery and improve treatment outcome. Aberrant activation of cyclin-dependent kinase 5 (Cdk5) has been implicated in neuronal injury and neurodegeneration. Cdk5 is a neuronal protein kinase activated via interaction with its cofactor p35 that regulates numerous neuronal functions, including synaptic remodeling and cognition. However, conversion of p35 to p25 via Ca(2+) -dependent activation of calpain results in an aberrantly active Cdk5/p25 complex that is associated with neuronal damage and cell death. Here, we show that mice subjected to controlled cortical impact (CCI), a well-established experimental TBI model, exhibit increased p25 levels and consistently elevated Cdk5-dependent phosphorylation of microtubule-associated protein tau and retinoblastoma (Rb) protein in hippocampal lysates. Moreover, CCI-induced neuroinflammation as indicated by increased astrocytic activation and number of reactive microglia. Brain-wide conditional Cdk5 knockout mice (Cdk5 cKO) subjected to CCI exhibited significantly reduced edema, ventricular dilation, and injury area. Finally, neurophysiological recordings revealed that CCI attenuated excitatory post-synaptic potential field responses in the hippocampal CA3-CA1 pathway 24 h after injury. This neurophysiological deficit was attenuated in Cdk5 cKO mice. Thus, TBI induces increased levels of p25 generation and aberrant Cdk5 activity, which contributes to pathophysiological processes underlying TBI progression. Hence, selectively preventing aberrant Cdk5 activity may be an effective acute strategy to improve recovery from TBI. Traumatic brain injury (TBI) increases astrogliosis and microglial activation. Moreover, TBI deregulates Ca(2+) -homeostasis triggering p25 production. The protein kinase Cdk5 is aberrantly activated by p25 leading to phosphorylation of substrates including tau and Rb protein. Loss of Cdk5 attenuates TBI lesion size, indicating that Cdk5 is a critical player in TBI pathogenesis and thus may be a suitable therapeutic target for TBI.
Collapse
Affiliation(s)
- Mohammad A Yousuf
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chunfeng Tan
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Melissa I Torres-Altoro
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Fang-Min Lu
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Erik Plautz
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shanrong Zhang
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Masaya Takahashi
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Adan Hernandez
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Steven G Kernie
- Department of Pediatrics and Pathology & Cell Biology, Columbia University, New York, New York, USA
| | - Florian Plattner
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - James A Bibb
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
13
|
Characterization of Closed Head Impact Injury in Rat. BIOMED RESEARCH INTERNATIONAL 2015; 2015:272976. [PMID: 26451365 PMCID: PMC4588353 DOI: 10.1155/2015/272976] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 03/10/2015] [Indexed: 02/06/2023]
Abstract
The closed head impact (CHI) rat models are commonly used for studying the traumatic brain injury. The impact parameters vary considerably among different laboratories, making the comparison of research findings difficult. In this work, numerical CHI experiments were conducted to investigate the sensitivities of intracranial responses to various impact parameters (e.g., impact depth, velocity, and position; impactor diameter, material, and shape). A three-dimensional finite element rat head model with anatomical details was subjected to impact loadings. Results revealed that impact depth and impactor shape were the two leading factors affecting intracranial responses. The influence of impactor diameter was region-specific and an increase in impactor diameter could substantially increase tissue strains in the region which located directly beneath the impactor. The lateral impact could induce higher strains in the brain than the central impact. An indentation depth instead of impact depth would be appropriate to characterize the influence of a large deformed rubber impactor. The experimentally observed velocity-dependent injury severity could be attributed to the “overshoot” phenomenon. This work could be used to better design or compare CHI experiments.
