1
|
Sun M, Symons GF, Spitz G, O'Brien WT, Baker TL, Fan J, Martins BD, Allen J, Giesler LP, Mychasiuk R, van Donkelaar P, Brand J, Christie B, O'Brien TJ, O'Sullivan MJ, Mitra B, Wellington C, McDonald SJ, Shultz SR. Pathophysiology, blood biomarkers, and functional deficits after intimate partner violence-related brain injury: Insights from emergency department patients and a new rat model. Brain Behav Immun 2024; 123:383-396. [PMID: 39349286 DOI: 10.1016/j.bbi.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/22/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024] Open
Abstract
Intimate partner violence is a serious, but underappreciated, issue that predominantly affects women and often results in concussion (i.e., mild traumatic brain injury). However, concussion in intimate partner violence is unique because it often involves a concomitant strangulation which may exacerbate or alter the physiology and clinical presentation of the brain injury. Therefore, here we conducted human and rodent studies to provide insight into knowledge gaps related to the detection, pathophysiology, and functional consequences of intimate partner violence-related brain injury. We conducted the first study to analyze blood biomarkers and symptoms of brain injury in intimate partner violence patients presenting to an emergency department within 72 h of concussion. Intimate partner violence concussion patients, some of whom had also experienced a concomitant strangulation, had elevated serum neurofilament light and worse brain injury symptoms compared to healthy control, orthopedic trauma, and non-intimate partner violence concussion groups. We also developed the first rat model of non-fatal strangulation and examined the consequences of strangulation and concussion in isolation and in combination on pathophysiology, blood biomarkers, and behavior at 2 h and 1wk post-injury. Rats exposed to combined strangulation and concussion had exacerbated motor and cognitive deficits, neuroinflammation, and serum glial fibrillary acidic protein levels compared with either injury in isolation. Taken together, these rodent findings demonstrate that a concomitant strangulation modifies and exacerbates concussion pathophysiology, biomarkers, and functional consequences. Overall, these findings provide novel insights into intimate partner violence-related brain injury and provides a foundation for future translational studies.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Georgia F Symons
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Centre for Trauma & Mental Health Research, Vancouver Island University, Nanaimo, BC, Canada
| | - Gershon Spitz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Monash-Epworth Rehabilitation Research Centre, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - William T O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Tamara L Baker
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Jianjia Fan
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Beatriz D Martins
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Josh Allen
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Centre for Trauma & Mental Health Research, Vancouver Island University, Nanaimo, BC, Canada; Neuroscience Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lauren P Giesler
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Paul van Donkelaar
- Health & Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Justin Brand
- Neuroscience Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Brian Christie
- Neuroscience Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Terence J O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Michael J O'Sullivan
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Biswadev Mitra
- Emergency Services, Alfred Health, Melbourne, VIC, Australia; School of Public Health & Preventative Medicine, Monash University, Melbourne, VIC, Australia
| | - Cheryl Wellington
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Stuart J McDonald
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Centre for Trauma & Mental Health Research, Vancouver Island University, Nanaimo, BC, Canada; Neuroscience Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
2
|
Quach M, Ali I, Shultz SR, Casillas-Espinosa PM, Hudson MR, Jones NC, Silva JC, Yamakawa GR, Braine EL, Immonen R, Staba RJ, Tohka J, Harris NG, Gröhn O, O'Brien TJ, Wright DK. ComBating inter-site differences in field strength: harmonizing preclinical traumatic brain injury MRI data. NMR IN BIOMEDICINE 2024; 37:e5142. [PMID: 38494895 DOI: 10.1002/nbm.5142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/09/2023] [Accepted: 02/15/2024] [Indexed: 03/19/2024]
Abstract
Integrating datasets from multiple sites and scanners can increase statistical power for neuroimaging studies but can also introduce significant inter-site confounds. We evaluated the effectiveness of ComBat, an empirical Bayes approach, to combine longitudinal preclinical MRI data acquired at 4.7 or 9.4 T at two different sites in Australia. Male Sprague Dawley rats underwent MRI on Days 2, 9, 28, and 150 following moderate/severe traumatic brain injury (TBI) or sham injury as part of Project 1 of the NIH/NINDS-funded Centre Without Walls EpiBioS4Rx project. Diffusion-weighted and multiple-gradient-echo images were acquired, and outcomes included QSM, FA, and ADC. Acute injury measures including apnea and self-righting reflex were consistent between sites. Mixed-effect analysis of ipsilateral and contralateral corpus callosum (CC) summary values revealed a significant effect of site on FA and ADC values, which was removed following ComBat harmonization. Bland-Altman plots for each metric showed reduced variability across sites following ComBat harmonization, including for QSM, despite appearing to be largely unaffected by inter-site differences and no effect of site observed. Following harmonization, the combined inter-site data revealed significant differences in the imaging metrics consistent with previously reported outcomes. TBI resulted in significantly reduced FA and increased susceptibility in the ipsilateral CC, and significantly reduced FA in the contralateral CC compared with sham-injured rats. Additionally, TBI rats also exhibited a reversal in ipsilateral CC ADC values over time with significantly reduced ADC at Day 9, followed by increased ADC 150 days after injury. Our findings demonstrate the need for harmonizing multi-site preclinical MRI data and show that this can be successfully achieved using ComBat while preserving phenotypical changes due to TBI.
Collapse
Affiliation(s)
- Mara Quach
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Victoria, Australia
- Melbourne Brain Centre Imaging Unit, The University of Melbourne, Melbourne, Victoria, Australia
| | - Idrish Ali
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
- Health Sciences, Vancouver Island University, Nanaimo, British Columbia, Canada
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Matthew R Hudson
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Nigel C Jones
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Juliana C Silva
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Emma L Braine
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Riikka Immonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Richard J Staba
- Department of Neurology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California, USA
| | - Jussi Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Neil G Harris
- Department of Neurology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California, USA
| | - Olli Gröhn
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Terence J O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Agoston DV, Helmy A. Fluid-Based Protein Biomarkers in Traumatic Brain Injury: The View from the Bedside. Int J Mol Sci 2023; 24:16267. [PMID: 38003454 PMCID: PMC10671762 DOI: 10.3390/ijms242216267] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
There has been an explosion of research into biofluid (blood, cerebrospinal fluid, CSF)-based protein biomarkers in traumatic brain injury (TBI) over the past decade. The availability of very large datasets, such as CENTRE-TBI and TRACK-TBI, allows for correlation of blood- and CSF-based molecular (protein), radiological (structural) and clinical (physiological) marker data to adverse clinical outcomes. The quality of a given biomarker has often been framed in relation to the predictive power on the outcome quantified from the area under the Receiver Operating Characteristic (ROC) curve. However, this does not in itself provide clinical utility but reflects a statistical association in any given population between one or more variables and clinical outcome. It is not currently established how to incorporate and integrate biofluid-based biomarker data into patient management because there is no standardized role for such data in clinical decision making. We review the current status of biomarker research and discuss how we can integrate existing markers into current clinical practice and what additional biomarkers do we need to improve diagnoses and to guide therapy and to assess treatment efficacy. Furthermore, we argue for employing machine learning (ML) capabilities to integrate the protein biomarker data with other established, routinely used clinical diagnostic tools, to provide the clinician with actionable information to guide medical intervention.
Collapse
Affiliation(s)
- Denes V. Agoston
- Department of Anatomy, Physiology and Genetic, School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK;
| |
Collapse
|
4
|
Allen J, Pham L, Bond ST, O’Brien WT, Spitz G, Shultz SR, Drew BG, Wright DK, McDonald SJ. Acute effects of single and repeated mild traumatic brain injury on levels of neurometabolites, lipids, and mitochondrial function in male rats. Front Mol Neurosci 2023; 16:1208697. [PMID: 37456524 PMCID: PMC10338885 DOI: 10.3389/fnmol.2023.1208697] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Mild traumatic brain injuries (mTBIs) are the most common form of acquired brain injury. Symptoms of mTBI are thought to be associated with a neuropathological cascade, potentially involving the dysregulation of neurometabolites, lipids, and mitochondrial bioenergetics. Such alterations may play a role in the period of enhanced vulnerability that occurs after mTBI, such that a second mTBI will exacerbate neuropathology. However, it is unclear whether mTBI-induced alterations in neurometabolites and lipids that are involved in energy metabolism and other important cellular functions are exacerbated by repeat mTBI, and if such alterations are associated with mitochondrial dysfunction. Methods In this experiment, using a well-established awake-closed head injury (ACHI) paradigm to model mTBI, male rats were subjected to a single injury, or five injuries delivered 1 day apart, and injuries were confirmed with a beam-walk task and a video observation protocol. Abundance of several neurometabolites was evaluated 24 h post-final injury in the ipsilateral and contralateral hippocampus using in vivo proton magnetic resonance spectroscopy (1H-MRS), and mitochondrial bioenergetics were evaluated 30 h post-final injury, or at 24 h in place of 1H-MRS, in the rostral half of the ipsilateral hippocampus. Lipidomic evaluations were conducted in the ipsilateral hippocampus and cortex. Results We found that behavioral deficits in the beam task persisted 1- and 4 h after the final injury in rats that received repetitive mTBIs, and this was paralleled by an increase and decrease in hippocampal glutamine and glucose, respectively, whereas a single mTBI had no effect on sensorimotor and metabolic measurements. No group differences were observed in lipid levels and mitochondrial bioenergetics in the hippocampus, although some lipids were altered in the cortex after repeated mTBI. Discussion The decrease in performance in sensorimotor tests and the presence of more neurometabolic and lipidomic abnormalities, after repeated but not singular mTBI, indicates that multiple concussions in short succession can have cumulative effects. Further preclinical research efforts are required to understand the underlying mechanisms that drive these alterations to establish biomarkers and inform treatment strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Josh Allen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Louise Pham
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Simon T. Bond
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - William T. O’Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Gershon Spitz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Monash-Epworth Rehabilitation Research Centre, Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Health Sciences, Vancouver Island University, Nanaimo, BC, Canada
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Brian G. Drew
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - David K. Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Eagle SR, Puccio AM, Agoston DV, Soose R, Mancinelli M, Nwafo R, McIntyre P, Agnone A, Tollefson S, Collins M, Kontos AP, Schneider W, Okonkwo DO. Evaluating Targeted Therapeutic Response With Predictive Blood-Based Biomarkers in Patients With Chronic Mild Traumatic Brain Injury. Neurotrauma Rep 2023; 4:404-409. [PMID: 37360545 PMCID: PMC10288300 DOI: 10.1089/neur.2023.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Chronic consequences of mild traumatic brain injury (mTBI) are heterogeneous, but may be treatable with targeted medical and rehabilitation interventions. A biological signature for the likelihood of response to therapy (i.e., "predictive" biomarkers) would empower personalized medicine post-mTBI. The purpose of this study was to correlate pre-intervention blood biomarker levels and the likelihood of response to targeted interventions for patients with chronic issues attributable to mTBI. Patients with chronic symptoms and/or disorders secondary to mTBI >3 months previous (104 days to 15 years; n = 74) were enrolled. Participants completed pre-intervention assessments of symptom burden, comprehensive clinical evaluation, and blood-based biomarker measurements. Multi-domain targeted interventions for specific symptoms and impairments across a 6-month treatment period were prescribed. Participants completed a follow-up testing after the treatment period. An all-possible model's backward logistic regression was built to identify predictors of improvement in relation to blood biomarker levels before intervention. The minimum clinically important difference (MCID) of the change score (post-intervention subtracted from pre-intervention) for the Post-Concussion Symptom Scale (PCSS) to identify treatment responders from non-responders was the primary outcome. The MCID for total PCSS score was 10. The model to predict change in PCSS score over the 6-month intervention was significant (R2 = 0.09; p = 0.01) and identified ubiquitin C-terminal hydrolase L1 (odds ratio [OR] = 2.53; 95% confidence interval [CI], 1.18-5.46; p = 0.02) and hyperphosphorylated tau (p-tau; OR = 0.70; 95% CI, 0.51-0.96; p = 0.03) as significant predictors of symptom improvement beyond the PCSS MCID. In this cohort of chronic TBI subjects, blood biomarkers before rehabilitation intervention predicted the likelihood of response to targeted therapy for chronic disorders post-TBI.
