1
|
Behan C, Greene C, Hanley N, Salla CV, Brennan D, Connolly R, Sweeney K, O'Brien D, Farrell M, Meaney J, Henshall DC, Campbell M, Doherty CP. Restoration of blood brain barrier integrity post neurosurgical resection in drug resistant epilepsy. Epilepsy Behav 2025; 168:110425. [PMID: 40267865 DOI: 10.1016/j.yebeh.2025.110425] [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: 01/01/2025] [Revised: 03/20/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
Surgery for temporal lobe epilepsy (TLE) is a well-recognised therapy for drug resistant seizures which occur in more than 50 % of patients with TLE. Blood-brain barrier (BBB) dysfunction is commonly observed in resected brain tissue from patients with treatment resistant epilepsy however, no studies have documented the recovery of BBB function following surgery. We firstly prospectively performed dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) on seven patients scheduled for temporal lobe resections before and after resection. DCE-MRI revealed BBB dysfunction in frontal and temporal brain regions. At 6-24 months post-resection, there was a reduction in the percentage of brain volume with BBB dysfunction in 5/7 patients. We then retrospectively characterised resected brain tissue from 6 further TLE cases (total n = 13) by q-RT-PCR and immunohistochemistry which revealed region-specific changes in markers of BBB integrity and inflammation with changes in CLDN12 and TJP1/2 in the hippocampus and CSF1R pathway genes in cortical and hippocampal tissue. BBB dysfunction is a key component of the molecular disruption caused by seizures and in longstanding early onset chronic epilepsy that is refractory to treatment. Here, we demonstrate for the first time the rescue of BBB dysfunction by controlling seizures after surgery.
Collapse
Affiliation(s)
- Claire Behan
- Academic Unit of Neurology, Room 5.41, Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Department of Neurology, Health Care Centre, Hospital 5, St James's Hospital, Dublin 8, Ireland; FutureNeuro Research Ireland Centre, Academic Unit of Neurology Trinity College, School of Medicine, Dublin 2, Ireland; School of Nursing and Midwifery, RSCI University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland; Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland; FutureNeuro Research Ireland Centre, Department or Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, 123 St Stephen's Green, Dublin 2, Ireland
| | - Nicole Hanley
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Carme Vila Salla
- Academic Unit of Neurology, Room 5.41, Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Department of Neurology, Health Care Centre, Hospital 5, St James's Hospital, Dublin 8, Ireland
| | - Declan Brennan
- Academic Unit of Neurology, Room 5.41, Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Department of Neurology, Health Care Centre, Hospital 5, St James's Hospital, Dublin 8, Ireland
| | - Ruairi Connolly
- Academic Unit of Neurology, Room 5.41, Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Department of Neurology, Health Care Centre, Hospital 5, St James's Hospital, Dublin 8, Ireland
| | - Kieron Sweeney
- Department of Neurosurgery, Beaumont Hospital, Dublin, Ireland
| | - Donncha O'Brien
- Department of Neurosurgery, Beaumont Hospital, Dublin, Ireland
| | - Michael Farrell
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - James Meaney
- Thomas Mitchell Centre for Advanced Medical Imaging (CAMI), St. James's Hospital & Trinity College Dublin, Dublin 8, Ireland
| | - David C Henshall
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland; FutureNeuro Research Ireland Centre, Department or Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, 123 St Stephen's Green, Dublin 2, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland; FutireNeuro Research Ireland Centre, Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland.
| | - Colin P Doherty
- Academic Unit of Neurology, Room 5.41, Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Department of Neurology, Health Care Centre, Hospital 5, St James's Hospital, Dublin 8, Ireland; FutureNeuro Research Ireland Centre, Academic Unit of Neurology Trinity College, School of Medicine, Dublin 2, Ireland
| |
Collapse
|
2
|
Zedde M, Piazza F, Pascarella R. Traumatic Brain Injury and Chronic Traumatic Encephalopathy: Not Only Trigger for Neurodegeneration but Also for Cerebral Amyloid Angiopathy? Biomedicines 2025; 13:881. [PMID: 40299513 PMCID: PMC12024568 DOI: 10.3390/biomedicines13040881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Traumatic brain injury (TBI) has been linked to the development of neurodegenerative diseases, particularly Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). This review critically assesses the relationship between TBI and cerebral amyloid angiopathy (CAA), highlighting the complexities of diagnosing CAA in the context of prior head trauma. While TBI has been shown to facilitate the accumulation of amyloid plaques and tau pathology, the interplay between neurodegenerative processes and vascular contributions remains underexplored. Epidemiological studies indicate that TBI increases the risk of various dementias, not solely AD, emphasizing the need for a comprehensive understanding of TBI-related neurodegeneration as a polypathological condition. This review further delineates the mechanisms by which TBI can lead to CAA, particularly focusing on the vascular changes that occur post-injury. It discusses the challenges associated with diagnosing CAA after TBI, particularly due to the overlapping symptoms and pathologies that complicate clinical evaluations. Notably, this review includes a clinical case that exemplifies the diagnostic challenges posed by TBI in patients with subsequent cognitive decline and vascular pathology. By synthesizing current research on TBI, CAA, and associated neurodegenerative conditions, this review aims to foster a more nuanced understanding of how these conditions interact and contribute to long-term cognitive outcomes. The findings underscore the importance of developing standardized diagnostic criteria and imaging techniques to better elucidate the relationship between TBI and vascular pathology, which could enhance clinical interventions and inform therapeutic strategies for affected individuals.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
- CAA and AD Translational Research and Biomarkers Lab, School of Medicine, University of Milano-Bicocca, 20900 Monza, Italy; (F.P.); (R.P.)
- Neuroradiology Unit, Ospedale Santa Maria della Misericordia, AULSS 5 Polesana, 45100 Rovigo, Italy
| | - Fabrizio Piazza
- CAA and AD Translational Research and Biomarkers Lab, School of Medicine, University of Milano-Bicocca, 20900 Monza, Italy; (F.P.); (R.P.)
- Neuroradiology Unit, Ospedale Santa Maria della Misericordia, AULSS 5 Polesana, 45100 Rovigo, Italy
- iCAβ International Network
| | - Rosario Pascarella
- CAA and AD Translational Research and Biomarkers Lab, School of Medicine, University of Milano-Bicocca, 20900 Monza, Italy; (F.P.); (R.P.)
- Neuroradiology Unit, Ospedale Santa Maria della Misericordia, AULSS 5 Polesana, 45100 Rovigo, Italy
- SINdem Study Group “The Inflammatory Cerebral Amyloid Angiopathy and Alzheimer’s Disease Biomarkers”
| |
Collapse
|
3
|
Cafri N, Mirloo S, Zarhin D, Kamintsky L, Serlin Y, Alhadeed L, Goldberg I, Maclean MA, Whatley B, Urman I, Doherty CP, Greene C, Behan C, Brennan D, Campbell M, Bowen C, Ben‐Arie G, Shelef I, Wandschneider B, Koepp M, Friedman A, Benninger F. Imaging blood-brain barrier dysfunction in drug-resistant epilepsy: A multi-center feasibility study. Epilepsia 2025; 66:195-206. [PMID: 39503526 PMCID: PMC11742632 DOI: 10.1111/epi.18145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE Blood-brain barrier dysfunction (BBBD) has been linked to various neurological disorders, including epilepsy. This study aims to utilize dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) to identify and compare brain regions with BBBD in patients with epilepsy (PWE) and healthy individuals. METHODS We scanned 50 drug-resistant epilepsy (DRE) patients and 58 control participants from four global specialized epilepsy centers using DCE-MRI. The presence and extent of BBBD were analyzed and compared between PWE and healthy controls. RESULTS Both greater brain volume and higher number of brain regions with BBBD were significantly present in PWE compared to healthy controls (p < 10-7). No differences in total brain volume with BBBD were observed in patients diagnosed with either focal seizures or generalized epilepsy, despite variations in the affected regions. Overall brain volume with BBBD did not differ in PWE with MRI-visible lesions compared with non-lesional cases. BBBD was observed in brain regions suspected to be related to the onset of seizures in 82% of patients (n = 39) and was typically identified in, adjacent to, and/or in the same hemisphere as the suspected epileptogenic lesion (n = 10). SIGNIFICANCE These findings are consistent with pre-clinical studies that highlight the role of BBBD in the development of DRE and identify microvascular stabilization as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Nir Cafri
- Department of Physiology, Professor Vladimir Zelman Inter‐Disciplinary Center of Brain SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
- Department of Cell Biology, Cognitive and Brain Sciences, Professor Vladimir Zelman Inter‐Disciplinary Center of Brain SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
- Department of NeurologyRabin Medical Center, Beilinson Hospital and Tel‐Aviv UniversityPetah TikvaIsrael
| | - Sheida Mirloo
- Department of Medical NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| | - Daniel Zarhin
- Department of NeurologyRabin Medical Center, Beilinson Hospital and Tel‐Aviv UniversityPetah TikvaIsrael
| | - Lyna Kamintsky
- Department of Medical NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| | - Yonatan Serlin
- Department of Medical NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
- Neurophysiology of Epilepsy UnitNational Institute of Neurological Disorders and Stroke, NIHBethesdaMarylandUSA
| | - Laith Alhadeed
- Department of Medical NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| | - Ilan Goldberg
- Department of NeurologyRabin Medical Center, Beilinson Hospital and Tel‐Aviv UniversityPetah TikvaIsrael
| | - Mark A. Maclean
- Division of NeurosurgeryDalhousie UniversityHalifaxNova ScotiaCanada
| | - Ben Whatley
- Division of NeurologyDalhousie UniversityHalifaxNova ScotiaCanada
| | - Ilia Urman
- Department of NeurologyRabin Medical Center, Beilinson Hospital and Tel‐Aviv UniversityPetah TikvaIsrael
| | - Colin P. Doherty
- Academic Unit of Neurology, School of MedicineTrinity CollegeDublinIreland
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in IrelandUniversity of Medicine and Health SciencesDublinIreland
| | - Chris Greene
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in IrelandUniversity of Medicine and Health SciencesDublinIreland
- Smurfit Institute of GeneticsTrinity College DublinDublinIreland
| | - Claire Behan
- Academic Unit of Neurology, School of MedicineTrinity CollegeDublinIreland
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in IrelandUniversity of Medicine and Health SciencesDublinIreland
| | - Declan Brennan
- Academic Unit of Neurology, School of MedicineTrinity CollegeDublinIreland
| | - Matthew Campbell
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in IrelandUniversity of Medicine and Health SciencesDublinIreland
- Smurfit Institute of GeneticsTrinity College DublinDublinIreland
| | - Chris Bowen
- Department of Diagnostic RadiologyDalhousie UniversityHalifaxNova ScotiaCanada
| | - Gal Ben‐Arie
- Department of Medical ImagingSoroka Medical CenterBeer ShevaIsrael
| | - Ilan Shelef
- Department of Clinical and Experimental EpilepsyUCL Queen Square Institute of NeurologyLondonUK
| | - Britta Wandschneider
- Department of Clinical and Experimental EpilepsyUCL Queen Square Institute of NeurologyLondonUK
- UCL‐Epilepsy Society MRI UnitChalfont Centre for EpilepsyChalfont St PeterUK
| | - Matthias Koepp
- Department of Clinical and Experimental EpilepsyUCL Queen Square Institute of NeurologyLondonUK
- UCL‐Epilepsy Society MRI UnitChalfont Centre for EpilepsyChalfont St PeterUK
| | - Alon Friedman
- Department of Physiology, Professor Vladimir Zelman Inter‐Disciplinary Center of Brain SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
- Department of Cell Biology, Cognitive and Brain Sciences, Professor Vladimir Zelman Inter‐Disciplinary Center of Brain SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
- Department of Medical NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| | - Felix Benninger
- Department of NeurologyRabin Medical Center, Beilinson Hospital and Tel‐Aviv UniversityPetah TikvaIsrael
| |
Collapse
|
4
|
Arena JD, Smith DH, Diaz Arrastia R, Cullen DK, Xiao R, Fan J, Harris DC, Lynch CE, Johnson VE. The neuropathological basis of elevated serum neurofilament light following experimental concussion. Acta Neuropathol Commun 2024; 12:189. [PMID: 39633506 PMCID: PMC11619522 DOI: 10.1186/s40478-024-01883-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Mild traumatic brain injury (mTBI) or concussion is a substantial health problem globally, with up to 15% of patients experiencing persisting symptoms that can significantly impact quality of life. Currently, the diagnosis of mTBI relies on clinical presentation with ancillary neuroimaging to exclude more severe forms of injury. However, identifying patients at risk for a poor outcome or protracted recovery is challenging, in part due to the lack of early objective tests that reflect the relevant underlying pathology. While the pathophysiology of mTBI is poorly understood, axonal damage caused by rotational forces is now recognized as an important consequence of injury. Moreover, serum measurement of the neurofilament light (NfL) protein has emerged as a potentially promising biomarker of injury. Understanding the pathological processes that determine serum NfL dynamics over time, and the ability of NfL to reflect underlying pathology will be critical for future clinical research aimed at reducing the burden of disability after mild TBI. Using a gyrencephalic model of head rotational acceleration scaled to human concussion, we demonstrate significant elevations in serum NfL, with a peak at 3 days post-injury. Moreover, increased serum NfL was detectable out to 2 weeks post-injury, with some evidence it follows a biphasic course. Subsequent quantitative histological examinations demonstrate that axonal pathology, including in the absence of neuronal somatic degeneration, was the likely source of elevated serum NfL. However, the extent of axonal pathology quantified via multiple markers did not correlate strongly with the extent of serum NfL. Interestingly, the extent of blood-brain barrier (BBB) permeability offered more robust correlations with serum NfL measured at multiple time points, suggesting BBB disruption is an important determinant of serum biomarker dynamics after mTBI. These data provide novel insights to the temporal course and pathological basis of serum NfL measurements that inform its utility as a biomarker in mTBI.