Collapse
|
14
|
Semple BD, Trivedi A, Gimlin K, Noble-Haeusslein LJ. Neutrophil elastase mediates acute pathogenesis and is a determinant of long-term behavioral recovery after traumatic injury to the immature brain. Neurobiol Dis 2014; 74:263-80. [PMID: 25497734 DOI: 10.1016/j.nbd.2014.12.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 11/19/2014] [Accepted: 12/01/2014] [Indexed: 12/21/2022] Open
Abstract
While neutrophil elastase (NE), released by activated neutrophils, is a key mediator of secondary pathogenesis in adult models of brain ischemia and spinal cord injury, no studies to date have examined this protease in the context of the injured immature brain, where there is notable vulnerability resulting from inadequate antioxidant reserves and prolonged exposure to infiltrating neutrophils. We thus reasoned that NE may be a key determinant of secondary pathogenesis, and as such, adversely influence long-term neurological recovery. To address this hypothesis, wild-type (WT) and NE knockout (KO) mice were subjected to a controlled cortical impact at post-natal day 21, approximating a toddler-aged child. To determine if NE is required for neutrophil infiltration into the injured brain, and whether this protease contributes to vasogenic edema, we quantified neutrophil numbers and measured water content in the brains of each of these genotypes. While leukocyte trafficking was indistinguishable between genotypes, vasogenic edema was markedly attenuated in the NE KO. To determine if early pathogenesis is dependent on NE, indices of cell death (TUNEL and activated caspase-3) were quantified across genotypes. NE KO mice showed a reduction in these markers of cell death in the injured hippocampus, which corresponded to greater preservation of neuronal integrity as well as reduced expression of heme oxygenase-1, a marker of oxidative stress. WT mice, treated with a competitive inhibitor of NE at 2, 6 and 12h post-injury, likewise showed a reduction in cell death and oxidative stress compared to vehicle-treated controls. We next examined the long-term behavioral and structural consequences of NE deficiency. NE KO mice showed an improvement in long-term spatial memory retention and amelioration of injury-induced hyperactivity. However, volumetric and stereological analyses found comparable tissue loss in the injured cortex and hippocampus independent of genotype. Further, WT mice treated acutely with the NE inhibitor showed no long-term behavioral or structural improvements. Together, these findings validate the central role of NE in both acute pathogenesis and chronic functional recovery, and support future exploration of the therapeutic window, taking into account the prolonged period of neutrophil trafficking into the injured immature brain.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA; Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC 3000, Australia.
| | - Alpa Trivedi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Kayleen Gimlin
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Linda J Noble-Haeusslein
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA; Department of Physical Therapy and Rehabilitation Sciences, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
15
|
Xu ZS, Yao A, Chu SS, Paun MK, McClintic AM, Murphy SP, Mourad PD. Detection of mild traumatic brain injury in rodent models using shear wave elastography: preliminary studies. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2014; 33:1763-1771. [PMID: 25253822 DOI: 10.7863/ultra.33.10.1763] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
OBJECTIVES Traumatic brain injury (TBI) can cause adverse physiologic changes in fluid content within the brain, which may lead to changes in tissue elasticity (eg, stiffness). This study evaluated the ability of ultrasonic shear wave elastography to observe these changes in the brain after TBI in vivo. METHODS Mice and rats received a mild TBI or sham surgery and were imaged acutely or 24 hours after injury using shear wave elastography, and the hemispheric stiffness values were compared. RESULTS Stiffness values were consistent across brain hemispheres of sham TBI rodents. By 24 hours after TBI, relative brain tissue stiffness values for mice and rats each decreased ipsilaterally and increased contralaterally, both relative to each other and compared to sham TBI rodents (P < .05). The absolute tissue elasticity value increased for rats (P < .05) but not for mice. CONCLUSIONS Differences between intrahemispheric stiffness values of rodent brains by 24 hours after mild TBI may reflect the observed edema and hemorrhage ipsilateral to TBI and the known reduction of cerebral blood flow in both brain hemispheres. If these hypotheses hold true, ultrasonic shear wave elastography may offer a method for detecting adverse changes in fluid content within the brain after mild TBI.