Collapse
Affiliation(s)
- Shawn R. Eagle
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ava M. Puccio
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Denes V. Agoston
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Ryan Soose
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael Mancinelli
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rachel Nwafo
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Peyton McIntyre
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Allison Agnone
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Savannah Tollefson
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael Collins
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony P. Kontos
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Walter Schneider
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David O. Okonkwo
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
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: 5.0] [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
|
7
|
Saletti PG, Mowrey WB, Liu W, Li Q, McCullough J, Aniceto R, Lin I, Eklund M, Casillas‐Espinosa PM, Ali I, Santana‐Gomez C, Coles L, Shultz SR, Jones N, Staba R, O'Brien TJ, Moshé SL, Agoston DV, Galanopoulou AS. Early preclinical plasma protein biomarkers of brain trauma are influenced by early seizures and levetiracetam. Epilepsia Open 2023; 8:586-608. [PMID: 37026764 PMCID: PMC10235584 DOI: 10.1002/epi4.12738] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
OBJECTIVE We used the lateral fluid percussion injury (LFPI) model of moderate-to-severe traumatic brain injury (TBI) to identify early plasma biomarkers predicting injury, early post-traumatic seizures or neuromotor functional recovery (neuroscores), considering the effect of levetiracetam, which is commonly given after severe TBI. METHODS Adult male Sprague-Dawley rats underwent left parietal LFPI, received levetiracetam (200 mg/kg bolus, 200 mg/kg/day subcutaneously for 7 days [7d]) or vehicle post-LFPI, and were continuously video-EEG recorded (n = 14/group). Sham (craniotomy only, n = 6), and naïve controls (n = 10) were also used. Neuroscores and plasma collection were done at 2d or 7d post-LFPI or equivalent timepoints in sham/naïve. Plasma protein biomarker levels were determined by reverse phase protein microarray and classified according to injury severity (LFPI vs. sham/control), levetiracetam treatment, early seizures, and 2d-to-7d neuroscore recovery, using machine learning. RESULTS Low 2d plasma levels of Thr231 -phosphorylated tau protein (pTAU-Thr231 ) and S100B combined (ROC AUC = 0.7790) predicted prior craniotomy surgery (diagnostic biomarker). Levetiracetam-treated LFPI rats were differentiated from vehicle treated by the 2d-HMGB1, 2d-pTAU-Thr231 , and 2d-UCHL1 plasma levels combined (ROC AUC = 0.9394) (pharmacodynamic biomarker). Levetiracetam prevented the seizure effects on two biomarkers that predicted early seizures only among vehicle-treated LFPI rats: pTAU-Thr231 (ROC AUC = 1) and UCHL1 (ROC AUC = 0.8333) (prognostic biomarker of early seizures among vehicle-treated LFPI rats). Levetiracetam-resistant early seizures were predicted by high 2d-IFNγ plasma levels (ROC AUC = 0.8750) (response biomarker). 2d-to-7d neuroscore recovery was best predicted by higher 2d-S100B, lower 2d-HMGB1, and 2d-to-7d increase in HMGB1 or decrease in TNF (P < 0.05) (prognostic biomarkers). SIGNIFICANCE Antiseizure medications and early seizures need to be considered in the interpretation of early post-traumatic biomarkers.
Collapse
Affiliation(s)
- Patricia G. Saletti
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Wenzhu B. Mowrey
- Department of Epidemiology & Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Wei Liu
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Qianyun Li
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Jesse McCullough
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Roxanne Aniceto
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - I‐Hsuan Lin
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Michael Eklund
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Pablo M. Casillas‐Espinosa
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Idrish Ali
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | | | - Lisa Coles
- University of Minnesota Twin CitiesMinneapolisMinnesotaUSA
| | - Sandy R. Shultz
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Nigel Jones
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | | | - Terence J. O'Brien
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
- Isabelle Rapin Division of Child NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Dominick P Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Denes V. Agoston
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
- Isabelle Rapin Division of Child NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Dominick P Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
| | | |
Collapse
|
8
|
Sub-acute Changes on MRI Measures of Cerebral Blood Flow and Venous Oxygen Saturation in Concussed Australian Rules Footballers. SPORTS MEDICINE - OPEN 2022; 8:45. [PMID: 35362855 PMCID: PMC8975948 DOI: 10.1186/s40798-022-00435-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/17/2022] [Indexed: 12/03/2022]
Abstract
Background Sports-related concussion (SRC) is common in collision sport athletes. There is growing evidence that repetitive SRC can have serious neurological consequences, particularly when the repetitive injuries occur when the brain has yet to fully recover from the initial injury. Hence, there is a need to identify biomarkers that are capable of determining SRC recovery so that they can guide clinical decisions pertaining to return-to-play. Cerebral venous oxygen saturation (SvO2) and cerebral blood flow (CBF) can be measured using magnetic resonance imaging (MRI) and may provide insights into changing energy demands and recovery following SRC. Results In this study we therefore investigated SvO2 and CBF in a cohort of concussed amateur Australian Football athletes (i.e., Australia’s most participated collision sport). Male and female Australian footballers (n = 13) underwent MRI after being cleared to return to play following a mandatory 13-day recovery period and were compared to a group of control Australian footballers (n = 16) with no recent history of SRC (i.e., > 3 months since last SRC). Despite the concussed Australian footballers being cleared to return to play at the time of MRI, we found evidence of significantly increased susceptibility in the global white matter (p = 0.020) and a trend (F5,21 = 2.404, p = 0.071) for reduced relative CBF (relCBF) compared to the control group. Further, there was evidence of an interaction between sex and injury in straight sinus susceptibility values (F1,25 = 3.858, p = 0.061) which were decreased in female SRC athletes (p = 0.053). Of note, there were significant negative correlations between straight sinus susceptibility and relCBF suggesting impaired metabolic function after SRC. Conclusions These findings support the use of quantitative susceptibility mapping (QSM) and relCBF as sensitive indicators of SRC, and raise further concerns related to SRC guidelines that allow for return-to-play in less than two weeks.
Collapse
|
9
|
San Martín Molina I, Fratini M, Campi G, Burghammer M, Grünewald TA, Salo RA, Narvaez O, Aggarwal M, Tohka J, Sierra A. A multiscale tissue assessment in a rat model of mild traumatic brain injury. J Neuropathol Exp Neurol 2022; 82:71-83. [PMID: 36331507 PMCID: PMC9764078 DOI: 10.1093/jnen/nlac100] [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] [Indexed: 11/06/2022] Open
Abstract
Diffusion tensor imaging (DTI) has demonstrated the potential to assess the pathophysiology of mild traumatic brain injury (mTBI) but correlations of DTI findings and pathological changes in mTBI are unclear. We evaluated the potential of ex vivo DTI to detect tissue damage in a mild mTBI rat model by exploiting multiscale imaging methods, histology and scanning micro-X-ray diffraction (SμXRD) 35 days after sham-operation (n = 2) or mTBI (n = 3). There were changes in DTI parameters rostral to the injury site. When examined by histology and SμXRD, there was evidence of axonal damage, reduced myelin density, gliosis, and ultrastructural alterations in myelin that were ongoing at the experimental time point of 35 days postinjury. We assessed the relationship between the 3 imaging modalities by multiple linear regression analysis. In this analysis, DTI and histological parameters were moderately related, whereas SμXRD parameters correlated weakly with DTI and histology. These findings suggest that while DTI appears to distinguish tissue changes at the microstructural level related to the loss of myelinated axons and gliosis, its ability to visualize alterations in myelin ultrastructure is limited. The use of several imaging techniques represents a novel approach to reveal tissue damage and provides new insights into mTBI detection.
Collapse
Affiliation(s)
| | - Michela Fratini
- Institute of Nanotechnology-CNR c/o Physics Department, Sapienza University of Rome, Rome, Italy,IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | | | - Tilman A Grünewald
- European Synchrotron Radiation Facility, Grenoble Cedex, France,Aix-Marseille Université, CNRS, Centrale Marseille, Institut Fresnel, Marseille, France
| | - Raimo A Salo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Omar Narvaez
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Manisha Aggarwal
- Russell H. Morgan Department of Radiology and Radiological Science, John Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jussi Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Alejandra Sierra
- Send correspondence to: Alejandra Sierra, PhD, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland (Kuopio Campus), PO Box 1627, Neulaniementie 2, FI-70211 Kuopio, Finland; E-mail:
| |
Collapse
|
10
|
Ribonuclease-1 treatment after traumatic brain injury preserves blood-brain barrier integrity and delays secondary brain damage in mice. Sci Rep 2022; 12:5731. [PMID: 35388024 PMCID: PMC8986812 DOI: 10.1038/s41598-022-09326-2] [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] [Received: 10/05/2021] [Accepted: 03/22/2022] [Indexed: 11/08/2022] Open
Abstract
Traumatic brain injury (TBI) involves primary mechanical damage and delayed secondary damage caused by vascular dysfunction and neuroinflammation. Intracellular components released into the parenchyma and systemic circulation, termed danger-associated molecular patterns (DAMPs), are major drivers of vascular dysfunction and neuroinflammation. These DAMPs include cell-free RNAs (cfRNAs), which damage the blood-brain barrier (BBB), thereby promoting edema, procoagulatory processes, and infiltration of inflammatory cells. We tested the hypothesis that intraperitoneal injection of Ribonuclease-1 (RNase1, two doses of 20, 60, or 180 µg/kg) at 30 min and 12 h after controlled-cortical-impact (CCI) can reduce secondary lesion expansion compared to vehicle treatment 24 h and 120 h post-CCI. The lowest total dose (40 µg/kg) was most effective at reducing lesion volume (- 31% RNase 40 µg/kg vs. vehicle), brain water accumulation (- 5.5%), and loss of BBB integrity (- 21.6%) at 24 h post-CCI. RNase1 also reduced perilesional leukocyte recruitment (- 53.3%) and microglial activation (- 18.3%) at 120 h post-CCI, but there was no difference in lesion volume at this time and no functional benefit. Treatment with RNase1 in the early phase following TBI stabilizes the BBB and impedes leukocyte immigration, thereby suppressing neuroinflammation. RNase1-treatment may be a novel approach to delay brain injury to extend the window for treatment opportunities after TBI.
Collapse
|
11
|
Zamani A, Walker AK, Rollo B, Ayers KL, Farah R, O'Brien TJ, Wright DK. Impaired glymphatic function in the early stages of disease in a TDP-43 mouse model of amyotrophic lateral sclerosis. Transl Neurodegener 2022; 11:17. [PMID: 35287738 PMCID: PMC8922788 DOI: 10.1186/s40035-022-00291-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Multiple lines of evidence suggest possible impairment of the glymphatic system in amyotrophic lateral sclerosis (ALS). To investigate this, we used in vivo magnetic resonance imaging (MRI) to assess glymphatic function early in the course of disease in a transgenic mouse with doxycycline (Dox)-controlled expression of cytoplasmic human TDP-43 (hTDP-43ΔNLS), mimicking the key pathology implicated in ALS. METHODS Adult TDP-43 transgenic and littermate monogenic control mice underwent longitudinal multimodal MRI one and three weeks after the cessation of Dox feed, together with weekly rotarod assessments of motor performance. Glymphatic function was assessed using dynamic contrast-enhanced MRI to track the clearance of an MR contrast agent injected into the cisterna magna. RESULTS Compared to their littermate controls, TDP-43 mice exhibited progressive neurodegeneration including that within the primary motor cortex, primary somatosensory cortex and corticospinal tract, significant weight loss including gastrocnemius atrophy, and shortened telomere length. Furthermore, in the presence of this ALS-like phenotype, these mice have significantly disrupted glymphatic function. CONCLUSIONS Although the relationship between glymphatic clearance and ALS disease progression remains to be elucidated, these changes occurred very early in the disease course. This provides initial evidence to suggest that the glymphatic system might be a potential therapeutic target in the treatment of ALS.
Collapse
Affiliation(s)
- Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Adam K Walker
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Ben Rollo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Katie L Ayers
- The Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, 3052, Australia.,Department of Pediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Raysha Farah
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
12
|
Sun M, Symons GF, O'Brien WT, Mccullough J, Aniceto R, Lin IH, Eklund M, Brady RD, Costello DM, Chen Z, O'Brien TJ, McDonald SJ, Agoston DV, Shultz SR. Serum protein biomarkers of inflammation, oxidative stress, and cerebrovascular and glial injury in concussed Australian football players. J Neurotrauma 2022; 39:800-808. [PMID: 35176905 DOI: 10.1089/neu.2021.0493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Clinical decisions related to sports-related concussion (SRC) are challenging due to the heterogenous nature of SRC symptoms coupled with the current reliance on subjective self-reported symptom measures. Sensitive and objective methods that can diagnose SRC and determine recovery would aid clinical management, and there is evidence that SRC induces changes in circulating protein biomarkers indicative of neuroaxonal injury. However, potential blood biomarkers related to other pathobiological responses linked to SRC are still poorly understood. Therefore, here we analyzed blood samples from concussed (male = 30; female = 9) and non-concussed (male = 74; female = 27) amateur Australian rules football players collected during the pre-season (i.e., baseline), and at 2-, 6-, and 13-days post-SRC to determine time dependent changes in serum levels of biomarkers related to glial (i.e., brain lipid-binding protein, BLBP; phosphoprotein enriched in astrocytes 15) and cerebrovascular injury (i.e., von Willebrand factor, claudin-5), inflammation (i.e., fibrinogen, high mobility group box protein 1), and oxidative stress (i.e., 4-hydroxynoneal). In females, BLBP levels were significantly decreased at 2-days post-SRC compared to their pre-season baseline; however, area under the receiver operating characteristic curve (AUROC) analysis found that BLBP was unable to distinguish between SRC and controls. In males, AUROC analysis revealed a statistically significant change at 2-days post-SRC in the serum levels of 4-hydroxynoneal, however the associated AUROC value (0.6373) indicated little clinical utility for this biomarker in distinguishing SRC from controls. There were no other statistically significant findings. These results indicate that the serum biomarkers tested in this study hold little clinical value in the management of SRC at 2-, 6-, and 13-days post-injury.