Collapse
Affiliation(s)
- John D Arena
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Douglas H Smith
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ramon Diaz Arrastia
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - D Kacy Cullen
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rui Xiao
- The Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jiaxin Fan
- The Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Danielle C Harris
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cillian E Lynch
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Victoria E Johnson
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
Hanael E, Baruch S, Altman RK, Chai O, Rapoport K, Peery D, Friedman A, Shamir MH. Blood-brain barrier dysfunction and decreased transcription of tight junction proteins in epileptic dogs. J Vet Intern Med 2024; 38:2237-2248. [PMID: 38842297 PMCID: PMC11256172 DOI: 10.1111/jvim.17099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/25/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Epilepsy in dogs and humans is associated with blood-brain barrier (BBB) dysfunction (BBBD), which may involve dysfunction of tight junction (TJ) proteins, matrix metalloproteases, and astrocytes. Imaging techniques to assess BBB integrity, to identify potential treatment strategies, have not yet been evaluated in veterinary medicine. HYPOTHESIS Some dogs with idiopathic epilepsy (IE) will exhibit BBBD. Identifying BBBD may improve antiepileptic treatment in the future. ANIMALS Twenty-seven dogs with IE and 10 healthy controls. METHODS Retrospective, prospective cohort study. Blood-brain barrier permeability (BBBP) scores were calculated for the whole brain and piriform lobe of all dogs by using dynamic contrast enhancement (DCE) magnetic resonance imaging (MRI) and subtraction enhancement analysis (SEA). Matrix metalloproteinase-9 (MMP9) activity in serum and cerebrospinal fluid (CSF) was measured and its expression in the piriform lobe was examined using immunofluorescent staining. Gene expression of TJ proteins and astrocytic transporters was analyzed in the piriform lobe. RESULTS The DCE-MRI analysis of the piriform lobe identified higher BBBP score in the IE group when compared with controls (34.5% vs 26.5%; P = .02). Activity and expression of MMP9 were increased in the serum, CSF, and piriform lobe of IE dogs as compared with controls. Gene expression of Kir4.1 and claudin-5 in the piriform lobe of IE dogs was significantly lower than in control dogs. CONCLUSIONS AND CLINICAL IMPORTANCE Our findings demonstrate BBBD in dogs with IE and were supported by increased MMP9 activity and downregulation of astrocytic potassium channels and some TJ proteins. Blood brain barrier dysfunction may be a novel antiepileptic therapy target.
Collapse
Affiliation(s)
- Erez Hanael
- Koret School of Veterinary Medicine, Neurology and NeurosurgeryThe Hebrew University of JerusalemRehovotIsrael
| | - Shelly Baruch
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, Neurology and NeurosurgeryThe Hebrew University of JerusalemRehovotIsrael
| | - Rotem Kalev Altman
- Koret School of Veterinary MedicineThe Hebrew University of JerusalemRehovotIsrael
| | - Orit Chai
- Koret School of Veterinary MedicineThe Hebrew University of JerusalemRehovotIsrael
| | - Kira Rapoport
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, Neurology and NeurosurgeryThe Hebrew University of JerusalemRehovotIsrael
| | - Dana Peery
- Koret School of Veterinary MedicineThe Hebrew University of JerusalemRehovotIsrael
| | | | - Merav H. Shamir
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, Neurology and NeurosurgeryThe Hebrew University of JerusalemRehovotIsrael
| |
Collapse
|
6
|
van Hameren G, Aboghazleh R, Parker E, Dreier JP, Kaufer D, Friedman A. From spreading depolarization to blood-brain barrier dysfunction: navigating traumatic brain injury for novel diagnosis and therapy. Nat Rev Neurol 2024; 20:408-425. [PMID: 38886512 DOI: 10.1038/s41582-024-00973-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/20/2024]
Abstract
Considerable strides in medical interventions during the acute phase of traumatic brain injury (TBI) have brought improved overall survival rates. However, following TBI, people often face ongoing, persistent and debilitating long-term complications. Here, we review the recent literature to propose possible mechanisms that lead from TBI to long-term complications, focusing particularly on the involvement of a compromised blood-brain barrier (BBB). We discuss evidence for the role of spreading depolarization as a key pathological mechanism associated with microvascular dysfunction and the transformation of astrocytes to an inflammatory phenotype. Finally, we summarize new predictive and diagnostic biomarkers and explore potential therapeutic targets for treating long-term complications of TBI.
Collapse
Affiliation(s)
- Gerben van Hameren
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Refat Aboghazleh
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| | - Ellen Parker
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Neurosurgery, Dalhousie University QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Jens P Dreier
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada.
- Department of Cell Biology, Cognitive and Brain Sciences, Zelman Inter-Disciplinary Center of Brain Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
7
|
Omo-Lamai S, Nong J, Savalia K, Kelley BJ, Wu J, Esteves-Reyes S, Chase LS, Muzykantov VR, Marcos-Contreras OA, Dollé JP, Smith DH, Brenner JS. Targeting of nanoparticles to the cerebral vasculature after traumatic brain injury. PLoS One 2024; 19:e0297451. [PMID: 38857220 PMCID: PMC11164327 DOI: 10.1371/journal.pone.0297451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/04/2024] [Indexed: 06/12/2024] Open
Abstract
Traumatic brain injury has faced numerous challenges in drug development, primarily due to the difficulty of effectively delivering drugs to the brain. However, there is a potential solution in targeted drug delivery methods involving antibody-drug conjugates or nanocarriers conjugated with targeting antibodies. Following a TBI, the blood-brain barrier (BBB) becomes permeable, which can last for years and allow the leakage of harmful plasma proteins. Consequently, an appealing approach for TBI treatment involves using drug delivery systems that utilize targeting antibodies and nanocarriers to help restore BBB integrity. In our investigation of this strategy, we examined the efficacy of free antibodies and nanocarriers targeting a specific endothelial surface marker called vascular cell adhesion molecule-1 (VCAM-1), which is known to be upregulated during inflammation. In a mouse model of TBI utilizing central fluid percussion injury, free VCAM-1 antibody did not demonstrate superior targeting when comparing sham vs. TBI brain. However, the administration of VCAM-1-targeted nanocarriers (liposomes) exhibited a 10-fold higher targeting specificity in TBI brain than in sham control. Flow cytometry and confocal microscopy analysis confirmed that VCAM-1 liposomes were primarily taken up by brain endothelial cells post-TBI. Consequently, VCAM-1 liposomes represent a promising platform for the targeted delivery of therapeutics to the brain following traumatic brain injury.
Collapse
Affiliation(s)
- Serena Omo-Lamai
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jia Nong
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Krupa Savalia
- Departments of Neurology & Neurological Surgery, University of California—Davis, Sacramento, California, United States of America
| | - Brian J. Kelley
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jichuan Wu
- Department of Medicine, Division of Pulmonary Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sahily Esteves-Reyes
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Liam S. Chase
- Department of Medicine, Division of Pulmonary Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Vladimir R. Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Oscar A. Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jean-Pierre Dollé
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Douglas H. Smith
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jacob S. Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, Division of Pulmonary Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
8
|
Brooks JS, Dickey JP. Effect of Repetitive Head Impacts on Saccade Performance in Canadian University Football Players. Clin J Sport Med 2024; 34:280-287. [PMID: 38150378 PMCID: PMC11042529 DOI: 10.1097/jsm.0000000000001202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 11/07/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE Investigate the effect of cumulative head impacts on saccade latency and errors, measured across two successive football seasons. DESIGN Participants were acquired from a sample of convenience-one Canadian university football team. Head impacts were collected during training camp, practices, eight regular season games, and four playoff games in each season. Saccade measurements were collected at five time points-before and after training camp, at midseason, after regular season, and after playoffs. SETTING Two seasons following players from a single USports football team during practices and games. PARTICIPANTS Players who completed a baseline saccade measurement and a minimum of one follow-up measurement were included in the study. A total of 127 players were monitored across two competitive seasons, including 61 players who participated in both seasons. INDEPENDENT VARIABLES Head impact measurements were collected using helmet-mounted sensors. MAIN OUTCOME MEASURES Saccade latency and number of errors were measured using high-speed video or electro-oculography. RESULTS On average, each head impact increased prosaccade latency by 5.16 × 10 -3 ms (95% confidence interval [CI], 2.26 × 10 -4 -1.00 × 10 -2 , P = 0.03) and antisaccade latency by 5.74 × 10 -3 ms (95% CI, 7.18 × 10 -4 -1.06 × 10 -2 , P = 0.02). These latency increases did not decrease between the two seasons; in fact, prosaccade latencies were 23.20 ms longer (95% CI, 19.40-27.14, P < 0.001) at the second season's baseline measurement than the first. The number of saccade errors was not affected by cumulative head impacts. CONCLUSIONS Repetitive head impacts in Canadian university football result in cumulative declines in brain function as measured by saccade performance. CLINICAL RELEVANCE Football organizations should consider implementing policies focused on reducing head impacts to improve player safety.