Collapse
Affiliation(s)
- Zinnia S Xu
- Department of Bioengineering (Z.S.X., A.Y., S.S.C., P.D.M.), Applied Physics Laboratory (M.K.P., P.D.M.), and Department of Neurological Surgery (A.M.M., S.P.M., P.D.M.), University of Washington, Seattle, Washington USA
| | - Anning Yao
- Department of Bioengineering (Z.S.X., A.Y., S.S.C., P.D.M.), Applied Physics Laboratory (M.K.P., P.D.M.), and Department of Neurological Surgery (A.M.M., S.P.M., P.D.M.), University of Washington, Seattle, Washington USA
| | - Stephanie S Chu
- Department of Bioengineering (Z.S.X., A.Y., S.S.C., P.D.M.), Applied Physics Laboratory (M.K.P., P.D.M.), and Department of Neurological Surgery (A.M.M., S.P.M., P.D.M.), University of Washington, Seattle, Washington USA
| | - Marla K Paun
- Department of Bioengineering (Z.S.X., A.Y., S.S.C., P.D.M.), Applied Physics Laboratory (M.K.P., P.D.M.), and Department of Neurological Surgery (A.M.M., S.P.M., P.D.M.), University of Washington, Seattle, Washington USA
| | - Abbi M McClintic
- Department of Bioengineering (Z.S.X., A.Y., S.S.C., P.D.M.), Applied Physics Laboratory (M.K.P., P.D.M.), and Department of Neurological Surgery (A.M.M., S.P.M., P.D.M.), University of Washington, Seattle, Washington USA
| | - Sean P Murphy
- Department of Bioengineering (Z.S.X., A.Y., S.S.C., P.D.M.), Applied Physics Laboratory (M.K.P., P.D.M.), and Department of Neurological Surgery (A.M.M., S.P.M., P.D.M.), University of Washington, Seattle, Washington USA
| | - Pierre D Mourad
- Department of Bioengineering (Z.S.X., A.Y., S.S.C., P.D.M.), Applied Physics Laboratory (M.K.P., P.D.M.), and Department of Neurological Surgery (A.M.M., S.P.M., P.D.M.), University of Washington, Seattle, Washington USA.
| |
Collapse
|
16
|
Macolino CM, Daiutolo BV, Albertson BK, Elliott MB. Mechanical allodynia induced by traumatic brain injury is independent of restraint stress. J Neurosci Methods 2014; 226:139-146. [PMID: 24486873 DOI: 10.1016/j.jneumeth.2014.01.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/20/2013] [Accepted: 01/13/2014] [Indexed: 01/28/2023]
Abstract
BACKGROUND This study identifies the relationship between a test for post-traumatic headache and a marker for acute stress in rodent models of traumatic brain injury. NEW METHOD C57BL/6 mice and Sprague Dawley rats were divided into Controlled Cortical Impact (CCI) injury, craniotomy (CR), and incision groups. Periorbital and paw allodynia were evaluated using the von Frey test prior to injury and up to four weeks post-operatively. Serum corticosterone was evaluated in groups with and without mild restraint. RESULTS Periorbital and forepaw thresholds, but not hindpaw thresholds, were reduced in CCI and CR mice compared to incision (p<0.0001 and p<0.01). In contrast to mice, reduced periorbital and forepaw periorbital thresholds were found in CCI rats but not CR rats compared to incision (p<0.0001). Right periorbital thresholds were reduced compared to left thresholds for both rat and mouse at one week (p<0.01), but there were no side differences for forepaw thresholds. Hindpaw thresholds did not change from baseline values for any groups of mice or rats. In mice serum corticosterone levels were increased at one, two and four weeks post-CCI and CR, while the levels for rats were not different from incision (p<0.0001). Corticosterone levels were not different in mice subjected to restraint compared to no restraint. COMPARISON WITH EXISTING METHODS This study presents novel data for allodynia in a rat model of TBI, and differences among mouse and rat species. CONCLUSIONS Mechanical allodynia occurs independent of evoked restraint stress, while hypothalamic pituitary adrenal axis activity is dependent on head trauma and species.