Collapse
Affiliation(s)
- Mujun Sun
- Monash University, Department of Neuroscience, Central Clinical School, Melbourne, Australia;
| | - Georgia F Symons
- Monash University, Neuroscience, Melbourne, Victoria, Australia;
| | | | | | | | | | | | - Rhys D Brady
- Monash University, Neuroscience, The Alfred Centre, Level 6, 99 Commercial Rd, Melbourne, Victoria, Australia, 3004;
| | - Daniel M Costello
- The University of Melbourne, 2281, Department of Medicine, Melbourne, Victoria, Australia;
| | - Zhibin Chen
- Monash University, Neuroscience, Melbourne, Victoria, Australia.,Monash University, 2541, Clinical Epidemiology, Melbourne, Victoria, Australia;
| | - Terence J O'Brien
- Monash University, Neuroscience, Melbourne, Victoria, Australia.,Melbourne Health, 6451, Department of Neurology, Parkville, Victoria, Australia.,Alfred Health, 5392, Department of Neurology, Melbourne, Victoria, Australia.,The University of Melbourne, 2281, Department of Medicine, Melbourne, Victoria, Australia;
| | - Stuart John McDonald
- Monash University Central Clinical School, 161666, Department of Neuroscience, 99 Commercial Road, Melbourne, Victoria, Australia, 3004;
| | - Denes V Agoston
- Uniformed Services University, APG, 4301 Jones Br Rd, Bethesda, Maryland, United States, 20814;
| | - Sandy R Shultz
- Monash University, Neuroscience, Level 6, Alfred Centre, 99 Commercial Road, Melbourne, Victoria, Australia, 3004;
| |
Collapse
|
13
|
Yeh FC, Irimia A, Bastos DCDA, Golby AJ. Tractography methods and findings in brain tumors and traumatic brain injury. Neuroimage 2021; 245:118651. [PMID: 34673247 PMCID: PMC8859988 DOI: 10.1016/j.neuroimage.2021.118651] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 10/05/2021] [Accepted: 10/11/2021] [Indexed: 12/31/2022] Open
Abstract
White matter fiber tracking using diffusion magnetic resonance imaging (dMRI) provides a noninvasive approach to map brain connections, but improving anatomical accuracy has been a significant challenge since the birth of tractography methods. Utilizing tractography in brain studies therefore requires understanding of its technical limitations to avoid shortcomings and pitfalls. This review explores tractography limitations and how different white matter pathways pose different challenges to fiber tracking methodologies. We summarize the pros and cons of commonly-used methods, aiming to inform how tractography and its related analysis may lead to questionable results. Extending these experiences, we review the clinical utilization of tractography in patients with brain tumors and traumatic brain injury, starting from tensor-based tractography to more advanced methods. We discuss current limitations and highlight novel approaches in the context of these two conditions to inform future tractography developments.
Collapse
Affiliation(s)
- Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA; Corwin D. Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | | | - Alexandra J Golby
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Baker TL, Agoston DV, Brady RD, Major B, McDonald SJ, Mychasiuk R, Wright DK, Yamakawa GR, Sun M, Shultz SR. Targeting the Cerebrovascular System: Next-Generation Biomarkers and Treatment for Mild Traumatic Brain Injury. Neuroscientist 2021; 28:594-612. [PMID: 33966527 DOI: 10.1177/10738584211012264] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The diagnosis, prognosis, and treatment of mild traumatic brain injuries (mTBIs), such as concussions, are significant unmet medical issues. The kinetic forces that occur in mTBI adversely affect the cerebral vasculature, making cerebrovascular injury (CVI) a pathophysiological hallmark of mTBI. Given the importance of a healthy cerebrovascular system in overall brain function, CVI is likely to contribute to neurological dysfunction after mTBI. As such, CVI and related pathomechanisms may provide objective biomarkers and therapeutic targets to improve the clinical management and outcomes of mTBI. Despite this potential, until recently, few studies have focused on the cerebral vasculature in this context. This article will begin by providing a brief overview of the cerebrovascular system followed by a review of the literature regarding how mTBI can affect the integrity and function of the cerebrovascular system, and how this may ultimately contribute to neurological dysfunction and neurodegenerative conditions. We then discuss promising avenues of research related to mTBI biomarkers and interventions that target CVI, and conclude that a clinical approach that takes CVI into account could result in substantial improvements in the care and outcomes of patients with mTBI.
Collapse
Affiliation(s)
- Tamara L Baker
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Brendan Major
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
15
|
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: 4.3] [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
|
16
|
Janigro D, Bailey DM, Lehmann S, Badaut J, O'Flynn R, Hirtz C, Marchi N. Peripheral Blood and Salivary Biomarkers of Blood-Brain Barrier Permeability and Neuronal Damage: Clinical and Applied Concepts. Front Neurol 2021; 11:577312. [PMID: 33613412 PMCID: PMC7890078 DOI: 10.3389/fneur.2020.577312] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
Within the neurovascular unit (NVU), the blood–brain barrier (BBB) operates as a key cerebrovascular interface, dynamically insulating the brain parenchyma from peripheral blood and compartments. Increased BBB permeability is clinically relevant for at least two reasons: it actively participates to the etiology of central nervous system (CNS) diseases, and it enables the diagnosis of neurological disorders based on the detection of CNS molecules in peripheral body fluids. In pathological conditions, a suite of glial, neuronal, and pericyte biomarkers can exit the brain reaching the peripheral blood and, after a process of filtration, may also appear in saliva or urine according to varying temporal trajectories. Here, we specifically examine the evidence in favor of or against the use of protein biomarkers of NVU damage and BBB permeability in traumatic head injury, including sport (sub)concussive impacts, seizure disorders, and neurodegenerative processes such as Alzheimer's disease. We further extend this analysis by focusing on the correlates of human extreme physiology applied to the NVU and its biomarkers. To this end, we report NVU changes after prolonged exercise, freediving, and gravitational stress, focusing on the presence of peripheral biomarkers in these conditions. The development of a biomarker toolkit will enable minimally invasive routines for the assessment of brain health in a broad spectrum of clinical, emergency, and sport settings.
Collapse
Affiliation(s)
- Damir Janigro
- Department of Physiology Case Western Reserve University, Cleveland, OH, United States.,FloTBI Inc., Cleveland, OH, United States
| | - Damian M Bailey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Wales, United Kingdom
| | - Sylvain Lehmann
- IRMB, INM, UFR Odontology, University Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Jerome Badaut
- Brain Molecular Imaging Lab, CNRS UMR 5287, INCIA, University of Bordeaux, Bordeaux, France
| | - Robin O'Flynn
- IRMB, INM, UFR Odontology, University Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Christophe Hirtz
- IRMB, INM, UFR Odontology, University Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS-U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
17
|
Sinke MRT, Otte WM, Meerwaldt AE, Franx BAA, Ali MHM, Rakib F, van der Toorn A, van Heijningen CL, Smeele C, Ahmed T, Blezer ELA, Dijkhuizen RM. Imaging Markers for the Characterization of Gray and White Matter Changes from Acute to Chronic Stages after Experimental Traumatic Brain Injury. J Neurotrauma 2021; 38:1642-1653. [PMID: 33198560 DOI: 10.1089/neu.2020.7151] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Despite clinical symptoms, a large majority of people with mild traumatic brain injury (TBI) have normal computed tomography (CT) and magnetic resonance imaging (MRI) scans. Therefore, present-day neuroimaging tools are insufficient to diagnose or classify low grades of TBI. Advanced neuroimaging techniques, such as diffusion-weighted and functional MRI, may yield novel biomarkers that may aid in the diagnosis of TBI. Therefore, the present study had two aims: first, to characterize the development of MRI-based measures of structural and functional changes in gray and white matter regions from acute to chronic stages after mild and moderate TBI; and second, to identify the imaging markers that can most accurately predict outcome after TBI. To these aims, 52 rats underwent serial functional (resting-state) and structural (T1-, T2-, and diffusion-weighted) MRI before and 1 h, 1 day, 1 week, 1 month and 3-4 months after mild or moderate experimental TBI. All rats underwent behavioral testing. Histology was performed in subgroups of rats at different time points. Early after moderate TBI, axial and radial diffusivities were increased, and fractional anisotropy was reduced in the corpus callosum and bilateral hippocampi, which normalized over time and was paralleled by recovery of sensorimotor function. Correspondingly, histology revealed decreased myelin staining early after TBI, which was not detected at chronic stages. No significant changes in individual outcome measures were detected after mild TBI. However, multivariate analysis showed a significant additive contribution of diffusion parameters in the distinction between control and different grades of TBI-affected brains. Therefore, combining multiple imaging markers may increase the sensitivity for TBI-related pathology.
Collapse
Affiliation(s)
- Michel R T Sinke
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| | - Willem M Otte
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078.,UMC Utrecht Brain Center, Department of Child Neurology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| | - Anu E Meerwaldt
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| | - Bart A A Franx
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| | - Mohamed H M Ali
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Fazle Rakib
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Annette van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| | - Caroline L van Heijningen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| | - Christel Smeele
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| | - Tariq Ahmed
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Erwin L A Blezer
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands. ORCID ID: 0000-0002-8185-9209; 0000-0002-4623-4078
| |
Collapse
|
18
|
White matter changes following experimental pediatric traumatic brain injury: an advanced diffusion-weighted imaging investigation. Brain Imaging Behav 2021; 15:2766-2774. [PMID: 33411159 DOI: 10.1007/s11682-020-00433-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/24/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
Pediatric traumatic brain injury (pTBI) is a major community health concern. Due to ongoing maturation, injury to the brain at a young age can have devastating consequences in later life. However, how pTBI affects brain development, including white matter maturation, is still poorly understood. Here, we used advanced diffusion weighted imaging (DWI) to assess chronic white matter changes after experimental pTBI. Mice at post-natal day 21 sustained a TBI using the controlled cortical impact model and magnetic resonance imaging (MRI) was performed at 6 months post-injury using a 4.7 T Bruker scanner. Four diffusion shells with 81 directions and b-values of 1000, 3000, 5000, and 7000s/mm2 were acquired and analyzed using MRtrix3 software. Advanced DWI metrics, including fiber density, fiber cross-section and a combined fiber density and cross-section measure, were investigated together with three track-weighted images (TWI): the average pathlength map, mean curvature and the track density image. These advanced metrics were compared to traditional diffusion tensor imaging (DTI) metrics which indicated that TBI injured mice had reduced fractional anisotropy and increased radial diffusivity in the white matter when compared to age-matched sham controls. Consistent with previous findings, fiber density and TWI metrics appeared to be more sensitive to white matter changes than DTI metrics, revealing widespread reductions in fiber density and TWI metrics in pTBI mice compared to sham controls. These results provide additional support for the use of advanced DWI metrics in assessing white matter degeneration following injury and highlight the chronic outcomes that can follow pTBI.
Collapse
|
19
|
Contrast enhanced magnetic resonance imaging highlights neurovasculature changes following experimental traumatic brain injury in the rat. Sci Rep 2020; 10:21252. [PMID: 33277513 PMCID: PMC7718275 DOI: 10.1038/s41598-020-77975-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 11/10/2020] [Indexed: 11/08/2022] Open
Abstract
Neurovascular injury has been proposed as a universal pathological hallmark of traumatic brain injury (TBI) with molecular markers of angiogenesis and endothelial function associated with injury severity and morbidity. Sex differences in the neurovasculature response post-TBI may contribute to the differences seen in how males and females respond to injury. Steady-state contrast enhanced magnetic resonance imaging (SSCE-MRI) can be used to non-invasively assess the neurovasculature and may be a useful tool in understanding and predicting outcomes post-TBI. Here we used SSCE-MRI to investigate the neurovasculature of male and female rats at 48 h after an experimental TBI, and how these changes related to neuromotor function at 1-week post-TBI. In addition to TBI induced changes, we found that female rats had greater vessel density, greater cerebral blood volumes and performed better on a neuromotor task than their male counterparts. These results suggest that acute post-TBI cerebrovascular function is worse in males, and that this may contribute to the greater functional deficits observed post-injury. Furthermore, these results highlight the potential of SSCE-MRI to provide insights into the cerebral microvasculature post-TBI. Future studies, incorporating both males and females, are warranted to investigate the evolution of these changes and the underlying mechanisms.
Collapse
|
20
|
Ondek K, Pevzner A, Tercovich K, Schedlbauer AM, Izadi A, Ekstrom AD, Cowen SL, Shahlaie K, Gurkoff GG. Recovery of Theta Frequency Oscillations in Rats Following Lateral Fluid Percussion Corresponds With a Mild Cognitive Phenotype. Front Neurol 2020; 11:600171. [PMID: 33343499 PMCID: PMC7746872 DOI: 10.3389/fneur.2020.600171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/21/2020] [Indexed: 01/31/2023] Open
Abstract
Whether from a fall, sports concussion, or even combat injury, there is a critical need to identify when an individual is able to return to play or work following traumatic brain injury (TBI). Electroencephalogram (EEG) and local field potentials (LFP) represent potential tools to monitor circuit-level abnormalities related to learning and memory: specifically, theta oscillations can be readily observed and play a critical role in cognition. Following moderate traumatic brain injury in the rat, lasting changes in theta oscillations coincide with deficits in spatial learning. We hypothesized, therefore, that theta oscillations can be used as an objective biomarker of recovery, with a return of oscillatory activity corresponding with improved spatial learning. In the current study, LFP were recorded from dorsal hippocampus and anterior cingulate in awake, behaving adult Sprague Dawley rats in both a novel environment on post-injury days 3 and 7, and Barnes maze spatial navigation on post-injury days 8–11. Theta oscillations, as measured by power, theta-delta ratio, peak theta frequency, and phase coherence, were significantly altered on day 3, but had largely recovered by day 7 post-injury. Injured rats had a mild behavioral phenotype and were not different from shams on the Barnes maze, as measured by escape latency. Injured rats did use suboptimal search strategies. Combined with our previous findings that demonstrated a correlation between persistent alterations in theta oscillations and spatial learning deficits, these new data suggest that neural oscillations, and particularly theta oscillations, have potential as a biomarker to monitor recovery of brain function following TBI. Specifically, we now demonstrate that oscillations are depressed following injury, but as oscillations recover, so does behavior.