Collapse
Affiliation(s)
- Jeffrey S. Brooks
- School of Kinesiology, Faculty of Health Sciences, Western University, London, ON, Canada
- Department of Mechanical and Materials Engineering, Faculty of Engineering, Western University, London, ON, Canada; and
| | - James P. Dickey
- School of Kinesiology, Faculty of Health Sciences, Western University, London, ON, Canada
- School of Biomedical Engineering, Western University, London, ON, Canada
| |
Collapse
|
9
|
Greene C, Connolly R, Brennan D, Laffan A, O'Keeffe E, Zaporojan L, O'Callaghan J, Thomson B, Connolly E, Argue R, Meaney JFM, Martin-Loeches I, Long A, Cheallaigh CN, Conlon N, Doherty CP, Campbell M. Blood-brain barrier disruption and sustained systemic inflammation in individuals with long COVID-associated cognitive impairment. Nat Neurosci 2024; 27:421-432. [PMID: 38388736 PMCID: PMC10917679 DOI: 10.1038/s41593-024-01576-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/09/2024] [Indexed: 02/24/2024]
Abstract
Vascular disruption has been implicated in coronavirus disease 2019 (COVID-19) pathogenesis and may predispose to the neurological sequelae associated with long COVID, yet it is unclear how blood-brain barrier (BBB) function is affected in these conditions. Here we show that BBB disruption is evident during acute infection and in patients with long COVID with cognitive impairment, commonly referred to as brain fog. Using dynamic contrast-enhanced magnetic resonance imaging, we show BBB disruption in patients with long COVID-associated brain fog. Transcriptomic analysis of peripheral blood mononuclear cells revealed dysregulation of the coagulation system and a dampened adaptive immune response in individuals with brain fog. Accordingly, peripheral blood mononuclear cells showed increased adhesion to human brain endothelial cells in vitro, while exposure of brain endothelial cells to serum from patients with long COVID induced expression of inflammatory markers. Together, our data suggest that sustained systemic inflammation and persistent localized BBB dysfunction is a key feature of long COVID-associated brain fog.
Collapse
Affiliation(s)
- Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Ruairi Connolly
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Declan Brennan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Aoife Laffan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Eoin O'Keeffe
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Lilia Zaporojan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | | | - Bennett Thomson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Emma Connolly
- The Irish Longitudinal Study on Ageing, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Ruth Argue
- Clinical Research Facility, St James's Hospital, Dublin, Ireland
| | - James F M Meaney
- Thomas Mitchell Centre for Advanced Medical Imaging (CAMI), St. James's Hospital & Trinity College Dublin, Dublin, Ireland
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization, Trinity Centre for Health Sciences, St James's University Hospital, Dublin, Ireland
| | - Aideen Long
- Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin, Ireland
| | - Cliona Ni Cheallaigh
- Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin, Ireland
- Department of Immunology, St James's Hospital, Dublin, Ireland
| | - Niall Conlon
- Department of Immunology, St James's Hospital, Dublin, Ireland
- St James's Hospital, Tallaght University Hospital, Trinity College Dublin Allied Researchers (STTAR) Bioresource, Trinity College Dublin, Dublin, Ireland
| | - Colin P Doherty
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland.
- Academic Unit of Neurology, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland.
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland.
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
10
|
Jones CMA, Kamintsky L, Parker E, Kureshi N, Audas L, Wilson L, Champagne AA, Boulanger MM, DiStefano V, Fenerty L, Bowen C, Beyea S, Atkinson C, Clarke DB, Friedman A. Blood-Brain Barrier Dysfunction and Exposure to Head Impacts in University Football Players. Clin J Sport Med 2024; 34:61-68. [PMID: 37285595 DOI: 10.1097/jsm.0000000000001164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/21/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To investigate the link between dysfunction of the blood-brain barrier (BBB) and exposure to head impacts in concussed football athletes. DESIGN This was a prospective, observational pilot study. SETTING Canadian university football. PARTICIPANTS The study population consisted of 60 university football players, aged 18 to 25. Athletes who sustained a clinically diagnosed concussion over the course of a single football season were invited to undergo an assessment of BBB leakage. INDEPENDENT VARIABLES Head impacts detected using impact-sensing helmets were the measured variables. MAIN OUTCOME MEASURES Clinical diagnosis of concussion and BBB leakage assessed using dynamic contrast-enhanced MRI (DCE-MRI) within 1 week of concussion were the outcome measures. RESULTS Eight athletes were diagnosed with a concussion throughout the season. These athletes sustained a significantly higher number of head impacts than nonconcussed athletes. Athletes playing in the defensive back position were significantly more likely to sustain a concussion than remain concussion free. Five of the concussed athletes underwent an assessment of BBB leakage. Logistic regression analysis indicated that region-specific BBB leakage in these 5 athletes was best predicted by impacts sustained in all games and practices leading up to the concussion-as opposed to the last preconcussion impact or the impacts sustained during the game when concussion occurred. CONCLUSIONS These preliminary findings raise the potential for the hypothesis that repeated exposure to head impacts may contribute to the development of BBB pathology. Further research is needed to validate this hypothesis and to test whether BBB pathology plays a role in the sequela of repeated head trauma.
Collapse
Affiliation(s)
- Casey M A Jones
- Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Lyna Kamintsky
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Ellen Parker
- Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Nelofar Kureshi
- Division of Neurosurgery, Dalhousie University QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, NS, Canada
| | - Lorelei Audas
- Division of Neurosurgery, Dalhousie University QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, NS, Canada
| | | | | | | | - Vincent DiStefano
- School of Applied Child Psychology, McGill University, Montréal, QC, Canada
| | - Lynne Fenerty
- Division of Neurosurgery, Dalhousie University QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, NS, Canada
| | - Chris Bowen
- Biomedical Translational Imaging Centre (BIOTIC), Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
- Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
| | - Steven Beyea
- Biomedical Translational Imaging Centre (BIOTIC), Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
- Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
| | - Christina Atkinson
- Department of Family Medicine, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; and
| | - David B Clarke
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Division of Neurosurgery, Dalhousie University QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, NS, Canada
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Departments of Cognitive and Brain Sciences, Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
11
|
MacLean MA, Muradov JH, Greene R, Van Hameren G, Clarke DB, Dreier JP, Okonkwo DO, Friedman A. Memantine inhibits cortical spreading depolarization and improves neurovascular function following repetitive traumatic brain injury. SCIENCE ADVANCES 2023; 9:eadj2417. [PMID: 38091390 PMCID: PMC10848720 DOI: 10.1126/sciadv.adj2417] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Cortical spreading depolarization (CSD) is a promising target for neuroprotective therapy in traumatic brain injury (TBI). We explored the effect of NMDA receptor antagonism on electrically triggered CSDs in healthy and brain-injured animals. Rats received either one moderate or four daily repetitive mild closed head impacts (rmTBI). Ninety-three animals underwent craniectomy with electrocorticographic (ECoG) and local blood flow monitoring. In brain-injured animals, ketamine or memantine inhibited CSDs in 44 to 88% and 50 to 67% of cases, respectively. Near-DC/AC-ECoG amplitude was reduced by 44 to 75% and 52 to 67%, and duration by 39 to 87% and 61 to 78%, respectively. Daily memantine significantly reduced spreading depression and oligemia following CSD. Animals (N = 31) were randomized to either memantine (10 mg/kg) or saline with daily neurobehavioral testing. Memantine-treated animals had higher neurological scores. We demonstrate that memantine improved neurovascular function following CSD in sham and brain-injured animals. Memantine also prevented neurological decline in a blinded, preclinical randomized rmTBI trial.
Collapse
Affiliation(s)
- Mark A. MacLean
- Division of Neurosurgery, Dalhousie University, Halifax, Canada
| | - Jamil H. Muradov
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | - Ryan Greene
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | - Gerben Van Hameren
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | - David B. Clarke
- Division of Neurosurgery, Dalhousie University, Halifax, Canada
| | - Jens P. Dreier
- Center for Stroke Research Berlin, Charite University, Berlin, Germany
| | - David O. Okonkwo
- Division of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alon Friedman
- Division of Neurosurgery, Dalhousie University, Halifax, Canada
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
- Departments of Brain and Cognitive Sciences, Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
12
|
McKee AC, Mez J, Abdolmohammadi B, Butler M, Huber BR, Uretsky M, Babcock K, Cherry JD, Alvarez VE, Martin B, Tripodis Y, Palmisano JN, Cormier KA, Kubilus CA, Nicks R, Kirsch D, Mahar I, McHale L, Nowinski C, Cantu RC, Stern RA, Daneshvar D, Goldstein LE, Katz DI, Kowall NW, Dwyer B, Stein TD, Alosco ML. Neuropathologic and Clinical Findings in Young Contact Sport Athletes Exposed to Repetitive Head Impacts. JAMA Neurol 2023; 80:1037-1050. [PMID: 37639244 PMCID: PMC10463175 DOI: 10.1001/jamaneurol.2023.2907] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/06/2023] [Indexed: 08/29/2023]
Abstract
Importance Young contact sport athletes may be at risk for long-term neuropathologic disorders, including chronic traumatic encephalopathy (CTE). Objective To characterize the neuropathologic and clinical symptoms of young brain donors who were contact sport athletes. Design, Setting, and Participants This case series analyzes findings from 152 of 156 brain donors younger than 30 years identified through the Understanding Neurologic Injury and Traumatic Encephalopathy (UNITE) Brain Bank who donated their brains from February 1, 2008, to September 31, 2022. Neuropathologic evaluations, retrospective telephone clinical assessments, and online questionnaires with informants were performed blinded. Data analysis was conducted between August 2021 and June 2023. Exposures Repetitive head impacts from contact sports. Main Outcomes and Measures Gross and microscopic neuropathologic assessment, including diagnosis of CTE, based on defined diagnostic criteria; and informant-reported athletic history and informant-completed scales that assess cognitive symptoms, mood disturbances, and neurobehavioral dysregulation. Results Among the 152 deceased contact sports participants (mean [SD] age, 22.97 [4.31] years; 141 [92.8%] male) included in the study, CTE was diagnosed in 63 (41.4%; median [IQR] age, 26 [24-27] years). Of the 63 brain donors diagnosed with CTE, 60 (95.2%) were diagnosed with mild CTE (stages I or II). Brain donors who had CTE were more likely to be older (mean difference, 3.92 years; 95% CI, 2.74-5.10 years) Of the 63 athletes with CTE, 45 (71.4%) were men who played amateur sports, including American football, ice hockey, soccer, rugby, and wrestling; 1 woman with CTE played collegiate soccer. For those who played football, duration of playing career was significantly longer in those with vs without CTE (mean difference, 2.81 years; 95% CI, 1.15-4.48 years). Athletes with CTE had more ventricular dilatation, cavum septum pellucidum, thalamic notching, and perivascular pigment-laden macrophages in the frontal white matter than those without CTE. Cognitive and neurobehavioral symptoms were frequent among all brain donors. Suicide was the most common cause of death, followed by unintentional overdose; there were no differences in cause of death or clinical symptoms based on CTE status. Conclusions and Relevance This case series found that young brain donors exposed to repetitive head impacts were highly symptomatic regardless of CTE status, and the causes of symptoms in this sample are likely multifactorial. Future studies that include young brain donors unexposed to repetitive head impacts are needed to clarify the association among exposure, white matter and microvascular pathologic findings, CTE, and clinical symptoms.