Collapse
Affiliation(s)
- Christine M Macolino
- Department of Neurological Surgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | - Brittany V Daiutolo
- Department of Neurological Surgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | - Brad K Albertson
- Jefferson Medical College, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | - Melanie B Elliott
- Department of Neuroscience, Jefferson Hospital for Neuroscience Chairs Office 900 Walnut Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
17
|
Tchantchou F, Zhang Y. Selective inhibition of alpha/beta-hydrolase domain 6 attenuates neurodegeneration, alleviates blood brain barrier breakdown, and improves functional recovery in a mouse model of traumatic brain injury. J Neurotrauma 2013; 30:565-79. [PMID: 23151067 DOI: 10.1089/neu.2012.2647] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
2-arachidonylglycerol (2-AG) is the most abundant endocannabinoid in the central nervous system and is elevated after brain injury. Because of its rapid hydrolysis, however, the compensatory and neuroprotective effect of 2-AG is short-lived. Although inhibition of monoacylglycerol lipase, a principal enzyme for 2-AG degradation, causes a robust increase of brain levels of 2-AG, it also leads to cannabinoid receptor desensitization and behavioral tolerance. Alpha/beta hydrolase domain 6 (ABHD6) is a novel 2-AG hydrolytic enzyme that accounts for a small portion of 2-AG hydrolysis, but its inhibition is believed to elevate the levels of 2-AG within the therapeutic window without causing side effect. Using a mouse model of traumatic brain injury (TBI), we found that post-insult chronic treatment with a selective ABHD6 inhibitor WWL70 improved motor coordination and working memory performance. WWL70 treatment reduced lesion volume in the cortex and neurodegeneration in the dendate gyrus. It also suppressed the expression of inducible nitric oxide synthase and cyclooxygenase-2 and enhanced the expression of arginase-1 in the ipsilateral cortex at 3 and 7 days post-TBI, suggesting microglia/macrophages shifted from M1 to M2 phenotypes after treatment. The blood-brain barrier dysfunction at 3 and 7 days post-TBI was dramatically reduced. Furthermore, the beneficial effects of WWL70 involved up-regulation and activation of cannabinoid type 1 and type 2 receptors and were attributable to the phosphorylation of the extracellular signal regulated kinase and the serine/threonine protein kinase AKT. This study indicates that the fine-tuning of 2-AG signaling by modulating ABHD6 activity can exert anti-inflammatory and neuroprotective effects in TBI.
Collapse
Affiliation(s)
- Flaubert Tchantchou
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20892, USA
| | | |
Collapse
|
18
|
Argenta LC, Zheng Z, Bryant A, Tatter SB, Morykwas MJ. A new method for modulating traumatic brain injury with mechanical tissue resuscitation. Neurosurgery 2012; 70:1281-95. [PMID: 22157550 DOI: 10.1227/neu.0b013e3182446760] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Traumatic brain injuries remain a treatment enigma with devastating late results. As terminally differentiated tissue, the brain retains little capacity to regenerate, making early attempts to preserve brain cells after brain injury essential. OBJECTIVE To resuscitate damaged tissue by modulating edema, soluble cytokines, and metabolic products in the "halo" of damaged tissue around the area of central injury that progressively becomes compromised. By re-equilibrating the zone of injury milieu, it is postulated neurons in this area will survive and function. METHODS Mechanical tissue resuscitation used localized, controlled, subatmospheric pressure directly to the area of controlled cortical impact injury and was compared with untreated injured controls and with sham surgery in a rat model. Functional outcome, T2 magnetic resonance imaging hyperintense volume, magnetic resonance imaging spectroscopy metabolite measurement, tissue water content, injury cavity area, and cortical volume were compared. RESULTS There were significant differences between mechanical tissue resuscitation treated and untreated groups in levels of myoinositol, N-acetylaspartate, and creatine. Treated animals had significantly less tissue swelling and density than the untreated animals. Nonviable brain tissue areas were smaller in treated animals than in untreated animals. Treated animals performed better than untreated animals in functional tests. Histological analysis showed the remaining viable ipsilateral cerebral area was 58% greater for treated animals than for untreated animals, and the cavity for treated animals was 95% smaller than for untreated animals 1 month after injury. CONCLUSION Mechanical tissue resuscitation with controlled subatmospheric pressure can significantly modulate levels of excitatory amino acids and lactate in traumatic brain injury, decrease the water content and volume of injured brain, improve neuronal survival, and speed functional recovery.