Collapse
Affiliation(s)
- Katelynn Ondek
- Department of Neurological Surgery, University of California, Davis, Davis, CA, United States.,Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Aleksandr Pevzner
- Department of Neurological Surgery, University of California, Davis, Davis, CA, United States
| | - Kayleen Tercovich
- Department of Neurological Surgery, University of California, Davis, Davis, CA, United States.,Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Amber M Schedlbauer
- Department of Neurological Surgery, University of California, Davis, Davis, CA, United States.,Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Ali Izadi
- Department of Neurological Surgery, University of California, Davis, Davis, CA, United States.,Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Arne D Ekstrom
- Department of Psychology, The University of Arizona, Tucson, AZ, United States.,McKnight Brain Institute, The University of Arizona, Tucson, AZ, United States
| | - Stephen L Cowen
- Department of Psychology, The University of Arizona, Tucson, AZ, United States.,McKnight Brain Institute, The University of Arizona, Tucson, AZ, United States
| | - Kiarash Shahlaie
- Department of Neurological Surgery, University of California, Davis, Davis, CA, United States
| | - Gene G Gurkoff
- Department of Neurological Surgery, University of California, Davis, Davis, CA, United States.,Center for Neuroscience, University of California, Davis, Davis, CA, United States
| |
Collapse
|
21
|
Symons GF, Clough M, O’Brien WT, Ernest J, Salberg S, Costello D, Sun M, Brady RD, McDonald SJ, Wright DK, White O, Abel L, O’Brien TJ, Mccullough J, Aniceto R, Lin IH, Agoston DV, Fielding J, Mychasiuk R, Shultz SR. Shortened telomeres and serum protein biomarker abnormalities in collision sport athletes regardless of concussion history and sex. JOURNAL OF CONCUSSION 2020. [DOI: 10.1177/2059700220975609] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mild brain injuries are frequent in athletes engaging in collision sports and have been linked to a range of long-term neurological abnormalities. There is a need to identify how these potential abnormalities manifest using objective measures; determine whether changes are due to concussive and/or sub-concussive injuries; and examine how biological sex affects outcomes. This study investigated cognitive, cellular, and molecular biomarkers in male and female amateur Australian footballers (i.e. Australia’s most participated collision sport). 95 Australian footballers (69 males, 26 females), both with and without a history of concussion, as well as 49 control athletes (28 males, 21 females) with no history of brain trauma or participation in collision sports were recruited to the study. Ocular motor assessment was used to examine cognitive function. Telomere length, a biomarker of cellular senescence and neurological health, was examined in saliva. Serum levels of tau, phosphorylated tau, neurofilament light chain, and 4-hydroxynonenal were used as markers to assess axonal injury and oxidative stress. Australian footballers had reduced telomere length (p = 0.031) and increased serum protein levels of 4-hydroxynonenal (p = 0.001), tau (p = 0.007), and phosphorylated tau (p = 0.036). These findings were independent of concussion history and sex. No significant ocular motor differences were found. Taken together, these findings suggest that engagement in collision sports, regardless of sex or a history of concussion, is associated with shortened telomeres, axonal injury, and oxidative stress. These saliva- and serum-based biomarkers may be useful to monitor neurological injury in collision sport athletes.
Collapse
Affiliation(s)
- Georgia F Symons
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Meaghan Clough
- Department of Neuroscience, Monash University, Melbourne, Australia
| | | | - Joel Ernest
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Daniel Costello
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | | | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Owen White
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Larry Abel
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Terence J O’Brien
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Jesse Mccullough
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - Roxanne Aniceto
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - I-Hsuan Lin
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - Joanne Fielding
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Psychology, University of Calgary, Calgary, AB, Canada
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
22
|
Shultz SR, McDonald SJ, Corrigan F, Semple BD, Salberg S, Zamani A, Jones NC, Mychasiuk R. Clinical Relevance of Behavior Testing in Animal Models of Traumatic Brain Injury. J Neurotrauma 2020; 37:2381-2400. [DOI: 10.1089/neu.2018.6149] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Frances Corrigan
- Department of Anatomy, University of South Australia, Adelaide, South Australia, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
23
|
Pham L, Wright DK, O'Brien WT, Bain J, Huang C, Sun M, Casillas-Espinosa PM, Shah AD, Schittenhelm RB, Sobey CG, Brady RD, O'Brien TJ, Mychasiuk R, Shultz SR, McDonald SJ. Behavioral, axonal, and proteomic alterations following repeated mild traumatic brain injury: Novel insights using a clinically relevant rat model. Neurobiol Dis 2020; 148:105151. [PMID: 33127468 DOI: 10.1016/j.nbd.2020.105151] [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: 08/05/2020] [Revised: 10/07/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022] Open
Abstract
A history of mild traumatic brain injury (mTBI) is linked to a number of chronic neurological conditions, however there is still much unknown about the underlying mechanisms. To provide new insights, this study used a clinically relevant model of repeated mTBI in rats to characterize the acute and chronic neuropathological and neurobehavioral consequences of these injuries. Rats were given four sham-injuries or four mTBIs and allocated to 7-day or 3.5-months post-injury recovery groups. Behavioral analysis assessed sensorimotor function, locomotion, anxiety, and spatial memory. Neuropathological analysis included serum quantification of neurofilament light (NfL), mass spectrometry of the hippocampal proteome, and ex vivo magnetic resonance imaging (MRI). Repeated mTBI rats had evidence of acute cognitive deficits and prolonged sensorimotor impairments. Serum NfL was elevated at 7 days post injury, with levels correlating with sensorimotor deficits; however, no NfL differences were observed at 3.5 months. Several hippocampal proteins were altered by repeated mTBI, including those associated with energy metabolism, neuroinflammation, and impaired neurogenic capacity. Diffusion MRI analysis at 3.5 months found widespread reductions in white matter integrity. Taken together, these findings provide novel insights into the nature and progression of repeated mTBI neuropathology that may underlie lingering or chronic neurobehavioral deficits.
Collapse
Affiliation(s)
- Louise Pham
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - William T O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Jesse Bain
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Cheng Huang
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Anup D Shah
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; Monash Bioinformatics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
24
|
Major BP, McDonald SJ, O'Brien WT, Symons GF, Clough M, Costello D, Sun M, Brady RD, Mccullough J, Aniceto R, Lin IH, Law M, Mychasiuk R, O'Brien TJ, Agoston DV, Shultz SR. Serum Protein Biomarker Findings Reflective of Oxidative Stress and Vascular Abnormalities in Male, but Not Female, Collision Sport Athletes. Front Neurol 2020; 11:549624. [PMID: 33117257 PMCID: PMC7561422 DOI: 10.3389/fneur.2020.549624] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022] Open
Abstract
Studies have indicated that concussive and sub-concussive brain injuries that are frequent during collision sports may lead to long-term neurological abnormalities, however there is a knowledge gap on how biological sex modifies outcomes. Blood-based biomarkers can help to identify the molecular pathology induced by brain injuries and to better understand how biological sex affects the molecular changes. We therefore analyzed serum protein biomarkers in male (n = 50) and female (n = 33) amateur Australian rules footballers (i.e., Australia's most participated collision sport), both with a history of concussion (HoC) and without a history of concussion (NoHoC). These profiles were compared to those of age-matched control male (n = 24) and female (n = 20) athletes with no history of neurotrauma or participation in collision sports. Serum levels of protein markers indicative of neuronal, axonal and glial injury (UCH-L1, NfL, tau, p-tau, GFAP, BLBP, PEA15), metabolic (4-HNE) and vascular changes (VEGF-A, vWF, CLDN5), and inflammation (HMGB1) were assessed using reverse phase protein microarrays. Male, but not female, footballers had increased serum levels of VEGF-A compared to controls regardless of concussion history. In addition, only male footballers who had HoC had increased serum levels of 4-HNE. These findings being restricted to males may be related to shorter collision sport career lengths for females compared to males. In summary, these findings show that male Australian rules footballers have elevated levels of serum biomarkers indicative of vascular abnormalities (VEGF-A) and oxidative stress (4-HNE) in comparison to non-collision control athletes. While future studies are required to determine how these findings relate to neurological function, serum levels of VEGF-A and 4-HNE may be useful to monitor subclinical neurological injury in males participating in collision sports.
Collapse
Affiliation(s)
- Brendan P Major
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - William T O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Georgia F Symons
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Meaghan Clough
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Daniel Costello
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Jesse Mccullough
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, United States
| | - Roxanne Aniceto
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, United States
| | - I-Hsuan Lin
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, United States
| | - Meng Law
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Melbourne, VIC, Australia.,Departments of Neurological Surgery and Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, United States
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
25
|
Müller HP, Roselli F, Rasche V, Kassubek J. Diffusion Tensor Imaging-Based Studies at the Group-Level Applied to Animal Models of Neurodegenerative Diseases. Front Neurosci 2020; 14:734. [PMID: 32982659 PMCID: PMC7487414 DOI: 10.3389/fnins.2020.00734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
The understanding of human and non-human microstructural brain alterations in the course of neurodegenerative diseases has substantially improved by the non-invasive magnetic resonance imaging (MRI) technique of diffusion tensor imaging (DTI). Animal models (including disease or knockout models) allow for a variety of experimental manipulations, which are not applicable to humans. Thus, the DTI approach provides a promising tool for cross-species cross-sectional and longitudinal investigations of the neurobiological targets and mechanisms of neurodegeneration. This overview with a systematic review focuses on the principles of DTI analysis as used in studies at the group level in living preclinical models of neurodegeneration. The translational aspect from in-vivo animal models toward (clinical) applications in humans is covered as well as the DTI-based research of the non-human brains' microstructure, the methodological aspects in data processing and analysis, and data interpretation at different abstraction levels. The aim of integrating DTI in multiparametric or multimodal imaging protocols will allow the interrogation of DTI data in terms of directional flow of information and may identify the microstructural underpinnings of neurodegeneration-related patterns.
Collapse
Affiliation(s)
| | - Francesco Roselli
- Department of Neurology, University of Ulm, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, University of Ulm, Ulm, Germany
| |
Collapse
|
26
|
Selvakumar GP, Ahmed ME, Iyer SS, Thangavel R, Kempuraj D, Raikwar SP, Bazley K, Wu K, Khan A, Kukulka K, Bussinger B, Zaheer S, Burton C, James D, Zaheer A. Absence of Glia Maturation Factor Protects from Axonal Injury and Motor Behavioral Impairments after Traumatic Brain Injury. Exp Neurobiol 2020; 29:230-248. [PMID: 32565489 PMCID: PMC7344375 DOI: 10.5607/en20017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) causes disability and death, accelerating the progression towards Alzheimer's disease and Parkinson's disease (PD). TBI causes serious motor and cognitive impairments, as seen in PD that arise during the period of the initial insult. However, this has been understudied relative to TBI induced neuroinflammation, motor and cognitive decline that progress towards PD. Neuronal ubiquitin-C-terminal hydrolase- L1 (UCHL1) is a thiol protease that breaks down ubiquitinated proteins and its level represents the severity of TBI. Previously, we demonstrated the molecular action of glia maturation factor (GMF); a proinflammatory protein in mediating neuroinflammation and neuronal loss. Here, we show that the weight drop method induced TBI neuropathology using behavioral tests, western blotting, and immunofluorescence techniques on sections from wild type (WT) and GMF-deficient (GMF-KO) mice. Results reveal a significant improvement in substantia nigral tyrosine hydroxylase and dopamine transporter expression with motor behavioral performance in GMF-KO mice following TBI. In addition, a significant reduction in neuroinflammation was manifested, as shown by activation of nuclear factor-kB, reduced levels of inducible nitric oxide synthase, and cyclooxygenase- 2 expressions. Likewise, neurotrophins including brain-derived neurotrophic factor and glial-derived neurotrophic factor were significantly improved in GMF-KO mice than WT 72 h post-TBI. Consistently, we found that TBI enhances GFAP and UCHL-1 expression and reduces the number of dopaminergic TH-positive neurons in WT compared to GMF-KO mice 72 h post-TBI. Interestingly, we observed a reduction of THpositive tanycytes in the median eminence of WT than GMF-KO mice. Overall, we found that absence of GMF significantly reversed these neuropathological events and improved behavioral outcome. This study provides evidence that PD-associated pathology progression can be initiated upon induction of TBI.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Klaudia Kukulka
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Bret Bussinger
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
27
|
In Vivo Diffusion Tensor Imaging in Acute and Subacute Phases of Mild Traumatic Brain Injury in Rats. eNeuro 2020; 7:ENEURO.0476-19.2020. [PMID: 32424056 PMCID: PMC7307627 DOI: 10.1523/eneuro.0476-19.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 04/27/2020] [Accepted: 05/11/2020] [Indexed: 12/23/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is the most common form of TBI with 10–25% of the patients experiencing long-lasting symptoms. The potential of diffusion tensor imaging (DTI) for evaluating microstructural damage after TBI is widely recognized, but the interpretation of DTI changes and their relationship with the underlying tissue damage is unclear. We studied how both axonal damage and gliosis contribute to DTI alterations after mTBI. We induced mTBI using the lateral fluid percussion (LFP) injury model in adult male Sprague Dawley rats and scanned them at 3 and 28 d post-mTBI. To characterize the DTI findings in the tissue, we assessed the histology by performing structure tensor (ST)-based analysis and cell counting on myelin-stained and Nissl-stained sections, respectively. In particular, we studied the contribution of two tissue components, myelinated axons and cellularity, to the DTI changes. Fractional anisotropy (FA), mean diffusivity (MD), and axial diffusivity (AD) were decreased in both white and gray matter areas in the acute phase post-mTBI, mainly at the primary lesion site. In the subacute phase, FA and AD were decreased in the white matter, external capsule, corpus callosum, and internal capsule. Our quantitative histologic assessment revealed axonal damage and gliosis throughout the brain in both white and gray matter, consistent with the FA and AD changes. Our findings suggest that the usefulness of in vivo DTI is limited in its detection of secondary damage distal to the primary lesion, while at the lesion site, DTI detected progressive microstructural damage in the white and gray matter after mTBI.