Collapse
Affiliation(s)
- Ann C. McKee
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- VA Bedford Healthcare System, US Department of Veteran Affairs, Bedford, Massachusetts
- National Center for PTSD, VA Boston Healthcare, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Jesse Mez
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Bobak Abdolmohammadi
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Morgane Butler
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Bertrand Russell Huber
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- VA Bedford Healthcare System, US Department of Veteran Affairs, Bedford, Massachusetts
- National Center for PTSD, VA Boston Healthcare, Boston, Massachusetts
| | - Madeline Uretsky
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Katharine Babcock
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Anatomy and Neurobiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Jonathan D. Cherry
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Victor E. Alvarez
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- VA Bedford Healthcare System, US Department of Veteran Affairs, Bedford, Massachusetts
| | - Brett Martin
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, Massachusetts
| | - Yorghos Tripodis
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, Massachusetts
| | - Joseph N. Palmisano
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, Massachusetts
| | - Kerry A. Cormier
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- VA Bedford Healthcare System, US Department of Veteran Affairs, Bedford, Massachusetts
| | - Caroline A. Kubilus
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- VA Bedford Healthcare System, US Department of Veteran Affairs, Bedford, Massachusetts
| | - Raymond Nicks
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- VA Bedford Healthcare System, US Department of Veteran Affairs, Bedford, Massachusetts
| | - Daniel Kirsch
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Ian Mahar
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Lisa McHale
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Concussion Legacy Foundation, Boston, Massachusetts
| | - Christopher Nowinski
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Concussion Legacy Foundation, Boston, Massachusetts
| | - Robert C. Cantu
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Concussion Legacy Foundation, Boston, Massachusetts
- Department of Neurosurgery, Emerson Hospital, Concord, Massachusetts
- Department of Neurosurgery, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Robert A. Stern
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurosurgery, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Daniel Daneshvar
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Rehabilitation Medicine, Harvard Medical School, Boston, Massachusetts
| | - Lee E. Goldstein
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Psychiatry, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Biomedical, Electrical, and Computer Engineering, Boston University College of Engineering, Boston, Massachusetts
| | - Douglas I. Katz
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Braintree Rehabilitation Hospital, Braintree, Massachusetts
| | - Neil W. Kowall
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Brigid Dwyer
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Braintree Rehabilitation Hospital, Braintree, Massachusetts
| | - Thor D. Stein
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- VA Bedford Healthcare System, US Department of Veteran Affairs, Bedford, Massachusetts
| | - Michael L. Alosco
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| |
Collapse
|
13
|
Hanael E, Baruch S, Chai O, Lishitsky L, Blum T, Rapoport K, Ruggeri M, Aizenberg Z, Peery D, Meyerhoff N, Volk HA, De Decker S, Tipold A, Baumgaertner W, Friedman A, Shamir M. Quantitative analysis of magnetic resonance images for characterization of blood-brain barrier dysfunction in dogs with brain tumors. J Vet Intern Med 2023; 37:606-617. [PMID: 36847997 DOI: 10.1111/jvim.16654] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) permeability can be assessed quantitatively using advanced imaging analysis. HYPOTHESIS/OBJECTIVES Quantification and characterization of blood-brain barrier dysfunction (BBBD) patterns in dogs with brain tumors can provide useful information about tumor biology and assist in distinguishing between gliomas and meningiomas. ANIMALS Seventy-eight hospitalized dogs with brain tumors and 12 control dogs without brain tumors. METHODS In a 2-arm study, images from a prospective dynamic contrast-enhanced (DCE; n = 15) and a retrospective archived magnetic resonance imaging study (n = 63) were analyzed by DCE and subtraction enhancement analysis (SEA) to quantify BBB permeability in affected dogs relative to control dogs (n = 6 in each arm). For the SEA method, 2 ranges of postcontrast intensity differences, that is, high (HR) and low (LR), were evaluated as possible representations of 2 classes of BBB leakage. BBB score was calculated for each dog and was associated with clinical characteristics and tumor location and class. Permeability maps were generated, using the slope values (DCE) or intensity difference (SEA) of each voxel, and analyzed. RESULTS Distinctive patterns and distributions of BBBD were identified for intra- and extra-axial tumors. At a cutoff of 0.1, LR/HR BBB score ratio yielded a sensitivity of 80% and specificity of 100% in differentiating gliomas from meningiomas. CONCLUSIONS AND CLINICAL IMPORTANCE Blood-brain barrier dysfunction quantification using advanced imaging analyses has the potential to be used for assessment of brain tumor characteristics and behavior and, particularly, to help differentiating gliomas from meningiomas.
Collapse
Affiliation(s)
- Erez Hanael
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Shelly Baruch
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Orit Chai
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Liron Lishitsky
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Tal Blum
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Kira Rapoport
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Marco Ruggeri
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Zahi Aizenberg
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Dana Peery
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Nina Meyerhoff
- School of Veterinary Medicine Hannover, Small Animal Medicine and Surgery, Hannover, Germany
| | - Holger Andreas Volk
- School of Veterinary Medicine Hannover, Small Animal Medicine and Surgery, Hannover, Germany
| | - Steven De Decker
- Department of Clinical Sciences, Royal Veterinary College, University of London, Hertfordshire, UK
| | - Andrea Tipold
- School of Veterinary Medicine Hannover, Small Animal Medicine and Surgery, Hannover, Germany
| | - Wolfgang Baumgaertner
- School of Veterinary Medicine Hannover, Small Animal Medicine and Surgery, Hannover, Germany
| | - Alon Friedman
- Faculty of Medicine, Department of Medical Neuroscience Halifax, Dalhousie University, Nova Scotia, Canada.,Departments of Physiology and Cell Biology, Brain, and Cognitive Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Merav Shamir
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| |
Collapse
|
14
|
Toman E, Hodgson S, Riley M, Welbury R, Di Pietro V, Belli A. Concussion in the UK: a contemporary narrative review. Trauma Surg Acute Care Open 2022; 7:e000929. [PMID: 36274785 PMCID: PMC9582316 DOI: 10.1136/tsaco-2022-000929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/03/2022] [Indexed: 11/18/2022] Open
Abstract
Concussion has been receiving an increasing amount of media exposure following several high-profile professional sports controversies and multimillion-dollar lawsuits. The potential life-changing sequalae of concussion and the rare, but devasting, second impact syndrome have also gained much attention. Despite this, our knowledge of the pathological processes involved is limited and often extrapolated from research into more severe brain injuries. As there is no objective diagnostic test for concussion. Relying on history and examination only, the diagnosis of concussion has become the rate-limiting step in widening research into the disease. Clinical study protocols therefore frequently exclude the most vulnerable groups of patients such as those with existing cognitive impairment, concurrent intoxication, mental health issues or learning difficulties. This up-to-date narrative review aims to summarize our current concussion knowledge and provides an insight into promising avenues for future research.
Collapse
Affiliation(s)
- Emma Toman
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK,Department of Neurosurgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Sam Hodgson
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Max Riley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Richard Welbury
- School of Dentistry, University of Central Lancashire, Preston, UK
| | - Valentina Di Pietro
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Antonio Belli
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK,Department of Neurosurgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
15
|
Zhang DF, Ma H, Yang GJ, Zhang ZP, He YF, Feng MY, Shan BC, Xu XF, Ding YY, Cheng YQ. Blood–brain barrier and brain structural changes in lung cancer patients with non-brain metastases. Front Oncol 2022; 12:1015011. [PMID: 36330467 PMCID: PMC9623018 DOI: 10.3389/fonc.2022.1015011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022] Open
Abstract
Purpose To explore the relationship between blood-brain barrier (BBB) leakage and brain structure in non-brain metastasis lung cancer (LC) by magnetic resonance imaging (MRI) as well as to indicate the possibility of brain metastasis (BM) occurrence. Patients and methods MRI were performed in 75 LC patients and 29 counterpart healthy peoples (HCs). We used the Patlak pharmacokinetic model to calculate the average leakage in each brain region according to the automated anatomical labeling (AAL) atlas. The thickness of the cortex and the volumes of subcortical structures were calculated using the FreeSurfer base on Destrieux atlas. We compared the thickness of the cerebral cortex, the volumes of subcortical structures, and the leakage rates of BBB, and evaluated the relationships between these parameters. Results Compared with HCs, the leakage rates of seven brain regions were higher in patients with advanced LC (aLC). In contrast to patients with early LC (eLC), the cortical thickness of two regions was decreased in aLCs. The volumes of twelve regions were also reduced in aLCs. Brain regions with increased BBB penetration showed negative correlations with thinner cortices and reduced subcortical structure volumes (P<0.05, R=-0.2 to -0.50). BBB penetration was positively correlated with tumor size and with levels of the tumor marker CYFRA21-1 (P<0.05, R=0.2–0.70). Conclusion We found an increase in BBB permeability in non-BM aLCs that corresponded to a thinner cortical thickness and smaller subcortical structure volumes. With progression in LC staging, BBB shows higher permeability and may be more likely to develop into BM.
Collapse
Affiliation(s)
- Da-Fu Zhang
- Department of Psychiatry, the First Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Huan Ma
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
- Department of Psychiatry, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guang-Jun Yang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Zhi-Ping Zhang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Yin-Fu He
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Mao-Yang Feng
- Department of Psychiatry, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bao-Ci Shan
- Laboratory of Nuclear Analysis Techniques, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Xiu-Feng Xu
- Department of Psychiatry, the First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Research Center for Mental Disorders, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Xiu-Feng Xu, ; Ying-Ying Ding, ; Yu-Qi Cheng,
| | - Ying-Ying Ding
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
- *Correspondence: Xiu-Feng Xu, ; Ying-Ying Ding, ; Yu-Qi Cheng,
| | - Yu-Qi Cheng
- Department of Psychiatry, the First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Research Center for Mental Disorders, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Xiu-Feng Xu, ; Ying-Ying Ding, ; Yu-Qi Cheng,
| |
Collapse
|
16
|
Ware JB, Sinha S, Morrison J, Walter AE, Gugger JJ, Schneider ALC, Dabrowski C, Zamore H, Wesley L, Magdamo B, Petrov D, Kim JJ, Diaz-Arrastia R, Sandsmark DK. Dynamic contrast enhanced MRI for characterization of blood-brain-barrier dysfunction after traumatic brain injury. Neuroimage Clin 2022; 36:103236. [PMID: 36274377 PMCID: PMC9668646 DOI: 10.1016/j.nicl.2022.103236] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/30/2022] [Accepted: 10/16/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND AND PURPOSE Dysfunction of the blood-brain-barrier (BBB) is a recognized pathological consequence of traumatic brain injury (TBI) which may play an important role in chronic TBI pathophysiology. We hypothesized that BBB disruption can be detected with dynamic contrast-enhanced (DCE) MRI not only in association with focal traumatic lesions but also in normal-appearing brain tissue of TBI patients, reflecting microscopic microvascular injury. We further hypothesized that BBB integrity would improve but not completely normalize months after TBI. MATERIALS AND METHODS DCE MRI was performed in 40 adult patients a median of 23 days after hospitalized TBI and in 21 healthy controls. DCE data was analyzed using Patlak and linear models, and derived metrics of BBB leakage including the volume transfer constant (Ktrans) and the normalized permeability index (NPI) were compared between groups. BBB metrics were compared with focal lesion distribution as well as with contemporaneous measures of symptomatology and cognitive function in TBI patients. Finally, BBB metrics were examined longitudinally among 18 TBI patients who returned for a second MRI a median of 204 days postinjury. RESULTS TBI patients exhibited higher mean Ktrans (p = 0.0028) and proportion of suprathreshold NPI voxels (p = 0.001) relative to controls. Tissue-based analysis confirmed greatest TBI-related BBB disruption in association with focal lesions, however elevated Ktrans was also observed in perilesional (p = 0.011) and nonlesional (p = 0.044) regions. BBB disruption showed inverse correlation with quality of life (rho = -0.51, corrected p = 0.016). Among the subset of TBI patients who underwent a second MRI several months after the initial evaluation, metrics of BBB disruption did not differ significantly at the group level, though variable longitudinal changes were observed at the individual subject level. CONCLUSIONS This pilot investigation suggests that TBI-related BBB disruption is detectable in the early post-injury period in association with focal and diffuse brain injury.