Collapse
Affiliation(s)
- Louis C Argenta
- Department of Plastic and Reconstructive Surgery, Wake Forest University Health Science, Winston-Salem, North Carolina 27157-1075, USA.
| | | | | | | | | |
Collapse
|
19
|
Elliott MB, Oshinsky ML, Amenta PS, Awe OO, Jallo JI. Nociceptive neuropeptide increases and periorbital allodynia in a model of traumatic brain injury. Headache 2012; 52:966-84. [PMID: 22568499 DOI: 10.1111/j.1526-4610.2012.02160.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE This study tests the hypothesis that injury to the somatosensory cortex is associated with periorbital allodynia and increases in nociceptive neuropeptides in the brainstem in a mouse model of controlled cortical impact (CCI) injury. METHODS Male C57BL/6 mice received either CCI or craniotomy-only followed by weekly periorbital von Frey (mechanical) sensory testing for up to 28 days post-injury. Mice receiving an incision only and naïve mice were included as control groups. Changes in calcitonin gene-related peptide (CGRP) and substance P (SP) within the brainstem were determined using enzyme-linked immunosorbent assay and immunohistochemistry, respectively. Activation of ionized calcium-binding adaptor molecule-1-labeled macrophages/microglia and glial fibrillary acidic protein (GFAP)-positive astrocytes were evaluated using immunohistochemistry because of their potential involvement in nociceptor sensitization. RESULTS Incision-only control mice showed no changes from baseline periorbital von Frey mechanical thresholds. CCI significantly reduced mean periorbital von Frey thresholds (periorbital allodynia) compared with baseline and craniotomy-only at each endpoint, analysis of variance P < .0001. Craniotomy significantly reduced periorbital threshold at 14 days but not 7, 21, or 28 days compared with baseline threshold, P < .01. CCI significantly increased SP immunoreactivity in the brainstem at 7 and 14 days but not 28 days compared with craniotomy-only and controls, P < .001. CGRP levels in brainstem tissues were significantly increased in CCI groups compared with controls (incision-only and naïve mice) or craniotomy-only mice at each endpoint examined, P < .0001. There was a significant correlation between CGRP and periorbital allodynia (P < .0001, r = -0.65) but not for SP (r = 0.20). CCI significantly increased the number of macrophage/microglia in the injured cortex at each endpoint up to 28 days, although cell numbers declined over weeks post-injury, P < .001. GFAP(+) immunoreactivity was significantly increased at 7 but not 14 or 28 days after CCI, P < .001. Craniotomy resulted in transient periorbital allodynia accompanied by transient increases in SP, CGRP, and GFAP immunoreactivity compared with control mice. There was no increase in the number of macrophage/microglia cells compared with controls after craniotomy. CONCLUSION Injury to the somatosensory cortex results in persistent periorbital allodynia and increases in brainstem nociceptive neuropeptides. Findings suggest that persistent allodynia and increased neuropeptides are maintained by mechanisms other than activation of macrophage/microglia or astrocyte in the injured somatosensory cortex.
Collapse
Affiliation(s)
- Melanie B Elliott
- Department of Neurological Surgery, Thomas Jefferson University, 1025 Walnut Street, Philadelphia, PA 19107, USA.