Collapse
|
28
|
Meningher I, Bernstein-Eliav M, Rubovitch V, Pick CG, Tavor I. Alterations in Network Connectivity after Traumatic Brain Injury in Mice. J Neurotrauma 2020; 37:2169-2179. [PMID: 32434427 DOI: 10.1089/neu.2020.7063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Victims of mild traumatic brain injury (mTBI) usually do not display clear morphological brain defects, but frequently have long-lasting cognitive deficits, emotional difficulties, and behavioral disturbances. In the present study we used diffusion magnetic resonance imaging (dMRI) combined with graph theory measurements to investigate the effects of mTBI on brain network connectivity. We employed a non-invasive closed-head weight-drop mouse model to produce mTBI. Mice were scanned at two time points, 24 h before the injury and either 7 or 30 days following the injury. Connectivity matrices were computed for each animal at each time point, and these were subsequently used to extract graph theory measures reflecting network integration and segregation, on both the global (i.e., whole brain) and local (i.e., single regions) levels. We found that cluster coefficient, reflecting network segregation, decreased 7 days post-injury and then returned to baseline level 30 days following the injury. Global efficiency, reflecting network integration, demonstrated opposite patterns in the left and right hemispheres, with an increase of right hemisphere efficiency at 7 days and then a decrease in efficiency following 30 days, and vice versa in the left hemisphere. These findings suggest a possible compensation mechanism acting to moderate the influence of mTBI on the global network. Moreover, these results highlight the importance of tracking the dynamic changes in mTBI over time, and the potential of structural connectivity as a promising approach for studying network integrity and pathology progression in mTBI.
Collapse
Affiliation(s)
- Inbar Meningher
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Michal Bernstein-Eliav
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel.,Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel-Aviv University, Tel-Aviv, Israel
| | - Ido Tavor
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
29
|
Venkatasubramanian PN, Keni P, Gastfield R, Li L, Aksenov D, Sherman SA, Bailes J, Sindelar B, Finan JD, Lee J, Bailes JE, Wyrwicz AM. Diffusion Tensor Imaging Detects Acute and Subacute Changes in Corpus Callosum in Blast-Induced Traumatic Brain Injury. ASN Neuro 2020; 12:1759091420922929. [PMID: 32403948 PMCID: PMC7238783 DOI: 10.1177/1759091420922929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
There is a critical need for understanding the progression of neuropathology in blast-induced traumatic brain injury using valid animal models to develop diagnostic approaches. In the present study, we used diffusion imaging and magnetic resonance (MR) morphometry to characterize axonal injury in white matter structures of the rat brain following a blast applied via blast tube to one side of the brain. Diffusion tensor imaging was performed on acute and subacute phases of pathology from which fractional anisotropy, mean diffusivity, axial diffusivity, and radial diffusivity were calculated for corpus callosum (CC), cingulum bundle, and fimbria. Ventricular volume and CC thickness were measured. Blast-injured rats showed temporally varying bilateral changes in diffusion metrics indicating persistent axonal pathology. Diffusion changes in the CC suggested vasogenic edema secondary to axonal injury in the acute phase. Axonal pathology persisted in the subacute phase marked by cytotoxic edema and demyelination which was confirmed by ultrastructural analysis. The evolution of pathology followed a different pattern in the cingulum bundle: axonal injury and demyelination in the acute phase followed by cytotoxic edema in the subacute phase. Spatially, structures close to midline were most affected. Changes in the genu were greater than in the body and splenium; the caudal cingulum bundle was more affected than the rostral cingulum. Thinning of CC and ventriculomegaly were greater only in the acute phase. Our results reveal the persistent nature of blast-induced axonal pathology and suggest that diffusion imaging may have potential for detecting the temporal evolution of blast injury.
Collapse
Affiliation(s)
- Palamadai N Venkatasubramanian
- Center for Basic M.R. Research, Department of Radiology, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Prachi Keni
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Roland Gastfield
- Center for Basic M.R. Research, Department of Radiology, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Limin Li
- Center for Basic M.R. Research, Department of Radiology, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Daniil Aksenov
- Center for Basic M.R. Research, Department of Radiology, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Sydney A Sherman
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Julian Bailes
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Brian Sindelar
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - John D Finan
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - John Lee
- Department of Pathology, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Julian E Bailes
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, Illinois, United States
| | - Alice M Wyrwicz
- Center for Basic M.R. Research, Department of Radiology, NorthShore University HealthSystem, Evanston, Illinois, United States
| |
Collapse
|
30
|
Gazdzinski LM, Mellerup M, Wang T, Adel SAA, Lerch JP, Sled JG, Nieman BJ, Wheeler AL. White Matter Changes Caused by Mild Traumatic Brain Injury in Mice Evaluated Using Neurite Orientation Dispersion and Density Imaging. J Neurotrauma 2020; 37:1818-1828. [PMID: 32242488 DOI: 10.1089/neu.2020.6992] [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] [Indexed: 12/12/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is common and can lead to persistent cognitive and behavioral symptoms. Although diffusion tensor imaging (DTI) has demonstrated some sensitivity to changes in white matter following mTBI, recent studies have suggested that more complex geometric models of diffusion, including the neurite orientation dispersion and density imaging (NODDI) model, may be more sensitive and specific. Here, we evaluate microstructural changes in white matter following mTBI using DTI and NODDI in a mouse model, and compare the time course of these changes to behavioral impairment and recovery. We also assess volumetric changes for a comprehensive picture of the structural alterations in the brain and histological staining to identify cellular changes that may contribute to the differences detected in the imaging data. Increased orientation dispersion index (ODI) was observed in the optic tracts of mTBI mice compared with shams. Changes in fractional anisotropy (FA) were not statistically significant. Volume deficits were detected in the optic tract as well as in several gray matter regions: the lateral geniculate nuclei of the thalamus, the entorhinal cortex, and the superior colliculi. Glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) staining was increased in the optic tracts of mTBI brains, and this staining correlated with ODI values. A transient impairment in working memory was observed, which resolved by 6 weeks, whereas increased ODI, GFAP, and Iba1 persisted to 18 weeks post-injury. We conclude that the optic tracts are particularly vulnerable to damage from the closed-skull impact model used in this study, and that ODI may be a more sensitive metric to this damage than FA. Differences in ODI and in histological measures of astrogliosis, neuroinflammation, and axonal degeneration persist beyond behavioral impairment in this model.
Collapse
Affiliation(s)
- Lisa M Gazdzinski
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Miranda Mellerup
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and University of Toronto, Toronto, Ontario, Canada
| | - Tong Wang
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and University of Toronto, Toronto, Ontario, Canada
| | - Seyed Amir Ali Adel
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and University of Toronto, Toronto, Ontario, Canada
| | - Jason P Lerch
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Wellcome Centre for Integrative Neuroimaging, Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - John G Sled
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada.,Mouse Imaging Centre at The Centre for Phenogenomics, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Brian J Nieman
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada.,Mouse Imaging Centre at The Centre for Phenogenomics, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Anne L Wheeler
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Hellewell SC, Nguyen VPB, Jayasena RN, Welton T, Grieve SM. Characteristic patterns of white matter tract injury in sport-related concussion: An image based meta-analysis. Neuroimage Clin 2020; 26:102253. [PMID: 32278315 PMCID: PMC7152675 DOI: 10.1016/j.nicl.2020.102253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/21/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022]
Abstract
Sports-related concussion (SRC) is sustained by millions of people per year, yet the spatiotemporal patterns of white matter (WM) injury remain poorly understood. Several SRC studies have implemented the standardised approach Tract-Based Spatial Statistics (TBSS). The aim of this image-based meta-analysis was to identify consensus patterns of SRC-related WM injury across TBSS studies. We included studies comparing the diffusion MRI measurement fractional anisotropy (FA) in SRC or subconcussive injury vs. controls using TBSS, as FA is the most frequently examined diffusion tensor imaging metric. Authors of eligible studies were contacted to request unthresholded statistical map outputs from TBSS, and image-based meta-analyses were performed using Seed-Based d-Mapping. Eight studies contributed to our meta-analyses, comprising 174 SRC or subconcussive injury participants and 160 controls. Our primary meta-analysis (n = 8), encompassing subjects with acute SRC (n = 2), chronic SRC (n = 4) and subconcussive injuries (n = 2) revealed dominant bilateral increased FA in the superior longitudinal fasciculus (SLF) and internal capsule. Subconcussive injury was associated with small clusters of increased and decreased FA in the arcuate fasciculus compared to control. In acute SRC, we found diffuse foci of raised FA at WM/grey matter border-zone associated with the bilateral SLF and right inferior fronto-occipital fasciculus. In contrast, chronic SRC had a pattern of deep WM alteration, asymmetrically located in the right optic radiations and arcuate fasciculus. Our findings represent the most powerful analysis of TBSS data in SRC, supporting the use of this approach to analyse diffusion data. TBSS is sensitive to WM abnormalities resulting from SRC or subconcussive injury over a range of temporal and clinical scenarios. Our data show spatially concordant patterns of WM injury unique to subconcussive, acute and chronic phases, highlighting the future utility of diffusion MRI for concussion diagnosis.
Collapse
Affiliation(s)
- Sarah C Hellewell
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Vy P B Nguyen
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Ruchira N Jayasena
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Thomas Welton
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Stuart M Grieve
- Sydney Translational Imaging Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia; Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Sydney, NSW, Australia.
| |
Collapse
|
32
|
Venkatasubramanian PN, Pina-Crespo JC, Mathews K, Rigby PH, Smith M, Duckworth JL, Wyrwicz AM, Spiess J. Initial Biphasic Fractional Anisotropy Response to Blast-Induced Mild Traumatic Brain Injury in a Mouse Model. Mil Med 2020; 185:243-247. [PMID: 32074348 PMCID: PMC7029837 DOI: 10.1093/milmed/usz307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 11/23/2022] Open
Abstract
Introduction Blast-induced mild traumatic brain injury was generated in a mouse model using a shock tube to investigate recovery and axonal injury from single blast. Methods A supersonic helium wave hit the head of anesthetized male young adult mice with a reflected pressure of 69 psi for 0.2 ms on Day 1. Subsequently, the mice were cardioperfused on Days 2, 5, or 12. The isolated brains were subjected to diffusion tensor imaging. Reduced fractional anisotropy (FA) indicated axonal injury. Results After single blast, FA showed a biphasic response in the corpus callosum with decrease on Days 2 and 12 and increase on Day 5. Conclusions Blast-induced mild traumatic brain injury in a mouse model follows a biphasic FA response within 12 days after a single blast similar to that reported for human subjects.