Collapse
Affiliation(s)
- Jeffrey B Ware
- Division of Neuroradiology, Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| | - Saurabh Sinha
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Justin Morrison
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Alexa E Walter
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - James J Gugger
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Andrea L C Schneider
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Cian Dabrowski
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Hannah Zamore
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Leroy Wesley
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Brigid Magdamo
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Dmitriy Petrov
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Junghoon J Kim
- Department of Molecular, Cellular, and Biomedical Sciences, CUNY School of Medicine at The City College of New York, Townsend Harris Hall, 160 Convent Avenue, New York, NY 10031, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Danielle K Sandsmark
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
van Vliet EA, Immonen R, Prager O, Friedman A, Bankstahl JP, Wright DK, O'Brien TJ, Potschka H, Gröhn O, Harris NG. A companion to the preclinical common data elements and case report forms for in vivo rodent neuroimaging: A report of the TASK3-WG3 Neuroimaging Working Group of the ILAE/AES Joint Translational Task Force. Epilepsia Open 2022. [PMID: 35962745 DOI: 10.1002/epi4.12643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/01/2022] [Indexed: 11/10/2022] Open
Abstract
The International League Against Epilepsy/American Epilepsy Society (ILAE/AES) Joint Translational Task Force established the TASK3 working groups to create common data elements (CDEs) for various aspects of preclinical epilepsy research studies, which could help improve the standardization of experimental designs. In this article, we discuss CDEs for neuroimaging data that are collected in rodent models of epilepsy, with a focus on adult rats and mice. We provide detailed CDE tables and case report forms (CRFs), and with this companion manuscript, we discuss the methodologies for several imaging modalities and the parameters that can be collected.
Collapse
Affiliation(s)
- Erwin A van Vliet
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Riikka Immonen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Ofer Prager
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- The Royal Melbourne Hospital, The University of Melbourne, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Olli Gröhn
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Neil G Harris
- Department of Neurosurgery UCLA, UCLA Brain Injury Research Center, Los Angeles, California, USA
- Intellectual and Developmental Disabilities Research Center, UCLA, Los Angeles, California, USA
| |
Collapse
|
18
|
Janigro D, Mondello S, Posti JP, Unden J. GFAP and S100B: What You Always Wanted to Know and Never Dared to Ask. Front Neurol 2022; 13:835597. [PMID: 35386417 PMCID: PMC8977512 DOI: 10.3389/fneur.2022.835597] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/03/2022] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a major global health issue, with outcomes spanning from intracranial bleeding, debilitating sequelae, and invalidity with consequences for individuals, families, and healthcare systems. Early diagnosis of TBI by testing peripheral fluids such as blood or saliva has been the focus of many research efforts, leading to FDA approval for a bench-top assay for blood GFAP and UCH-L1 and a plasma point-of-care test for GFAP. The biomarker S100B has been included in clinical guidelines for mTBI (mTBI) in Europe. Despite these successes, several unresolved issues have been recognized, including the robustness of prior data, the presence of biomarkers in tissues beyond the central nervous system, and the time course of biomarkers in peripheral body fluids. In this review article, we present some of these issues and provide a viewpoint derived from an analysis of existing literature. We focus on two astrocytic proteins, S100B and GFAP, the most commonly employed biomarkers used in mTBI. We also offer recommendations that may translate into a broader acceptance of these clinical tools.
Collapse
Affiliation(s)
- Damir Janigro
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States.,FloTBI, Cleveland, OH, United States
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Jussi P Posti
- Department of Neurosurgery, Neurocenter, Turku Brain Injury Center, Turku University Hospital, University of Turku, Turku, Finland
| | - Johan Unden
- Department of Operation and Intensive Care, Hallands Hospital Halmstad, Lund University, Lund, Sweden
| |
Collapse
|
19
|
Hanael E, Baruch S, Chai O, Nir Z, Rapoport K, Ruggeri M, Eizenberg I, Peery D, Friedman A, Shamir MH. Detection of blood‐brain barrier dysfunction using advanced imaging methods to predict seizures in dogs with meningoencephalitis of unknown origin. J Vet Intern Med 2022; 36:702-712. [PMID: 35285550 PMCID: PMC8965229 DOI: 10.1111/jvim.16396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 12/01/2022] Open
Abstract
Background The blood‐brain barrier (BBB), which separates the intravascular and neuropil compartments, characterizes the vascular bed of the brain and is essential for its proper function. Recent advances in imaging techniques have driven the development of methods for quantitative assessment of BBB permeability. Hypothesis/Objectives Permeability of the BBB can be assessed quantitatively in dogs with meningoencephalitis of unknown origin (MUO) and its status is associated with the occurrence of seizures. Animals Forty dogs with MUO and 12 dogs without MUO. Methods Retrospective, prospective cohort study. Both dynamic contrast enhancement (DCE) and subtraction enhancement analysis (SEA) methods were used to evaluate of BBB permeability in affected (DCE, n = 8; SEA, n = 32) and control dogs (DCE, n = 6; SEA, n = 6). Association between BBB dysfunction (BBBD) score and clinical characteristics was examined. In brain regions where BBBD was identified by DCE or SEA magnetic resonance imaging (MRI) analysis, immunofluorescent staining for albumin, glial fibrillary acidic protein, ionized calcium binding adaptor molecule, and phosphorylated mothers against decapentaplegic homolog 2 were performed to detect albumin extravasation, reactive astrocytes, activated microglia, and transforming growth factor beta signaling, respectively. Results Dogs with BBBD had significantly higher seizure prevalence (72% vs 19%; P = .01) when compared to MUO dogs with no BBBD. The addition of SEA to routine MRI evaluation increased the identification rate of brain pathology in dogs with MUO from 50% to 72%. Conclusions and Clinical Importance Imaging‐based assessment of BBB integrity has the potential to predict risk of seizures in dogs with MUO.
Collapse
Affiliation(s)
- Erez Hanael
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Shelly Baruch
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Orit Chai
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Zohar Nir
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Kira Rapoport
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Marco Ruggeri
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Itzhak Eizenberg
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Dana Peery
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Brain, and Cognitive Sciences, Zlotowski Center for Neuroscience Ben‐Gurion University of the Negev Beer Sheva Israel
- Department of Medical Neuroscience, Faculty of Medicine Dalhousie University Halifax NS Canada
| | - Merav H. Shamir
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| |
Collapse
|
20
|
Salivary S100 calcium-binding protein beta (S100B) and neurofilament light (NfL) after acute exposure to repeated head impacts in collegiate water polo players. Sci Rep 2022; 12:3439. [PMID: 35236877 PMCID: PMC8891257 DOI: 10.1038/s41598-022-07241-0] [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: 09/03/2021] [Accepted: 02/04/2022] [Indexed: 11/08/2022] Open
Abstract
Blood-based biomarkers of brain injury may be useful for monitoring brain health in athletes at risk for concussions. Two putative biomarkers of sport-related concussion, neurofilament light (NfL), an axonal structural protein, and S100 calcium-binding protein beta (S100B), an astrocyte-derived protein, were measured in saliva, a biofluid which can be sampled in an athletic setting without the risks and burdens associated with blood sampled by venipuncture. Samples were collected from men’s and women’s collegiate water polo players (n = 65) before and after a competitive tournament. Head impacts were measured using sensors previously evaluated for use in water polo, and video recordings were independently reviewed for the purpose of validating impacts recorded by the sensors. Athletes sustained a total of 107 head impacts, all of which were asymptomatic (i.e., no athlete was diagnosed with a concussion or more serious). Post-tournament salivary NfL was directly associated with head impact frequency (RR = 1.151, p = 0.025) and cumulative head impact magnitude (RR = 1.008, p = 0.014), while controlling for baseline salivary NfL. Change in S100B was not associated with head impact exposure (RR < 1.001, p > 0.483). These patterns suggest that repeated head impacts may cause axonal injury, even in asymptomatic athletes.
Collapse
|
21
|
Vike NL, Bari S, Stetsiv K, Walter A, Newman S, Kawata K, Bazarian JJ, Martinovich Z, Nauman EA, Talavage TM, Papa L, Slobounov SM, Breiter HC. A preliminary model of football-related neural stress that integrates metabolomics with transcriptomics and virtual reality. iScience 2022; 25:103483. [PMID: 35106455 PMCID: PMC8786649 DOI: 10.1016/j.isci.2021.103483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/23/2021] [Accepted: 11/19/2021] [Indexed: 12/06/2022] Open
Abstract
Research suggests contact sports affect neurological health. This study used permutation-based mediation statistics to integrate measures of metabolomics, neuroinflammatory miRNAs, and virtual reality (VR)-based motor control to investigate multi-scale relationships across a season of collegiate American football. Fourteen significant mediations (six pre-season, eight across-season) were observed where metabolites always mediated the statistical relationship between miRNAs and VR-based motor control (pSobelperm≤ 0.05; total effect > 50%), suggesting a hypothesis that metabolites sit in the statistical pathway between transcriptome and behavior. Three results further supported a model of chronic neuroinflammation, consistent with mitochondrial dysfunction: (1) Mediating metabolites were consistently medium-to-long chain fatty acids, (2) tricarboxylic acid cycle metabolites decreased across-season, and (3) accumulated head acceleration events statistically moderated pre-season metabolite levels to directionally model post-season metabolite levels. These preliminary findings implicate potential mitochondrial dysfunction and highlight probable peripheral blood biomarkers underlying repetitive head impacts in otherwise healthy collegiate football athletes. Permutation-based mediation statistics can be applied to multi-scale biology problems Fatty acids were a critical link between elevated miRNAs and motor control HAEs interacted with pre-season metabolite levels to model post-season levels Together, our observations point to brain-related mitochondrial dysfunction
Collapse
Affiliation(s)
- Nicole L Vike
- Warren Wright Adolescent Center Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sumra Bari
- Warren Wright Adolescent Center Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Khrystyna Stetsiv
- Warren Wright Adolescent Center Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Alexa Walter
- Department of Kinesiology, Pennsylvania State University, University Park, PA 16801, USA
| | - Sharlene Newman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Keisuke Kawata
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, IN 47405, USA.,Program in Neuroscience, College of Arts and Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Jeffrey J Bazarian
- Department of Emergency Medicine, University of Rochester, Rochester, NY 14627, USA
| | - Zoran Martinovich
- Warren Wright Adolescent Center Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eric A Nauman
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.,School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.,Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Thomas M Talavage
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.,School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47907, USA.,Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Linda Papa
- Department of Emergency Medicine, Orlando Regional Medical Center, Orlando, FL 32806, USA
| | - Semyon M Slobounov
- Department of Kinesiology, Pennsylvania State University, University Park, PA 16801, USA
| | - Hans C Breiter
- Warren Wright Adolescent Center Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.,Laboratory of Neuroimaging and Genetics, Department of Psychiatry, Massachusetts General Hospital and Harvard School of Medicine, Boston, MA 02114, USA
| |
Collapse
|
22
|
Parker E, Aboghazleh R, Mumby G, Veksler R, Ofer J, Newton J, Smith R, Kamintsky L, Jones CMA, O'Keeffe E, Kelly E, Doelle K, Roach I, Yang LT, Moradi P, Lin JM, Gleason AJ, Atkinson C, Bowen C, Brewer KD, Doherty CP, Campbell M, Clarke DB, van Hameren G, Kaufer D, Friedman A. Concussion susceptibility is mediated by spreading depolarization-induced neurovascular dysfunction. Brain 2021; 145:2049-2063. [PMID: 34927674 PMCID: PMC9246711 DOI: 10.1093/brain/awab450] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/17/2021] [Accepted: 11/14/2021] [Indexed: 11/21/2022] Open
Abstract
The mechanisms underlying the complications of mild traumatic brain injury, including post-concussion syndrome, post-impact catastrophic death, and delayed neurodegeneration remain poorly understood. This limited pathophysiological understanding has hindered the development of diagnostic and prognostic biomarkers and has prevented the advancement of treatments for the sequelae of mild traumatic brain injury. We aimed to characterize the early electrophysiological and neurovascular alterations following repetitive mild traumatic brain injury and sought to identify new targets for the diagnosis and treatment of individuals at risk of severe post-impact complications. We combined behavioural, electrophysiological, molecular, and neuroimaging techniques in a rodent model of repetitive mild traumatic brain injury. In humans, we used dynamic contrast-enhanced MRI to quantify blood–brain barrier dysfunction after exposure to sport-related concussive mild traumatic brain injury. Rats could clearly be classified based on their susceptibility to neurological complications, including life-threatening outcomes, following repetitive injury. Susceptible animals showed greater neurological complications and had higher levels of blood–brain barrier dysfunction, transforming growth factor β (TGFβ) signalling, and neuroinflammation compared to resilient animals. Cortical spreading depolarizations were the most common electrophysiological events immediately following mild traumatic brain injury and were associated with longer recovery from impact. Triggering cortical spreading depolarizations in mild traumatic brain injured rats (but not in controls) induced blood–brain barrier dysfunction. Treatment with a selective TGFβ receptor inhibitor prevented blood–brain barrier opening and reduced injury complications. Consistent with the rodent model, blood–brain barrier dysfunction was found in a subset of human athletes following concussive mild traumatic brain injury. We provide evidence that cortical spreading depolarization, blood–brain barrier dysfunction, and pro-inflammatory TGFβ signalling are associated with severe, potentially life-threatening outcomes following repetitive mild traumatic brain injury. Diagnostic-coupled targeting of TGFβ signalling may be a novel strategy in treating mild traumatic brain injury.