| | | | | | | | | |
Collapse
|
20
|
Elliott MB, Tuma RF, Amenta PS, Barbe MF, Jallo JI. Acute effects of a selective cannabinoid-2 receptor agonist on neuroinflammation in a model of traumatic brain injury. J Neurotrauma 2011; 28:973-81. [PMID: 21332427 DOI: 10.1089/neu.2010.1672] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Proposed therapeutic strategies for attenuating secondary traumatic brain injury (TBI) include modulation of acute neuroimmune responses. The goal of this study was to examine the acute effects of cannabinoid-2 receptor (CB(2)R) modulation on behavioral deficits, cerebral edema, perivascular substance P, and macrophage/microglial activation in a murine model of TBI. Thirty male C57BL/6 mice underwent sham surgery, or cortical contusion impact injury (CCI). CCI mice received vehicle or the CB(2)R agonist 0-1966 at 1 and 24 h after injury. Performance on the rotarod, forelimb cylinder, and open-field tests were evaluated before and at 48 h after sham or CCI surgery. Cerebral edema was evaluated using the wet-dry weight technique. Immunohistochemical analysis was used to examine changes in substance P and macrophage/microglia-specific Iba1 protein immunoreactivity. Locomotor performance and exploratory behavior were significantly improved in mice receiving 0-1966 (CB(2)R agonist) compared to vehicle-treated mice. Significant reductions were found for cerebral edema, number of perivascular areas of substance P immunoreactivity, and number of activated macrophages/microglial cells in the injured brains of 0-1966-treated mice compared to vehicle-treated mice. The findings show that the effects of the CB(2)R agonist 0-1966 on edema, substance P immunoreactivity, and macrophage/microglial activation, were associated with recovery of acute motor and exploratory deficits. This study provides evidence of acute neuroprotective effects derived from selective CB(2)R activation that may represent an avenue for further development of novel therapeutic agents in the treatment of TBI.
Collapse
Affiliation(s)
- Melanie B Elliott
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| | | | | | | | | |
Collapse
|
21
|
Levene HB, Elliott MB, Gaughan JP, Loftus CM, Tuma RF, Jallo JI. A murine model of hypertonic saline as a treatment for acute spinal cord injury: effects on autonomic outcome. J Neurosurg Spine 2011; 14:131-8. [DOI: 10.3171/2010.9.spine08314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Spinal cord injury (SCI) continues to be a problem without a definitive cure. Research based on improved understanding of the immunological aspects of SCI has revealed targets for treating and ameliorating the extent of secondary injury. Hypertonic saline (HTS), a substance both easy to create and to transport, has been investigated as an immunologically active material that can be used in a clinically relevant interval after injury. In this pilot study, HTS was investigated in a murine model for its abilities to ameliorate secondary injury after a severe spinal cord contusion.
Methods
Female C57Bl/6 mice with severe T8–10 contusion injuries were used as the model subjects. A group of 41 mice were studied in a blinded fashion. Mice received treatments with HTS (HTS, 7.5%) or normal saline solution (NSS, 0.9%) at 2 discreet time points (3 and 24 hours after injury.) A separate group of 9 untreated animals were also used as controls. Animals were assessed for autonomic outcome (bladder function). In a group of 33 mice, histological assessment (cellular infiltration) was also measured.
Results
Bladder function was found to be improved significantly in those treated with HTS compared with those who received NSS and also at later treatment times (24 hours) than at earlier treatment times (3 hours). Decreased cellular infiltration in each group correlated with bladder recovery.
Conclusions
The increased effectiveness of later administration time of the more osmotically active and immunomodulatory substance (HTS) suggests that interaction with events occurring around 24 hours after injury is critical. These events may be related to the invasion of leukocytes peaking at 8–24 hours postinjury and/or the peak benefit time of subject rehydration.
Collapse
Affiliation(s)
| | | | | | | | - Ronald F. Tuma
- 4Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | | |
Collapse
|
22
|
Mao H, Jin X, Zhang L, Yang KH, Igarashi T, Noble-Haeusslein LJ, King AI. Finite element analysis of controlled cortical impact-induced cell loss. J Neurotrauma 2010; 27:877-88. [PMID: 20199194 DOI: 10.1089/neu.2008.0616] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The controlled cortical impact (CCI) model has been extensively used to study region-specific patterns of neuronal injury and cell death after a focal traumatic brain injury. Although external parameters such as impact velocity and depth of penetration have been defined in this injury model, little is known about the intracranial mechanical responses within cortical and subcortical brain regions where neuronal loss is prevalent. At present, one of the best methods to determine the internal responses of the brain is finite element (FE) modeling. A previously developed and biomechanically validated detailed three-dimensional FE rat brain model, consisting of 255,700 hexahedral elements and representing all essential anatomical features of a rat brain, was used to study intracranial responses in a series of CCI experiments in which injury severity ranged from mild to severe. A linear relationship was found between the percentage of the neuronal loss observed in vivo and the FE model-predicted maximum principal strain (R(2) = 0.602). Interestingly, the FE model also predicted some risk of injury in the cerebellum, located remote from the point of impact, with a 25% neuronal loss for the "severe" impact condition. More research is needed to examine other regions that do not have histological data for comparison with FE model predictions before this injury mechanism and the associated injury threshold can be fully established.