Collapse
Affiliation(s)
| | - Juan C Pina-Crespo
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd, La Jolla, CA 92037
| | - Kiran Mathews
- L3 Applied Technologies, 10180 Barnes Canyon Rd, San Diego, CA 92121
| | - Paul H Rigby
- L3 Applied Technologies, 10180 Barnes Canyon Rd, San Diego, CA 92121
| | - Matthew Smith
- Northshore University Health System, 1001 University Place, Evanston, IL 60201
| | - Josh L Duckworth
- Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814
| | - Alice M Wyrwicz
- Northshore University Health System, 1001 University Place, Evanston, IL 60201
| | - Joachim Spiess
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd, La Jolla, CA 92037.,Cortrop Inc., 271 Cerro Street, Encinitas, CA 92024
| |
Collapse
|
33
|
Mohamed AZ, Corrigan F, Collins-Praino LE, Plummer SL, Soni N, Nasrallah FA. Evaluating spatiotemporal microstructural alterations following diffuse traumatic brain injury. Neuroimage Clin 2019; 25:102136. [PMID: 31865019 PMCID: PMC6931220 DOI: 10.1016/j.nicl.2019.102136] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diffuse traumatic brain injury (TBI) is known to lead to microstructural changes within both white and grey matter detected in vivo with diffusion tensor imaging (DTI). Numerous studies have shown alterations in fractional anisotropy (FA) and mean diffusivity (MD) within prominent white matter tracts, but few have linked these to changes within the grey matter with confirmation via histological assessment. This is especially important as alterations in the grey matter may be predictive of long-term functional deficits. METHODS A total of 33 male Sprague Dawley rats underwent severe closed-head TBI. Eight animals underwent tensor-based morphometry (TBM) and DTI at baseline (pre-TBI), 24 hours (24 h), 7, 14, and 30 days post-TBI. Immunohistochemical analysis for the detection of ionised calcium-binding adaptor molecule 1 (IBA1) to assess microglia number and percentage of activated cells, β-amyloid precursor protein (APP) as a marker of axonal injury, and myelin basic protein (MBP) to investigate myelination was performed at each time-point. RESULTS DTI showed significant alterations in FA and RD in numerous white matter tracts including the corpus callosum, internal and external capsule, and optic tract and in the grey-matter in the cortex, thalamus, and hippocampus, with the most significant effects observed at 14 D post-TBI. TBM confirmed volumetric changes within the hippocampus and thalamus. Changes in DTI were in line with significant axonal injury noted at 24 h post-injury via immunohistochemical analysis of APP, with widespread microglial activation seen within prominent white matter tracts and the grey matter, which persisted to 30 D within the hippocampus and thalamus. Microstructural alterations in MBP+ve fibres were also noted within the hippocampus and thalamus, as well as the cortex. CONCLUSION This study confirms the widespread effects of diffuse TBI on white matter tracts which could be detected via DTI and extends these findings to key grey matter regions, with a comprehensive investigation of the whole brain. In particular, the hippocampus and thalamus appear to be vulnerable to ongoing pathology post-TBI, with DTI able to detect these alterations supporting the clinical utility in evaluating these regions post-TBI.
Collapse
Affiliation(s)
- Abdalla Z Mohamed
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia
| | - Frances Corrigan
- Head Injury Laboratory, Division of Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Lyndsey E Collins-Praino
- Cognition, Aging and Neurodegenerative Disease Laboratory (CANDL), Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Stephanie L Plummer
- Translational Neuropathology Laboratory, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Neha Soni
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia
| | - Fatima A Nasrallah
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia.
| |
Collapse
|
34
|
Clément T, Lee JB, Ichkova A, Rodriguez-Grande B, Fournier ML, Aussudre J, Ogier M, Haddad E, Canini F, Koehl M, Abrous DN, Obenaus A, Badaut J. Juvenile mild traumatic brain injury elicits distinct spatiotemporal astrocyte responses. Glia 2019; 68:528-542. [PMID: 31670865 DOI: 10.1002/glia.23736] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/14/2022]
Abstract
Mild-traumatic brain injury (mTBI) represents ~80% of all emergency room visits and increases the probability of developing long-term cognitive disorders in children. To date, molecular and cellular mechanisms underlying post-mTBI cognitive dysfunction are unknown. Astrogliosis has been shown to significantly alter astrocytes' properties following brain injury, potentially leading to significant brain dysfunction. However, such alterations have never been investigated in the context of juvenile mTBI (jmTBI). A closed-head injury model was used to study jmTBI on postnatal-day 17 mice. Astrogliosis was evaluated using glial fibrillary acidic protein (GFAP), vimentin, and nestin immunolabeling in somatosensory cortex (SSC), dentate gyrus (DG), amygdala (AMY), and infralimbic area (ILA) of prefrontal cortex in both hemispheres from 1 to 30 days postinjury (dpi). In vivo T2-weighted-imaging (T2WI) and diffusion tensor imaging (DTI) were performed at 7 and 30 dpi to examine tissue level structural alterations. Increased GFAP-labeling was observed up to 30 dpi in the ipsilateral SSC, the initial site of the impact. However, vimentin and nestin expression was not perturbed by jmTBI. The morphology of GFAP positive cells was significantly altered in the SSC, DG, AMY, and ILA up to 7 dpi that some correlated with magnetic resonance imaging changes. T2WI and DTI values were significantly altered at 30 dpi within these brain regions most prominently in regions distant from the impact site. Our data show that jmTBI triggers changes in astrocytic phenotype with a distinct spatiotemporal pattern. We speculate that the presence and time course of astrogliosis may contribute to pathophysiological processes and long-term structural alterations following jmTBI.
Collapse
Affiliation(s)
| | - Jeong B Lee
- Department of Physiology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | | | | | | | | | - Michael Ogier
- Département des Neurosciences et Sciences Cognitives, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Elizabeth Haddad
- Department of Pediatrics, University of California, Irvine, Irvine, California
| | - Frederic Canini
- Département des Neurosciences et Sciences Cognitives, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Muriel Koehl
- Neurocentre Magendie INSERM U1215, Bordeaux, France
| | | | - Andre Obenaus
- Department of Physiology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California.,Department of Pediatrics, University of California, Irvine, Irvine, California
| | - Jerome Badaut
- CNRS UMR5287, University of Bordeaux, Bordeaux, France.,Department of Physiology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
35
|
Wright DK, Brady RD, Kamnaksh A, Trezise J, Sun M, McDonald SJ, Mychasiuk R, Kolbe SC, Law M, Johnston LA, O'Brien TJ, Agoston DV, Shultz SR. Repeated mild traumatic brain injuries induce persistent changes in plasma protein and magnetic resonance imaging biomarkers in the rat. Sci Rep 2019; 9:14626. [PMID: 31602002 PMCID: PMC6787341 DOI: 10.1038/s41598-019-51267-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/28/2019] [Indexed: 01/05/2023] Open
Abstract
A single mild traumatic brain injury (mTBI) typically causes only transient symptoms, but repeated mTBI (RmTBI) is associated with cumulative and chronic neurological abnormalities. Clinical management of mTBI is challenging due to the heterogeneous, subjective and transient nature of symptoms, and thus would be aided by objective biomarkers. Promising biomarkers including advanced magnetic resonance imaging (MRI) and plasma levels of select proteins were examined here in a rat model of RmTBI. Rats received either two mild fluid percussion or sham injuries administered five days apart. Rats underwent MRI and behavioral testing 1, 3, 5, 7, and 30 days after the second injury and blood samples were collected on days 1, 7, and 30. Structural and diffusion-weighted MRI revealed that RmTBI rats had abnormalities in the cortex and corpus callosum. Proteomic analysis of plasma found that RmTBI rats had abnormalities in markers indicating axonal and vascular injury, metabolic and mitochondrial dysfunction, and glial reactivity. These changes occurred in the presence of ongoing cognitive and sensorimotor deficits in the RmTBI rats. Our findings demonstrate that RmTBI can result in chronic neurological abnormalities, provide insight into potential contributing pathophysiological mechanisms, and supports the use of MRI and plasma protein measures as RmTBI biomarkers.
Collapse
Affiliation(s)
- David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Alaa Kamnaksh
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Jack Trezise
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Scott C Kolbe
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Leigh A Johnston
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia.,Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia. .,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
36
|
Thau-Zuchman O, Ingram R, Harvey GG, Cooke T, Palmas F, Pallier PN, Brook J, Priestley JV, Dalli J, Tremoleda JL, Michael-Titus AT. A Single Injection of Docosahexaenoic Acid Induces a Pro-Resolving Lipid Mediator Profile in the Injured Tissue and a Long-Lasting Reduction in Neurological Deficit after Traumatic Brain Injury in Mice. J Neurotrauma 2019; 37:66-79. [PMID: 31256709 DOI: 10.1089/neu.2019.6420] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) can lead to life-changing neurological deficits, which reflect the fast-evolving secondary injury post-trauma. There is a need for acute protective interventions, and the aim of this study was to explore in an experimental TBI model the neuroprotective potential of a single bolus of a neuroactive omega-3 fatty acid, docosahexaenoic acid (DHA), administered in a time window feasible for emergency services. Adult mice received a controlled cortical impact injury (CCI) and neurological impairment was assessed with the modified Neurological Severity Score (mNSS) up to 28 days post-injury. DHA (500 nmol/kg) or saline were injected intravenously at 30 min post-injury. The lipid mediator profile was assessed in the injured hemisphere at 3 h post-CCI. After completion of behavioral tests and lesion assessment using magnetic resonance imaging, over 7 days or 28 days post-TBI, the tissue was analyzed by immunohistochemistry. The single DHA bolus significantly reduced the injury-induced neurological deficit and increased pro-resolving mediators in the injured brain. DHA significantly reduced lesion size, the microglia and astrocytic reaction, and oxidation, and decreased the accumulation of beta-amyloid precursor protein (APP), indicating a reduced axonal injury at 7 days post-TBI. DHA reduced the neurofilament light levels in plasma at 28 days. Therefore, an acute single bolus of DHA post-TBI, in a time window relevant for acute emergency intervention, can induce a long-lasting and significant improvement in neurological outcome, and this is accompanied by a marked upregulation of neuroprotective mediators, including the DHA-derived resolvins and protectins.
Collapse
Affiliation(s)
- Orli Thau-Zuchman
- Center for Neuroscience, Surgery and Trauma,z Queen Mary University of London, London, United Kingdom
| | - Rachael Ingram
- Center for Neuroscience, Surgery and Trauma,z Queen Mary University of London, London, United Kingdom
| | - Georgina G Harvey
- Center for Neuroscience, Surgery and Trauma,z Queen Mary University of London, London, United Kingdom
| | - Thomas Cooke
- Center for Neuroscience, Surgery and Trauma,z Queen Mary University of London, London, United Kingdom
| | - Francesco Palmas
- Lipid Mediator Unit, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Patrick N Pallier
- Center for Neuroscience, Surgery and Trauma,z Queen Mary University of London, London, United Kingdom
| | - Joseph Brook
- Center for Molecular Oncology, Queen Mary University of London, London, United Kingdom
| | - John V Priestley
- Center for Neuroscience, Surgery and Trauma,z Queen Mary University of London, London, United Kingdom
| | - Jesmond Dalli
- Lipid Mediator Unit, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Jordi L Tremoleda
- Center for Neuroscience, Surgery and Trauma,z Queen Mary University of London, London, United Kingdom
| | - Adina T Michael-Titus
- Center for Neuroscience, Surgery and Trauma,z Queen Mary University of London, London, United Kingdom
| |
Collapse
|
37
|
Kulkarni P, Morrison TR, Cai X, Iriah S, Simon N, Sabrick J, Neuroth L, Ferris CF. Neuroradiological Changes Following Single or Repetitive Mild TBI. Front Syst Neurosci 2019; 13:34. [PMID: 31427931 PMCID: PMC6688741 DOI: 10.3389/fnsys.2019.00034] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 07/10/2019] [Indexed: 11/13/2022] Open
Abstract
Objectives To test the hypothesis that there are differences in neuroradiological measures between single and repeated mild traumatic brain injury using multimodal MRI. Methods A closed-head momentum exchange model was used to produce one or three mild head injuries in young adult male rats compared to non-injured, age and weight-matched controls. Six-seven weeks post-injury, rats were studied for deficits in cognitive and motor function. Seven-eight weeks post-injury changes in brain anatomy and function were evaluated through analysis of high resolution T2 weighted images, resting-state BOLD functional connectivity, and diffusion weighted imaging with quantitative anisotropy. Results Head injuries occurred without skull fracture or signs of intracranial bleeding or contusion. There were no significant differences in cognitive or motors behaviors between experimental groups. With a single mild hit, the affected areas were limited to the caudate/putamen and central amygdala. Rats hit three times showed altered diffusivity in white matter tracts, basal ganglia, central amygdala, brainstem, and cerebellum. Comparing three hits to one hit showed a similar pattern of change underscoring a dose effect of repeated head injury on the brainstem and cerebellum. Disruption of functional connectivity was pronounced with three mild hits. The midbrain dopamine system, hippocampus, and brainstem/cerebellum showed hypoconnectivity. Interestingly, rats exposed to one hit showed enhanced functional connectivity (or hyperconnectivity) across brain sites, particularly between the olfactory system and the cerebellum. Interpretation Neuroradiological evidence of altered brain structure and function, particularly in striatal and midbrain dopaminergic areas, persists long after mild repetitive head injury. These changes may serve as biomarkers of neurodegeneration and risk for dementia later in life.