Collapse
Affiliation(s)
- Ellen Parker
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada.,Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Refat Aboghazleh
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Griffin Mumby
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Ronel Veksler
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Jonathan Ofer
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Jillian Newton
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Rylan Smith
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada.,Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Lyna Kamintsky
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Casey M A Jones
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada.,Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Eoin O'Keeffe
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Eoin Kelly
- FutureNeuro SFI Research Centre, The Royal College of Surgeons in Ireland, Dublin, Ireland.,Academic Unit of Neurology, Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Klara Doelle
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Isabelle Roach
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Lynn T Yang
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Pooyan Moradi
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Jessica M Lin
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Allison J Gleason
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christina Atkinson
- Department of Family Medicine, Dalhousie University, Halifax, NS, Canada
| | - Chris Bowen
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada.,Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Kimberly D Brewer
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada.,Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Colin P Doherty
- FutureNeuro SFI Research Centre, The Royal College of Surgeons in Ireland, Dublin, Ireland.,Academic Unit of Neurology, Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - David B Clarke
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada.,Department of Surgery (Neurosurgery), Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gerben van Hameren
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,Helen Wills Neuroscience Institute & Berkeley Stem Cell Center, University of California Berkeley, Berkeley, CA 94720, USA
| | - Alon Friedman
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada.,Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
23
|
Amoo M, Henry J, O'Halloran PJ, Brennan P, Husien MB, Campbell M, Caird J, Javadpour M, Curley GF. S100B, GFAP, UCH-L1 and NSE as predictors of abnormalities on CT imaging following mild traumatic brain injury: a systematic review and meta-analysis of diagnostic test accuracy. Neurosurg Rev 2021; 45:1171-1193. [PMID: 34709508 DOI: 10.1007/s10143-021-01678-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 09/03/2021] [Accepted: 10/20/2021] [Indexed: 12/25/2022]
Abstract
Biomarkers such as calcium channel binding protein S100 subunit beta (S100B), glial fibrillary acidic protein (GFAP), ubiquitin c-terminal hydrolase L1 (UCH-L1) and neuron-specific enolase (NSE) have been proposed to aid in screening patients presenting with mild traumatic brain injury (mTBI). As such, we aimed to characterise their accuracy at various thresholds. MEDLINE, SCOPUS and EMBASE were searched, and articles reporting the diagnostic performance of included biomarkers were eligible for inclusion. Risk of bias was assessed using the QUADAS-II criteria. A meta-analysis was performed to assess the predictive value of biomarkers for imaging abnormalities on CT. A total of 2939 citations were identified, and 38 studies were included. Thirty-two studies reported data for S100B. At its conventional threshold of 0.1 μg/L, S100B had a pooled sensitivity of 91% (95%CI 87-94) and a specificity of 30% (95%CI 26-34). The optimal threshold for S100B was 0.72 μg/L, with a sensitivity of 61% (95% CI 50-72) and a specificity of 69% (95% CI 64-74). Nine studies reported data for GFAP. The optimal threshold for GFAP was 626 pg/mL, at which the sensitivity was 71% (95%CI 41-91) and specificity was 71% (95%CI 43-90). Sensitivity of GFAP was maximised at a threshold of 22 pg/mL, which had a sensitivity of 93% (95%CI 73-99) and a specificity of 36% (95%CI 12-68%). Three studies reported data for NSE and two studies for UCH-L1, which precluded meta-analysis. There is evidence to support the use of S100B as a screening tool in mild TBI, and potential advantages to the use of GFAP, which requires further investigation.
Collapse
Affiliation(s)
- Michael Amoo
- Department of Neurosurgery, Royal College of Surgeons in Ireland, Dublin, Ireland. .,National Neurosurgical Centre, Beaumont Hospital, Dublin 9, Ireland. .,Beacon Academy, Beacon Hospital, Sandyford, Dublin 18, Ireland.
| | - Jack Henry
- National Neurosurgical Centre, Beaumont Hospital, Dublin 9, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Philip J O'Halloran
- Department of Neurosurgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paul Brennan
- Department of Neurosurgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Radiology, Beaumont Hospital, Dublin 9, Ireland
| | - Mohammed Ben Husien
- Department of Neurosurgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Neurosurgical Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Matthew Campbell
- Department of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - John Caird
- Department of Neurosurgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Neurosurgical Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Mohsen Javadpour
- Department of Neurosurgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Neurosurgical Centre, Beaumont Hospital, Dublin 9, Ireland.,Department of Academic Neurology, Trinity College Dublin, Dublin 2, Ireland
| | - Gerard F Curley
- Department of Neurosurgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
24
|
Leaston J, Qiao J, Harding IC, Kulkarni P, Gharagouzloo C, Ebong E, Ferris CF. Quantitative Imaging of Blood-Brain Barrier Permeability Following Repetitive Mild Head Impacts. Front Neurol 2021; 12:729464. [PMID: 34659094 PMCID: PMC8515019 DOI: 10.3389/fneur.2021.729464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/24/2021] [Indexed: 12/28/2022] Open
Abstract
This was an exploratory study designed to evaluate the feasibility of a recently established imaging modality, quantitative ultrashort time-to-echo contrast enhanced (QUTE-CE), to follow the early pathology and vulnerability of the blood brain barrier in response to single and repetitive mild head impacts. A closed-head, momentum exchange model was used to produce three consecutive mild head impacts aimed at the forebrain separated by 24 h each. Animals were measured at baseline and within 1 h of impact. Anatomical images were collected to assess the extent of structural damage. QUTE-CE biomarkers for BBB permeability were calculated on 420,000 voxels in the brain and were registered to a bilateral 3D brain atlas providing site-specific information on 118 anatomical regions. Blood brain barrier permeability was confirmed by extravasation of labeled dextran. All head impacts occurred in the absence of any structural brain damage. A single mild head impact had measurable effects on blood brain barrier permeability and was more significant after the second and third impacts. Affected regions included the prefrontal ctx, basal ganglia, hippocampus, amygdala, and brainstem. Our findings support the concerns raised by the healthcare community regarding mild head injuries in participants in organized contact sports and military personnel in basic training and combat.
Collapse
Affiliation(s)
| | - Ju Qiao
- Center for Translational Neuroimaging, Northeastern University, Boston, MA, United States
| | - Ian C. Harding
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | | | - Codi Gharagouzloo
- Imaginostics, Inc., Cambridge, MA, United States
- Center for Translational Neuroimaging, Northeastern University, Boston, MA, United States
| | - Eno Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Craig F. Ferris
- Center for Translational Neuroimaging, Northeastern University, Boston, MA, United States
- Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| |
Collapse
|
25
|
Hier DB, Obafemi-Ajayi T, Thimgan MS, Olbricht GR, Azizi S, Allen B, Hadi BA, Wunsch DC. Blood biomarkers for mild traumatic brain injury: a selective review of unresolved issues. Biomark Res 2021; 9:70. [PMID: 34530937 PMCID: PMC8447604 DOI: 10.1186/s40364-021-00325-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/26/2021] [Indexed: 01/03/2023] Open
Abstract
Background The use of blood biomarkers after mild traumatic brain injury (mTBI) has been widely studied. We have identified eight unresolved issues related to the use of five commonly investigated blood biomarkers: neurofilament light chain, ubiquitin carboxy-terminal hydrolase-L1, tau, S100B, and glial acidic fibrillary protein. We conducted a focused literature review of unresolved issues in three areas: mode of entry into and exit from the blood, kinetics of blood biomarkers in the blood, and predictive capacity of the blood biomarkers after mTBI. Findings Although a disruption of the blood brain barrier has been demonstrated in mild and severe traumatic brain injury, biomarkers can enter the blood through pathways that do not require a breach in this barrier. A definitive accounting for the pathways that biomarkers follow from the brain to the blood after mTBI has not been performed. Although preliminary investigations of blood biomarkers kinetics after TBI are available, our current knowledge is incomplete and definitive studies are needed. Optimal sampling times for biomarkers after mTBI have not been established. Kinetic models of blood biomarkers can be informative, but more precise estimates of kinetic parameters are needed. Confounding factors for blood biomarker levels have been identified, but corrections for these factors are not routinely made. Little evidence has emerged to date to suggest that blood biomarker levels correlate with clinical measures of mTBI severity. The significance of elevated biomarker levels thirty or more days following mTBI is uncertain. Blood biomarkers have shown a modest but not definitive ability to distinguish concussed from non-concussed subjects, to detect sub-concussive hits to the head, and to predict recovery from mTBI. Blood biomarkers have performed best at distinguishing CT scan positive from CT scan negative subjects after mTBI.
Collapse
Affiliation(s)
- Daniel B Hier
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA.
| | - Tayo Obafemi-Ajayi
- Cooperative Engineering Program, Missouri State University, Springfield, MO 65897, United States
| | - Matthew S Thimgan
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO 65409, United States
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, Rolla, MO 65409, United States
| | - Sima Azizi
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA
| | - Blaine Allen
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA
| | - Bassam A Hadi
- Department of Surgery, Mercy Hospital, St. Louis MO, Missouri, MO 63141, United States
| | - Donald C Wunsch
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA.,National Science Foundation, ECCS Division, Virginia, 22314, USA
| |
Collapse
|
26
|
Calkin C, McClelland C, Cairns K, Kamintsky L, Friedman A. Insulin Resistance and Blood-Brain Barrier Dysfunction Underlie Neuroprogression in Bipolar Disorder. Front Psychiatry 2021; 12:636174. [PMID: 34113269 PMCID: PMC8185298 DOI: 10.3389/fpsyt.2021.636174] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Bipolar disorder (BD) often progresses to a more chronic and treatment resistant (neuroprogressive) course. Identifying which patients are at risk could allow for early intervention and prevention. Bipolar disorder is highly comorbid with metabolic disorders including type II diabetes mellitus (T2DM), hypertension, obesity, and dyslipidemia. Our studies have shown that insulin resistance (IR) is present in over 50% of patients with BD and that IR might underlie the progression of BD. While no confirmed predictors exist for identifying which patients with BD are likely to develop a more chronic course, emerging evidence including our own studies suggest that IR and related inflammatory pathways lead to impairments in blood-brain barrier (BBB) functioning. For the first time in living psychiatric patients, we have shown that the severity of BBB leakage is proportional to BD severity and is associated with IR. In this hypothesis paper we (i) highlight the evidence for a key role of IR in BD, (ii) show how IR in BD relates to shared inflammatory pathways, and (iii) hypothesize that these modulations result in BBB leakage and worse outcomes in BD. We further hypothesize that (iv) reversing IR through lifestyle changes or the actions of insulin sensitizing medications such as metformin, or optimizing BBB function using vascular protective drugs, such as losartan, could provide novel strategies for the prevention or treatment of neuroprogressive BD.