Collapse
Affiliation(s)
- Haojie Mao
- Bioengineering Center, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Albert-Weissenberger C, Sirén AL. Experimental traumatic brain injury. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2010; 2:16. [PMID: 20707892 PMCID: PMC2930598 DOI: 10.1186/2040-7378-2-16] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 08/13/2010] [Indexed: 12/03/2022]
Abstract
Traumatic brain injury, a leading cause of death and disability, is a result of an outside force causing mechanical disruption of brain tissue and delayed pathogenic events which collectively exacerbate the injury. These pathogenic injury processes are poorly understood and accordingly no effective neuroprotective treatment is available so far. Experimental models are essential for further clarification of the highly complex pathology of traumatic brain injury towards the development of novel treatments. Among the rodent models of traumatic brain injury the most commonly used are the weight-drop, the fluid percussion, and the cortical contusion injury models. As the entire spectrum of events that might occur in traumatic brain injury cannot be covered by one single rodent model, the design and choice of a specific model represents a major challenge for neuroscientists. This review summarizes and evaluates the strengths and weaknesses of the currently available rodent models for traumatic brain injury.
Collapse
|
24
|
Ibrahim NG, Ralston J, Smith C, Margulies SS. Physiological and pathological responses to head rotations in toddler piglets. J Neurotrauma 2010; 27:1021-35. [PMID: 20560753 PMCID: PMC2943503 DOI: 10.1089/neu.2009.1212] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Closed head injury is the leading cause of death in children less than 4 years of age, and is thought to be caused in part by rotational inertial motion of the brain. Injury patterns associated with inertial rotations are not well understood in the pediatric population. To characterize the physiological and pathological responses of the immature brain to inertial forces and their relationship to neurological development, toddler-age (4-week-old) piglets were subjected to a single non-impact head rotation at either low (31.6 +/- 4.7 rad/sec(2), n = 4) or moderate (61.0 +/- 7.5 rad/sec(2), n = 6) angular acceleration in the axial direction. Graded outcomes were observed for both physiological and histopathological responses such that increasing angular acceleration and velocity produced more severe responses. Unlike low-acceleration rotations, moderate-acceleration rotations produced marked EEG amplitude suppression immediately post-injury, which remained suppressed for the 6-h survival period. In addition, significantly more severe subarachnoid hemorrhage, ischemia, and axonal injury by beta-amyloid precursor protein (beta-APP) were observed in moderate-acceleration animals than low-acceleration animals. When compared to infant-age (5-day-old) animals subjected to similar (54.1 +/- 9.6 rad/sec(2)) acceleration rotations, 4-week-old moderate-acceleration animals sustained similar severities of subarachnoid hemorrhage and axonal injury at 6 h post-injury, despite the larger, softer brain in the older piglets. We conclude that the traditional mechanical engineering approach of scaling by brain mass and stiffness cannot explain the vulnerability of the infant brain to acceleration-deceleration movements, compared with the toddler.
Collapse
Affiliation(s)
- Nicole G. Ibrahim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jill Ralston
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Colin Smith
- Department of Neuropathology, University of Edinburgh, Edinburgh, United Kingdom
| | - Susan S. Margulies
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Elliott MB, Jallo JJ, Barbe MF, Tuma RF. Hypertonic saline attenuates tissue loss and astrocyte hypertrophy in a model of traumatic brain injury. Brain Res 2009; 1305:183-91. [DOI: 10.1016/j.brainres.2009.09.104] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 09/24/2009] [Accepted: 09/26/2009] [Indexed: 11/26/2022]
|