Collapse
Affiliation(s)
- Praveen Kulkarni
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Thomas R Morrison
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Xuezhu Cai
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Sade Iriah
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Neal Simon
- Azevan Pharmaceuticals, Bethlehem, PA, United States.,Department of Biological Sciences, College of Arts and Sciences, Lehigh University, Bethlehem, PA, United States
| | - Julia Sabrick
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Lucas Neuroth
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Craig F Ferris
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| |
Collapse
|
38
|
Pham L, Shultz SR, Kim HA, Brady RD, Wortman RC, Genders SG, Hale MW, O'Shea RD, Djouma E, van den Buuse M, Church JE, Christie BR, Drummond GR, Sobey CG, McDonald SJ. Mild Closed-Head Injury in Conscious Rats Causes Transient Neurobehavioral and Glial Disturbances: A Novel Experimental Model of Concussion. J Neurotrauma 2019; 36:2260-2271. [DOI: 10.1089/neu.2018.6169] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Louise Pham
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Sandy R. Shultz
- Department Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Hyun Ah Kim
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Rhys D. Brady
- Department Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Ryan C. Wortman
- Department Neuroscience, Monash University, Melbourne, Australia
| | - Shannyn G. Genders
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Matthew W. Hale
- Department of Psychology and Counseling, La Trobe University, Bundoora, Australia
| | - Ross D. O'Shea
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Elvan Djouma
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Maarten van den Buuse
- Department of Psychology and Counseling, La Trobe University, Bundoora, Australia
- Department of Pharmacology, University of Melbourne, Melbourne, Australia
- The College of Public Health, Medical, and Veterinary Sciences, James Cook University, Queensland, Australia
| | - Jarrod E. Church
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Grant R. Drummond
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Christopher G. Sobey
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Stuart J. McDonald
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
- Department Neuroscience, Monash University, Melbourne, Australia
| |
Collapse
|
39
|
Kao YCJ, Lui YW, Lu CF, Chen HL, Hsieh BY, Chen CY. Behavioral and Structural Effects of Single and Repeat Closed-Head Injury. AJNR Am J Neuroradiol 2019; 40:601-608. [PMID: 30923084 DOI: 10.3174/ajnr.a6014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/16/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE The effects of multiple head impacts, even without detectable primary injury, on subsequent behavioral impairment and structural abnormality is yet well explored. Our aim was to uncover the dynamic changes and long-term effects of single and repetitive head injury without focal contusion on tissue microstructure and macrostructure. MATERIALS AND METHODS We introduced a repetitive closed-head injury rodent model (n = 70) without parenchymal lesions. We performed a longitudinal MR imaging study during a 50-day study period (T2-weighted imaging, susceptibility-weighted imaging, and diffusion tensor imaging) as well as sequential behavioral assessment. Immunohistochemical staining for astrogliosis was examined in a subgroup of animals. Paired and independent t tests were used to evaluate the outcome change after injury and the cumulative effects of impact load, respectively. RESULTS There was no gross morphologic evidence for head injury such as skull fracture, contusion, or hemorrhage on micro-CT and MR imaging. A significant decrease of white matter fractional anisotropy from day 21 on and an increase of gray matter fractional anisotropy from day 35 on were observed. Smaller mean cortical volume in the double-injury group was shown at day 50 compared with sham and single injury (P < .05). Behavioral deficits (P < .05) in neurologic outcome, balance, and locomotor activity were also aggravated after double injury. Histologic analysis showed astrogliosis 24 hours after injury, which persisted throughout the study period. CONCLUSIONS There are measurable and dynamic changes in microstructure, cortical volume, behavior, and histopathology after both single and double injury, with more severe effects seen after double injury. This work bridges cross-sectional evidence from human subject and pathologic studies using animal models with a multi-time point, longitudinal research paradigm.
Collapse
Affiliation(s)
- Y-C J Kao
- From the Neuroscience Research Center (Y.-C.J.K., C.-Y.C.).,Translational Imaging Research Center (Y.-C.J.K., C.-Y.C.), Taipei Medical University, Taipei, Taiwan.,Department of Radiology (Y.-C.J.K., C.-Y.C.), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Radiogenomic Research Center (Y.-C.J.K., C.-Y.C.), Taipei Medical University Hospital, Taipei, Taiwan
| | - Y W Lui
- Department of Radiology (Y.W.L.), NYU School of Medicine/NYU Langone Health, New York, New York
| | - C-F Lu
- Department of Biomedical Imaging and Radiological Sciences (C.-F.L.), National Yang-Ming University, Taipei, Taiwan
| | - H-L Chen
- Departments of Medical Research (H.-L.C.)
| | - B-Y Hsieh
- Department of Biomedical Imaging and Radiological Science (B.-Y.H.), China Medical University, Taichung, Taiwan
| | - C-Y Chen
- From the Neuroscience Research Center (Y.-C.J.K., C.-Y.C.) .,Translational Imaging Research Center (Y.-C.J.K., C.-Y.C.), Taipei Medical University, Taipei, Taiwan.,Department of Radiology (Y.-C.J.K., C.-Y.C.), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Medical Imaging (C.-Y.C.).,Radiogenomic Research Center (Y.-C.J.K., C.-Y.C.), Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
40
|
Agoston DV, Kamnaksh A. Protein biomarkers of epileptogenicity after traumatic brain injury. Neurobiol Dis 2019; 123:59-68. [PMID: 30030023 PMCID: PMC6800147 DOI: 10.1016/j.nbd.2018.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a major risk factor for acquired epilepsy. Post-traumatic epilepsy (PTE) develops over time in up to 50% of patients with severe TBI. PTE is mostly unresponsive to traditional anti-seizure treatments suggesting distinct, injury-induced pathomechanisms in the development of this condition. Moderate and severe TBIs cause significant tissue damage, bleeding, neuron and glia death, as well as axonal, vascular, and metabolic abnormalities. These changes trigger a complex biological response aimed at curtailing the physical damage and restoring homeostasis and functionality. Although a positive correlation exists between the type and severity of TBI and PTE, there is only an incomplete understanding of the time-dependent sequelae of TBI pathobiologies and their role in epileptogenesis. Determining the temporal profile of protein biomarkers in the blood (serum or plasma) and cerebrospinal fluid (CSF) can help to identify pathobiologies underlying the development of PTE, high-risk individuals, and disease modifying therapies. Here we review the pathobiological sequelae of TBI in the context of blood- and CSF-based protein biomarkers, their potential role in epileptogenesis, and discuss future directions aimed at improving the diagnosis and treatment of PTE.
Collapse
Affiliation(s)
- Denes V Agoston
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, USA.
| | - Alaa Kamnaksh
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, USA
| |
Collapse
|
41
|
Papesh MA, Elliott JE, Callahan ML, Storzbach D, Lim MM, Gallun FJ. Blast Exposure Impairs Sensory Gating: Evidence from Measures of Acoustic Startle and Auditory Event-Related Potentials. J Neurotrauma 2019; 36:702-712. [PMID: 30113267 PMCID: PMC6387566 DOI: 10.1089/neu.2018.5801] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Many military service members and veterans who have been exposed to high-intensity blast waves experience traumatic brain injury (TBI), resulting in chronic auditory deficits despite normal hearing sensitivity. The current study sought to examine the neurological cause of this chronic dysfunction by testing the hypothesis that blast exposure leads to impaired filtering of sensory information at brainstem and early cortical levels. Groups of blast-exposed and non-blast-exposed participants completed self-report measures of auditory and neurobehavioral status, auditory perceptual tasks involving degraded and competing speech stimuli, and physiological measures of sensory gating, including pre-pulse inhibition and habituation of the acoustic startle reflex and electrophysiological assessment of a paired-click sensory gating paradigm. Blast-exposed participants showed significantly reduced habituation to acoustic startle stimuli and impaired filtering of redundant sensory information at the level the auditory cortex. Multiple linear regression analyses revealed that poorer sensory gating at the cortical level was primarily influenced by a diagnosis of TBI, whereas reduced habituation was primarily influenced by a diagnosis of post-traumatic stress disorder. A statistical model was created including cortical sensory gating and habituation to acoustic startle, which strongly predicted performance on a degraded speech task. These results support the hypothesis that blast exposure impairs central auditory processing via impairment of neural mechanisms underlying habituation and sensory gating.
Collapse
Affiliation(s)
- Melissa A. Papesh
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, Portland, Oregon
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
| | - Jonathan E. Elliott
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, Portland, Oregon
- Department of Neurology, Oregon Health & Science University, Portland, Oregon
| | - Megan L. Callahan
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, Portland, Oregon
- Department of Psychiatry, Oregon Health & Science University, Portland, Oregon
| | - Daniel Storzbach
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, Portland, Oregon
- Department of Neurology, Oregon Health & Science University, Portland, Oregon
- Department of Psychiatry, Oregon Health & Science University, Portland, Oregon
| | - Miranda M. Lim
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, Portland, Oregon
- Department of Neurology, Oregon Health & Science University, Portland, Oregon
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon
- Department of Behavioral Neuroscience and Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon
| | - Frederick J. Gallun
- National Center for Rehabilitative Auditory Research, VA Portland Health Care System, Portland, Oregon
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
42
|
Wortman RC, Meconi A, Neale KJ, Brady RD, McDonald SJ, Christie BR, Wright DK, Shultz SR. Diffusion MRI abnormalities in adolescent rats given repeated mild traumatic brain injury. Ann Clin Transl Neurol 2018; 5:1588-1598. [PMID: 30564624 PMCID: PMC6292182 DOI: 10.1002/acn3.667] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/12/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
Objective Mild traumatic brain injury (mTBI) is a serious health concern in the adolescent population. Repeated mTBI may result in more pronounced deficits, and has been associated with long‐term neurological consequences including neurodegeneration. As such, there is a critical need for the development of objective mTBI biomarkers to help guide medical management. Diffusion‐weighted imaging (DWI) is an advanced magnetic resonance imaging (MRI) technique that may detect brain abnormalities after mTBI. Diffusion tensor imaging (DTI) is the most commonly applied DWI method, and initial studies have reported DTI changes in mTBI patients. Furthermore, new DWI methods (e.g., track‐weighted imaging; TWI) are being developed that may also be sensitive to mTBIs, but remain to be comprehensively studied. Methods This study utilized the Awake Closed Head Injury (ACHI) model of mTBI to investigate changes in DTI and TWI following repeated mTBI in adolescent male and female rats. A total of four ACHI impacts, two/day over two consecutive days, were delivered beginning on postnatal day 25. At 1 day and 7 days postinjury, rats were euthanized and brains were collected for DWI analyses. Results Rats given repeated mTBI displayed changes in fractional anisotropy and radial diffusivity (i.e., DTI measures), as well as track density (i.e., TWI). Interpretation These findings are consistent with initial DTI findings in mTBI patients, suggest that TWI may complement DTI, support the utility of DWI measures as biomarkers in mTBI, and further validate the ACHI rat model of mTBI.
Collapse
Affiliation(s)
- Ryan C Wortman
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia.,Division of Medical Sciences University of Victoria Victoria BC V8P 5C2 Canada
| | - Alicia Meconi
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia
| | - Katie J Neale
- Division of Medical Sciences University of Victoria Victoria BC V8P 5C2 Canada
| | - Rhys D Brady
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy, and Microbiology La Trobe University Bundoora Victoria 3086 Australia
| | - Brian R Christie
- Division of Medical Sciences University of Victoria Victoria BC V8P 5C2 Canada
| | - David K Wright
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia.,The Florey Institute of Neuroscience and Mental Health Parkville Victoria 3052 Australia
| | - Sandy R Shultz
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia.,Division of Medical Sciences University of Victoria Victoria BC V8P 5C2 Canada.,Department of Medicine The Royal Melbourne Hospital The University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
43
|
Imaging biomarkers of epileptogenecity after traumatic brain injury - Preclinical frontiers. Neurobiol Dis 2018; 123:75-85. [PMID: 30321600 DOI: 10.1016/j.nbd.2018.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/04/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
Posttraumatic epilepsy (PTE) is a major neurodegenerative disease accounting for 20% of symptomatic epilepsy cases. A long latent phase offers a potential window for prophylactic treatment strategies to prevent epilepsy onset, provided that the patients at risk can be identified. Some promising imaging biomarker candidates for posttraumatic epileptogenesis have been identified, but more are required to provide the specificity and sensitivity for accurate prediction. Experimental models and preclinical longitudinal, multimodal imaging studies allow follow-up of complex cascade of events initiated by traumatic brain injury, as well as monitoring of treatment effects. Preclinical imaging data from the posttraumatic brain are rich in information, yet examination of their specific relevance to epilepsy is lacking. Accumulating evidence from ongoing preclinical studies in TBI support insight into processes involved in epileptogenesis, e.g. inflammation and changes in functional and structural brain-wide connectivity. These efforts are likely to produce both new biomarkers and treatment targets for PTE.
Collapse
|
44
|
Missault S, Anckaerts C, Blockx I, Deleye S, Van Dam D, Barriche N, De Pauw G, Aertgeerts S, Valkenburg F, De Deyn PP, Verhaeghe J, Wyffels L, Van der Linden A, Staelens S, Verhoye M, Dedeurwaerdere S. Neuroimaging of Subacute Brain Inflammation and Microstructural Changes Predicts Long-Term Functional Outcome after Experimental Traumatic Brain Injury. J Neurotrauma 2018; 36:768-788. [PMID: 30032713 DOI: 10.1089/neu.2018.5704] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
There is currently a lack of prognostic biomarkers to predict the different sequelae following traumatic brain injury (TBI). The present study investigated the hypothesis that subacute neuroinflammation and microstructural changes correlate with chronic TBI deficits. Rats were subjected to controlled cortical impact (CCI) injury, sham surgery, or skin incision (naïve). CCI-injured (n = 18) and sham-operated rats (n = 6) underwent positron emission tomography (PET) imaging with the translocator protein 18 kDa (TSPO) radioligand [18F]PBR111 and diffusion tensor imaging (DTI) in the subacute phase (≤3 weeks post-injury) to quantify inflammation and microstructural alterations. CCI-injured, sham-operated, and naïve rats (n = 8) underwent behavioral testing in the chronic phase (5.5-10 months post-injury): open field and sucrose preference tests, two one-week video-electroencephalogram (vEEG) monitoring periods, pentylenetetrazole (PTZ) seizure susceptibility tests, and a Morris water maze (MWM) test. In vivo imaging revealed pronounced neuroinflammation, decreased fractional anisotropy, and increased diffusivity in perilesional cortex and ipsilesional hippocampus of CCI-injured rats. Behavioral analysis revealed disinhibition, anhedonia, increased seizure susceptibility, and impaired learning in CCI-injured rats. Subacute TSPO expression and changes in DTI metrics significantly correlated with several chronic deficits (Pearson's |r| = 0.50-0.90). Certain specific PET and DTI parameters had good sensitivity and specificity (area under the receiver operator characteristic [ROC] curve = 0.85-1.00) to distinguish between TBI animals with and without particular behavioral deficits. Depending on the investigated behavioral deficit, PET or DTI data alone, or the combination, could very well predict the variability in functional outcome data (adjusted R2 = 0.54-1.00). Taken together, both TSPO PET and DTI seem promising prognostic biomarkers to predict different chronic TBI sequelae.