Collapse
Affiliation(s)
- Cynthia Calkin
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | | | | | - Lyna Kamintsky
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Alon Friedman
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
- Departments of Cell Biology and Physiology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
27
|
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: 18] [Impact Index Per Article: 4.5] [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
|
28
|
Amoo M, O'Halloran PJ, Henry J, Husien MB, Brennan P, Campbell M, Caird J, Curley GF. Permeability of the Blood-Brain Barrier after Traumatic Brain Injury; Radiological Considerations. J Neurotrauma 2021; 39:20-34. [PMID: 33632026 DOI: 10.1089/neu.2020.7545] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability, especially in young persons, and constitutes a major socioeconomic burden worldwide. It is regarded as the leading cause of mortality and morbidity in previously healthy young persons. Most of the mechanisms underpinning the development of secondary brain injury are consequences of disruption of the complex relationship between the cells and proteins constituting the neurovascular unit or a direct result of loss of integrity of the tight junctions (TJ) in the blood-brain barrier (BBB). A number of changes have been described in the BBB after TBI, including loss of TJ proteins, pericyte loss and migration, and altered expressions of water channel proteins at astrocyte end-feet processes. There is a growing research interest in identifying optimal biological and radiological biomarkers of severity of BBB dysfunction and its effects on outcomes after TBI. This review explores the microscopic changes occurring at the neurovascular unit, after TBI, and current radiological adjuncts for its evaluation in pre-clinical and clinical practice.
Collapse
Affiliation(s)
- Michael Amoo
- National Centre for Neurosurgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.,Royal College of Surgeons in Ireland, Dublin, Ireland.,Beacon Academy, Beacon Hospital, Sandyford, Dublin, Ireland
| | - Philip J O'Halloran
- Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Neurosurgery, Royal London Hospital, Whitechapel, London, United Kingdom
| | - Jack Henry
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Mohammed Ben Husien
- National Centre for Neurosurgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.,Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paul Brennan
- Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Radiology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | | | - John Caird
- National Centre for Neurosurgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Gerard F Curley
- Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
29
|
Clark AL, Weigand AJ, Bangen KJ, Merritt VC, Bondi MW, Delano-Wood L. Repetitive mTBI is associated with age-related reductions in cerebral blood flow but not cortical thickness. J Cereb Blood Flow Metab 2021; 41:431-444. [PMID: 32248731 PMCID: PMC8369996 DOI: 10.1177/0271678x19897443] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mild traumatic brain injury (mTBI) is a risk factor for Alzheimer's disease (AD), and evidence suggests cerebrovascular dysregulation initiates deleterious neurodegenerative cascades. We examined whether mTBI history alters cerebral blood flow (CBF) and cortical thickness in regions vulnerable to early AD-related changes. Seventy-four young to middle-aged Veterans (mean age = 34, range = 23-48) underwent brain scans. Participants were divided into: (1) Veteran Controls (n = 27), (2) 1-2 mTBIs (n = 26), and (2) 3+ mTBIs (n = 21) groups. Resting CBF was measured using MP-PCASL. T1 structural scans were processed with FreeSurfer. CBF and cortical thickness estimates were extracted from nine AD-vulnerable regions. Regression analyses examined whether mTBI moderated the association between age, CBF, and cortical thickness. Regressions adjusting for sex and posttraumatic stress revealed mTBI moderated the association between age and CBF of the precuneus as well as superior and inferior parietal cortices (p's < .05); increasing age was associated with lower CBF in the 3+ mTBIs group, but not in the VCs or 1-2 mTBIs groups. mTBI did not moderate associations between age and cortical thickness (p's >.05). Repetitive mTBI is associated with cerebrovascular dysfunction in AD-vulnerable regions and may accelerate pathological aging trajectories.
Collapse
Affiliation(s)
- Alexandra L Clark
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Alexandra J Weigand
- San Diego State University/University of California, San Diego (SDSU/UCSD) Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Katherine J Bangen
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Victoria C Merritt
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Mark W Bondi
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Lisa Delano-Wood
- VA San Diego Healthcare System (VASDHS), San Diego, CA, USA.,School of Medicine, Department of Psychiatry, University of California San Diego, San Diego, CA, USA.,Center of Excellence for Stress and Mental Health, VASDHS, San Diego, CA, USA
| |
Collapse
|
30
|
Veksler R, Vazana U, Serlin Y, Prager O, Ofer J, Shemen N, Fisher AM, Minaeva O, Hua N, Saar-Ashkenazy R, Benou I, Riklin-Raviv T, Parker E, Mumby G, Kamintsky L, Beyea S, Bowen CV, Shelef I, O'Keeffe E, Campbell M, Kaufer D, Goldstein LE, Friedman A. Slow blood-to-brain transport underlies enduring barrier dysfunction in American football players. Brain 2021; 143:1826-1842. [PMID: 32464655 PMCID: PMC7297017 DOI: 10.1093/brain/awaa140] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/27/2020] [Accepted: 03/11/2020] [Indexed: 12/14/2022] Open
Abstract
Repetitive mild traumatic brain injury in American football players has garnered increasing public attention following reports of chronic traumatic encephalopathy, a progressive tauopathy. While the mechanisms underlying repetitive mild traumatic brain injury-induced neurodegeneration are unknown and antemortem diagnostic tests are not available, neuropathology studies suggest a pathogenic role for microvascular injury, specifically blood–brain barrier dysfunction. Thus, our main objective was to demonstrate the effectiveness of a modified dynamic contrast-enhanced MRI approach we have developed to detect impairments in brain microvascular function. To this end, we scanned 42 adult male amateur American football players and a control group comprising 27 athletes practicing a non-contact sport and 26 non-athletes. MRI scans were also performed in 51 patients with brain pathologies involving the blood–brain barrier, namely malignant brain tumours, ischaemic stroke and haemorrhagic traumatic contusion. Based on data from prolonged scans, we generated maps that visualized the permeability value for each brain voxel. Our permeability maps revealed an increase in slow blood-to-brain transport in a subset of amateur American football players, but not in sex- and age-matched controls. The increase in permeability was region specific (white matter, midbrain peduncles, red nucleus, temporal cortex) and correlated with changes in white matter, which were confirmed by diffusion tensor imaging. Additionally, increased permeability persisted for months, as seen in players who were scanned both on- and off-season. Examination of patients with brain pathologies revealed that slow tracer accumulation characterizes areas surrounding the core of injury, which frequently shows fast blood-to-brain transport. Next, we verified our method in two rodent models: rats and mice subjected to repeated mild closed-head impact injury, and rats with vascular injury inflicted by photothrombosis. In both models, slow blood-to-brain transport was observed, which correlated with neuropathological changes. Lastly, computational simulations and direct imaging of the transport of Evans blue-albumin complex in brains of rats subjected to recurrent seizures or focal cerebrovascular injury suggest that increased cellular transport underlies the observed slow blood-to-brain transport. Taken together, our findings suggest dynamic contrast-enhanced-MRI can be used to diagnose specific microvascular pathology after traumatic brain injury and other brain pathologies.
Collapse
Affiliation(s)
- Ronel Veksler
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Udi Vazana
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yonatan Serlin
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Neurology Residency Training Program, McGill University, Montreal, QC, Canada
| | - Ofer Prager
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Jonathan Ofer
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nofar Shemen
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Andrew M Fisher
- Molecular Aging and Development Laboratory, Boston University School of Medicine, College of Engineering, Alzheimer's Disease and CTE Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Olga Minaeva
- Molecular Aging and Development Laboratory, Boston University School of Medicine, College of Engineering, Alzheimer's Disease and CTE Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Ning Hua
- Molecular Aging and Development Laboratory, Boston University School of Medicine, College of Engineering, Alzheimer's Disease and CTE Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Rotem Saar-Ashkenazy
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Psychology and the School of Social-work, Ashkelon Academic College, Israel
| | - Itay Benou
- Department of Electrical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tammy Riklin-Raviv
- Department of Electrical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ellen Parker
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Griffin Mumby
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Lyna Kamintsky
- Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Steven Beyea
- Biomedical Translational Imaging Centre (BIOTIC), IWK Health Centre and QEII Health Sciences Center, Dalhousie University, Halifax, NS, Canada
| | - Chris V Bowen
- Biomedical Translational Imaging Centre (BIOTIC), IWK Health Centre and QEII Health Sciences Center, Dalhousie University, Halifax, NS, Canada
| | - Ilan Shelef
- Department of Medical Imaging, Soroka University Medical Center, Beer-Sheva, Israel
| | - Eoin O'Keeffe
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Daniela Kaufer
- Department of Integrative Biology and the Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Lee E Goldstein
- Molecular Aging and Development Laboratory, Boston University School of Medicine, College of Engineering, Alzheimer's Disease and CTE Center, and Photonics Center, Boston University, Boston, MA, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Medical Neuroscience, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| |
Collapse
|
31
|
Kamintsky L, Beyea SD, Fisk JD, Hashmi JA, Omisade A, Calkin C, Bardouille T, Bowen C, Quraan M, Mitnitski A, Matheson K, Friedman A, Hanly JG. Blood-brain barrier leakage in systemic lupus erythematosus is associated with gray matter loss and cognitive impairment. Ann Rheum Dis 2020; 79:1580-1587. [PMID: 33004325 DOI: 10.1136/annrheumdis-2020-218004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/22/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To examine the association between blood-brain barrier (BBB) integrity, brain volume and cognitive dysfunction in adult patients with systemic lupus erythematosus (SLE). METHODS A total of 65 ambulatory patients with SLE and 9 healthy controls underwent dynamic contrast-enhanced MRI scanning, for quantitative assessment of BBB permeability. Volumetric data were extracted using the VolBrain pipeline. Global cognitive function was evaluated using a screening battery consisting of tasks falling into five broad cognitive domains, and was compared between patients with normal versus extensive BBB leakage. RESULTS Patients with SLE had significantly higher levels of BBB leakage compared with controls (p=0.04). Extensive BBB leakage (affecting over >9% of brain volume) was identified only in patients with SLE (16/65; 24.6%), who also had smaller right and left cerebral grey matter volumes compared with controls (p=0.04). Extensive BBB leakage was associated with lower global cognitive scores (p=0.02), and with the presence of impairment on one or more cognitive tasks (p=0.01). CONCLUSION Our findings provide evidence for a link between extensive BBB leakage and changes in both brain structure and cognitive function in patients with SLE. Future studies should investigate the mechanisms underlying BBB-mediated cognitive impairment, validate the diagnostic utility of BBB imaging, and determine the potential of targeting the BBB as a therapeutic strategy in patients with SLE.