Collapse
Affiliation(s)
- Stephan Missault
- 1 Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium .,2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Cynthia Anckaerts
- 2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Ines Blockx
- 2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Steven Deleye
- 3 Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Debby Van Dam
- 4 Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Belgium; Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen (UMCG) , Groningen, The Netherlands
| | - Nora Barriche
- 1 Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Glenn De Pauw
- 1 Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Stephanie Aertgeerts
- 1 Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Femke Valkenburg
- 4 Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Belgium; Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen (UMCG) , Groningen, The Netherlands
| | - Peter Paul De Deyn
- 4 Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Belgium; Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen (UMCG) , Groningen, The Netherlands
| | - Jeroen Verhaeghe
- 3 Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Leonie Wyffels
- 3 Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium .,5 Department of Nuclear Medicine, University Hospital Antwerp , Edegem, Belgium
| | - Annemie Van der Linden
- 2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Steven Staelens
- 3 Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Marleen Verhoye
- 2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Stefanie Dedeurwaerdere
- 6 Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| |
Collapse
|
45
|
Semple BD, Zamani A, Rayner G, Shultz SR, Jones NC. Affective, neurocognitive and psychosocial disorders associated with traumatic brain injury and post-traumatic epilepsy. Neurobiol Dis 2018; 123:27-41. [PMID: 30059725 DOI: 10.1016/j.nbd.2018.07.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/08/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
Survivors of traumatic brain injury (TBI) often develop chronic neurological, neurocognitive, psychological, and psychosocial deficits that can have a profound impact on an individual's wellbeing and quality of life. TBI is also a common cause of acquired epilepsy, which is itself associated with significant behavioral morbidity. This review considers the clinical and preclinical evidence that post-traumatic epilepsy (PTE) acts as a 'second-hit' insult to worsen chronic behavioral outcomes for brain-injured patients, across the domains of emotional, cognitive, and psychosocial functioning. Surprisingly, few well-designed studies have specifically examined the relationship between seizures and behavioral outcomes after TBI. The complex mechanisms underlying these comorbidities remain incompletely understood, although many of the biological processes that precipitate seizure occurrence and epileptogenesis may also contribute to the development of chronic behavioral deficits. Further, the relationship between PTE and behavioral dysfunction is increasingly recognized to be a bidirectional one, whereby premorbid conditions are a risk factor for PTE. Clinical studies in this arena are often challenged by the confounding effects of anti-seizure medications, while preclinical studies have rarely examined an adequately extended time course to fully capture the time course of epilepsy development after a TBI. To drive the field forward towards improved treatment strategies, it is imperative that both seizures and neurobehavioral outcomes are assessed in parallel after TBI, both in patient populations and preclinical models.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Akram Zamani
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia.
| | - Genevieve Rayner
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre (Austin Campus), Heidelberg, VIC, Australia; Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, Australia; Comprehensive Epilepsy Program, Alfred Health, Australia.
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Nigel C Jones
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| |
Collapse
|
46
|
Costello DM, Kaye AH, O'Brien TJ, Shultz SR. Sport related concussion - Potential for biomarkers to improve acute management. J Clin Neurosci 2018; 56:1-6. [PMID: 30055944 DOI: 10.1016/j.jocn.2018.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/08/2018] [Indexed: 12/14/2022]
Abstract
Sport-related concussion is a common form of mild traumatic brain injury that is now recognised as a serious health issue. Growing evidence suggests concussion may result in long-term and severe neurological disabilities. Recent research into the diagnosis and management of concussion may provide new approaches to concussion management that limit the potential long-term adverse effects of concussion. This paper summarises the problem of sport-related concussion and reviews key factors (sex, age, genetics) that may modify concussion outcomes. Current sport-related concussion tools are described. Analysis of emerging methods of acute concussion diagnosis using objective fluid and neuroimaging biomarkers is provided. These new concussion biomarkers have the potential to change management of sport-related concussion.
Collapse
Affiliation(s)
- Daniel M Costello
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3010, Australia.
| | - Andrew H Kaye
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, VIC 3010, Australia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3010, Australia; Departments of Neuroscience and Medicine, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, VIC, 3004, Australia
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3010, Australia; Departments of Neuroscience and Medicine, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, VIC, 3004, Australia
| |
Collapse
|
47
|
Lyons DN, Vekaria H, Macheda T, Bakshi V, Powell DK, Gold BT, Lin AL, Sullivan PG, Bachstetter AD. A Mild Traumatic Brain Injury in Mice Produces Lasting Deficits in Brain Metabolism. J Neurotrauma 2018; 35:2435-2447. [PMID: 29808778 PMCID: PMC6196750 DOI: 10.1089/neu.2018.5663] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Metabolic uncoupling has been well-characterized during the first minutes-to-days after a traumatic brain injury (TBI), yet mitochondrial bioenergetics during the weeks-to-months after a brain injury is poorly defined, particularly after a mild TBI. We hypothesized that a closed head injury (CHI) would be associated with deficits in mitochondrial bioenergetics at one month after the injury. A significant decrease in state-III (adenosine triphosphate production) and state-V (complex-I) driven mitochondrial respiration was found at one month post-injury in adult C57Bl/6J mice. Isolation of synaptic mitochondria demonstrated that the deficit in state-III and state-V was primarily neuronal. Injured mice had a temporally consistent deficit in memory recall at one month post-injury. Using proton magnetic resonance spectroscopy (1H MRS) at 7-Tesla, we found significant decreases in phosphocreatine, N-Acetylaspartic acid, and total choline. We also found regional variations in cerebral blood flow, including both hypo- and hyperperfusion, as measured by a pseudocontinuous arterial spin labeling MR sequence. Our results highlight a chronic deficit in mitochondrial bioenergetics associated with a CHI that may lead toward a novel approach for neurorestoration after a mild TBI. MRS provides a potential biomarker for assessing the efficacy of candidate treatments targeted at improving mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Danielle N Lyons
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Hemendra Vekaria
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Teresa Macheda
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Vikas Bakshi
- 4 Sanders-Brown Center on Aging, University of Kentucky , Lexington Kentucky.,5 Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington Kentucky
| | - David K Powell
- 2 Department of Neuroscience, University of Kentucky , Lexington Kentucky.,3 Department of Biomedical Engineering, University of Kentucky , Lexington Kentucky
| | - Brian T Gold
- 2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Ai-Ling Lin
- 4 Sanders-Brown Center on Aging, University of Kentucky , Lexington Kentucky.,5 Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington Kentucky
| | - Patrick G Sullivan
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| | - Adam D Bachstetter
- 1 Spinal Cord & Brain Injury Research Center, University of Kentucky , Lexington Kentucky.,2 Department of Neuroscience, University of Kentucky , Lexington Kentucky
| |
Collapse
|
48
|
McColl TJ, Brady RD, Shultz SR, Lovick L, Webster KM, Sun M, McDonald SJ, O'Brien TJ, Semple BD. Mild Traumatic Brain Injury in Adolescent Mice Alters Skull Bone Properties to Influence a Subsequent Brain Impact at Adulthood: A Pilot Study. Front Neurol 2018; 9:372. [PMID: 29887828 PMCID: PMC5980957 DOI: 10.3389/fneur.2018.00372] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/07/2018] [Indexed: 12/24/2022] Open
Abstract
Mild traumatic brain injuries (mTBI) are common during adolescence, and limited clinical evidence suggests that a younger age at first exposure to a mTBI may lead to worse long-term outcomes. In this study, we hypothesized that a mTBI during adolescence would predispose toward poorer neurobehavioral and neuropathological outcomes after a subsequent injury at adulthood. Mice received a mild weight drop injury (mTBI) at adolescence (postnatal day 35; P35) and/or at adulthood (P70). Mice were randomized to 6 groups: 'sham' (sham-surgery at P35 only); 'P35' (mTBI at P35 only); 'P35 + sham' (mTBI at P35 + sham at P70); 'sham + P70' (sham at P35 + mTBI at P70); 'sham + sham' (sham at both P35 and P70); or 'P35 + P70' (mTBI at both P35 and P70). Acute apnea and an extended righting reflex time confirmed a mTBI injury at P35 and/or P70. Cognitive, psychosocial, and sensorimotor function was assessed over 1-week post-injury. Injured groups performed similarly to sham controls across all tasks. Immunofluorescence staining at 1 week detected an increase in glial activation markers in Sham + P70 brains only. Strikingly, 63% of Sham + P70 mice exhibited a skull fracture at impact, compared to 13% of P35 + P70 mice. Micro computed tomography of parietal skull bones found that a mTBI at P35 resulted in increased bone volume and strength, which may account for the difference in fracture incidence. In summary, a single mTBI to the adolescent mouse brain did not exacerbate the cerebral effects of a subsequent mTBI in adulthood. However, the head impact at P35 induced significant changes in skull bone structure and integrity. These novel findings support future investigation into the consequences of mTBI on skull bone.
Collapse
Affiliation(s)
- Thomas J McColl
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Rhys D Brady
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Lauren Lovick
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Kyria M Webster
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Mujun Sun
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Terence J O'Brien
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Bridgette D Semple
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
49
|
Meconi A, Wortman RC, Wright DK, Neale KJ, Clarkson M, Shultz SR, Christie BR. Repeated mild traumatic brain injury can cause acute neurologic impairment without overt structural damage in juvenile rats. PLoS One 2018; 13:e0197187. [PMID: 29738554 PMCID: PMC5940222 DOI: 10.1371/journal.pone.0197187] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 04/27/2018] [Indexed: 11/19/2022] Open
Abstract
Repeated concussion is becoming increasingly recognized as a serious public health concern around the world. Moreover, there is a greater awareness amongst health professionals of the potential for repeated pediatric concussions to detrimentally alter the structure and function of the developing brain. To better study this issue, we developed an awake closed head injury (ACHI) model that enabled repeated concussions to be performed reliably and reproducibly in juvenile rats. A neurological assessment protocol (NAP) score was generated immediately after each ACHI to help quantify the cumulative effects of repeated injury on level of consciousness, and basic motor and reflexive capacity. Here we show that we can produce a repeated ACHI (4 impacts in two days) in both male and female juvenile rats without significant mortality or pain. We show that both single and repeated injuries produce acute neurological deficits resembling clinical concussion symptoms that can be quantified using the NAP score. Behavioural analyses indicate repeated ACHI acutely impaired spatial memory in the Barnes maze, and an interesting sex effect was revealed as memory impairment correlated moderately with poorer NAP score performance in a subset of females. These cognitive impairments occurred in the absence of motor impairments on the Rotarod, or emotional changes in the open field and elevated plus mazes. Cresyl violet histology and structural magnetic resonance imaging (MRI) indicated that repeated ACHI did not produce significant structural damage. MRI also confirmed there was no volumetric loss in the cortex, hippocampus, or corpus callosum of animals at 1 or 7 days post-ACHI. Together these data indicate that the ACHI model can provide a reliable, high throughput means to study the effects of concussions in juvenile rats.
Collapse
Affiliation(s)
- Alicia Meconi
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Ryan C. Wortman
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - David K. Wright
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Katie J. Neale
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Melissa Clarkson
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Sandy R. Shultz
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Centre for Brain Health and Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
50
|
Bylicky MA, Mueller GP, Day RM. Mechanisms of Endogenous Neuroprotective Effects of Astrocytes in Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6501031. [PMID: 29805731 PMCID: PMC5901819 DOI: 10.1155/2018/6501031] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/19/2018] [Indexed: 12/11/2022]
Abstract
Astrocytes, once believed to serve only as "glue" for the structural support of neurons, have been demonstrated to serve critical functions for the maintenance and protection of neurons, especially under conditions of acute or chronic injury. There are at least seven distinct mechanisms by which astrocytes protect neurons from damage; these are (1) protection against glutamate toxicity, (2) protection against redox stress, (3) mediation of mitochondrial repair mechanisms, (4) protection against glucose-induced metabolic stress, (5) protection against iron toxicity, (6) modulation of the immune response in the brain, and (7) maintenance of tissue homeostasis in the presence of DNA damage. Astrocytes support these critical functions through specialized responses to stress or toxic conditions. The detoxifying activities of astrocytes are essential for maintenance of the microenvironment surrounding neurons and in whole tissue homeostasis. Improved understanding of the mechanisms by which astrocytes protect the brain could lead to the development of novel targets for the development of neuroprotective strategies.
Collapse
Affiliation(s)
- Michelle A. Bylicky
- Department of Anatomy, Physiology, and Genetics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Gregory P. Mueller
- Department of Anatomy, Physiology, and Genetics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, The Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|