Collapse
Affiliation(s)
- Lyna Kamintsky
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Steven D Beyea
- Biomedical Translational Imaging Centre (BIOTIC), Queen Elizabeth ll Health Sciences Centre, Halifax, Nova Scotia, Canada
- Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - John D Fisk
- Psychiatry, Psychology & Neuroscience and Medicine, Dalhousie University and Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Javeria A Hashmi
- Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University and Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Antonina Omisade
- Acquired Brain Injury (Epilepsy Program), Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Cynthia Calkin
- Psychiatry and Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
- Mood Disorders Clinic, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Tim Bardouille
- Physics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Chris Bowen
- Biomedical Translational Imaging Centre (BIOTIC), Queen Elizabeth ll Health Sciences Centre, Halifax, Nova Scotia, Canada
- Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Maher Quraan
- Biomedical Translational Imaging Centre (BIOTIC), Queen Elizabeth ll Health Sciences Centre, Halifax, Nova Scotia, Canada
- Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Kara Matheson
- Research Methods Unit, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Alon Friedman
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
- Cognitive and Brain Sciences, Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - John G Hanly
- Medicine and Pathology, Queen Elizabeth ll Health Sciences Centre and Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
32
|
Eyolfson E, Khan A, Mychasiuk R, Lohman AW. Microglia dynamics in adolescent traumatic brain injury. J Neuroinflammation 2020; 17:326. [PMID: 33121516 PMCID: PMC7597018 DOI: 10.1186/s12974-020-01994-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Repetitive, mild traumatic brain injuries (RmTBIs) are increasingly common in adolescents and encompass one of the largest neurological health concerns in the world. Adolescence is a critical period for brain development where RmTBIs can substantially impact neurodevelopmental trajectories and life-long neurological health. Our current understanding of RmTBI pathophysiology suggests key roles for neuroinflammation in negatively regulating neural health and function. Microglia, the brain’s resident immune population, play important roles in brain development by regulating neuronal number, and synapse formation and elimination. In response to injury, microglia activate to inflammatory phenotypes that may detract from these normal homeostatic, physiological, and developmental roles. To date, however, little is known regarding the impact of RmTBIs on microglia function during adolescent brain development. This review details key concepts surrounding RmTBI pathophysiology, adolescent brain development, and microglia dynamics in the developing brain and in response to injury, in an effort to formulate a hypothesis on how the intersection of these processes may modify long-term trajectories.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N1N4, Canada.,Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, NW, Calgary, AB, T2N4N1, Canada
| | - Asher Khan
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, NW, Calgary, AB, T2N4N1, Canada
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N1N4, Canada.,Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, NW, Calgary, AB, T2N4N1, Canada.,Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Alexander W Lohman
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, NW, Calgary, AB, T2N4N1, Canada. .,Department of Cell Biology and Anatomy, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.
| |
Collapse
|
33
|
Kulkarni P, Bhosle MR, Lu SF, Simon NS, Iriah S, Brownstein MJ, Ferris CF. Evidence of early vasogenic edema following minor head impact that can be reduced with a vasopressin V1a receptor antagonist. Brain Res Bull 2020; 165:218-227. [PMID: 33053434 DOI: 10.1016/j.brainresbull.2020.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 10/01/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Does minor head impact without signs of structural brain damage cause short-term changes in vasogenic edema as measured by an increase apparent diffusion coefficient (ADC) using diffusion weighted imaging? If so, could the increase in vasogenic edema be treated with a vasopressin V1a receptor antagonist? We hypothesized that SRX251, a highly selective V1a antagonist, would reduce vasogenic edema in response to a single minor head impact. METHODS Lightly anesthetized male rats were subjected to a sham procedure or a single hit to the forehead using a closed skull, momentum exchange model. Animals recovered in five min and were injected with saline vehicle (n = 8) or SRX251 (n = 8) at 15 min post head impact and again 7-8 hrs later. At 2 h, 6 h, and 24 h post injury, rats were anesthetized and scanned for increases in ADC, a neurological measure of vasogenic edema. Sham rats (n = 6) were exposed to anesthesia and scanned at all time points but were not hit or treated. Images were registered to and analyzed using a 3D MRI rat atlas providing site-specific data on 150 different brain areas. These brain areas were parsed into 11 major brain regions. RESULTS Untreated rats with brain injury showed a significant increase in global brain vasogenic edema as compared to sham and SRX251 treated rats. Edema peaked at 6 h in injured, untreated rats in three brain regions where changes in ADC were observed, but returned to sham levels by 24 h. There were regional variations in the time course of vasogenic edema and drug efficacy. Edema was significantly reduced in cerebellum and thalamus with SRX251 treatment while the basal ganglia did not show a response to treatment. CONCLUSION A single minor impact to the forehead causes regional increases in vasogenic edema that peak at 6 h but return to baseline within a day in a subset of brain regions. Treatment with a selective V1a receptor antagonist can reduce much of the edema.
Collapse
Affiliation(s)
- Praveen Kulkarni
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA, United States
| | - Mansi R Bhosle
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA, United States
| | - Shi-Fang Lu
- Azevan Pharmaceuticals, Bethlehem, PA, United States; Dept.of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| | - Neal S Simon
- Azevan Pharmaceuticals, Bethlehem, PA, United States; Dept.of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| | - Sade Iriah
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA, United States
| | | | - Craig F Ferris
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA, United States; Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, United States.
| |
Collapse
|
34
|
Zlokovic BV, Gottesman RF, Bernstein KE, Seshadri S, McKee A, Snyder H, Greenberg SM, Yaffe K, Schaffer CB, Yuan C, Hughes TM, Daemen MJ, Williamson JD, González HM, Schneider J, Wellington CL, Katusic ZS, Stoeckel L, Koenig JI, Corriveau RA, Fine L, Galis ZS, Reis J, Wright JD, Chen J. Vascular contributions to cognitive impairment and dementia (VCID): A report from the 2018 National Heart, Lung, and Blood Institute and National Institute of Neurological Disorders and Stroke Workshop. Alzheimers Dement 2020; 16:1714-1733. [PMID: 33030307 DOI: 10.1002/alz.12157] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) are characterized by the aging neurovascular unit being confronted with and failing to cope with biological insults due to systemic and cerebral vascular disease, proteinopathy including Alzheimer's biology, metabolic disease, or immune response, resulting in cognitive decline. This report summarizes the discussion and recommendations from a working group convened by the National Heart, Lung, and Blood Institute and the National Institute of Neurological Disorders and Stroke to evaluate the state of the field in VCID research, identify research priorities, and foster collaborations. As discussed in this report, advances in understanding the biological mechanisms of VCID across the wide spectrum of pathologies, chronic systemic comorbidities, and other risk factors may lead to potential prevention and new treatment strategies to decrease the burden of dementia. Better understanding of the social determinants of health that affect risks for both vascular disease and VCID could provide insight into strategies to reduce racial and ethnic disparities in VCID.
Collapse
Affiliation(s)
| | | | | | - Sudha Seshadri
- University of Texas Health Science Center, San Antonio and Boston University, San Antonio, Texas, USA
| | - Ann McKee
- VA Boston Healthcare System and Boston University, Boston, Massachusetts, USA
| | | | - Steven M Greenberg
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kristine Yaffe
- University of California, San Francisco, San Francisco, California, USA
| | | | - Chun Yuan
- University of Washington, Seattle, Washington, USA
| | - Timothy M Hughes
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mat J Daemen
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | - Luke Stoeckel
- National Institute on Aging, Bethesda, Maryland, USA
| | - James I Koenig
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Roderick A Corriveau
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Lawrence Fine
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Zorina S Galis
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Jared Reis
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | | | - Jue Chen
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
35
|
Brett BL, Savitz J, Nitta M, España L, Teague TK, Nelson LD, McCrea MA, Meier TB. Systemic inflammation moderates the association of prior concussion with hippocampal volume and episodic memory in high school and collegiate athletes. Brain Behav Immun 2020; 89:380-388. [PMID: 32717401 PMCID: PMC7572869 DOI: 10.1016/j.bbi.2020.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/13/2020] [Accepted: 07/19/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND There is a need to determine why prior concussion has been associated with adverse outcomes in some retired and active athletes. We examined whether serum inflammatory markers moderate the associations of prior concussion with hippocampal volumes and neurobehavioral functioning in active high school and collegiate athletes. METHODS Athletes (N = 201) completed pre-season clinical testing and serum collection (C-reactive protein [CRP]; Interleukin-6 [IL]-6; IL-1 receptor antagonist [RA]) and in-season neuroimaging. Linear mixed-effects models examined associations of prior concussion with inflammatory markers, self-reported symptoms, neurocognitive function, and hippocampal volumes. Models examined whether inflammatory markers moderated associations of concussion history and hippocampal volume and/or clinical measures. RESULTS Concussion history was significantly associated with higher symptom severity, p = 0.012, but not hippocampal volume or inflammatory markers (ps > 0.05). A significant interaction of prior concussion and CRP was observed for hippocampal volume, p = 0.006. Follow-up analyses showed that at high levels of CRP, athletes with two or more prior concussions had smaller hippocampal volume compared to athletes without prior concussion, p = 0.008. There was a significant interaction between prior concussion and levels of IL-1RA on memory scores, p = 0.044, i.e., at low levels of IL-1RA, athletes with two or more concussions had worse memory performance than those without prior concussion (p = 0.014). CONCLUSION Findings suggest that certain markers of systemic inflammation moderate the association between prior concussion and hippocampal volume and episodic memory performance. Current findings highlight potential markers for predicting at-risk individuals and identify therapeutic targets for mitigating the long-term adverse consequences of cumulative concussion.
Collapse
Affiliation(s)
- Benjamin L Brett
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, United States; Oxley College of Health Sciences, Tulsa, OK, United States
| | - Morgan Nitta
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Psychology, Marquette University, Milwaukee, WI, United States
| | - Lezlie España
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - T Kent Teague
- Departments of Surgery and Psychiatry, The University of Oklahoma, School of Community Medicine, United States; Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, United States
| | - Lindsay D Nelson
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michael A McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Timothy B Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States.
| |
Collapse
|
36
|
Rawlings S, Takechi R, Lavender AP. Effects of sub-concussion on neuropsychological performance and its potential mechanisms: A narrative review. Brain Res Bull 2020; 165:56-62. [PMID: 33011196 DOI: 10.1016/j.brainresbull.2020.09.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/12/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
Concussion and mild traumatic brain injury (mTBI) are recognised as serious medical events that are relatively common in contact sports. Recently, the seemingly non-injurious phenomenon of sub-concussion has gained interest among neuroscience researchers and early studies are showing that there may be some acute and chronic effects on brain health and function with repeated sub-concussive events of the type seen in soccer, where players strike the ball with the head, and collision sports like the rugby codes. The aim of this narrative review is to describe sub-concussion and the current understanding of short and long term effects of repeated minor impacts that have been found to occur in human and animal models. Here, potential mechanisms for cognitive dysfunction following sub-concussion and recommend directions for future research are discussed. The Potential mechanisms of injuries resulting from sub-concussion such as changes in blood brain barrier integrity, neuroinflammation, cognitive impairment, and oxidative stress damage, among other changes in central nervous system function vary considerably making understanding of the underlying causative mechanism challenging for researchers. Some evidence suggests a link between impaired cerebrovascular function and cognitive impairment which poses a potential mechanism linking the two. It is hoped that this review helps guide researchers toward a potential direction of investigations.
Collapse
Affiliation(s)
- Samuel Rawlings
- School of Physiotherapy and Exercise Science, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Curtin University, Perth, Australia; School of Public Health, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Andrew P Lavender
- School of Physiotherapy and Exercise Science, Faculty of Health Sciences, Curtin University, Perth, Australia; School of Science, Psychology and Sport, Federation University Australia, Ballarat, Australia.
| |
Collapse
|
37
|
Milikovsky DZ, Ofer J, Senatorov VV, Friedman AR, Prager O, Sheintuch L, Elazari N, Veksler R, Zelig D, Weissberg I, Bar-Klein G, Swissa E, Hanael E, Ben-Arie G, Schefenbauer O, Kamintsky L, Saar-Ashkenazy R, Shelef I, Shamir MH, Goldberg I, Glik A, Benninger F, Kaufer D, Friedman A. Paroxysmal slow cortical activity in Alzheimer's disease and epilepsy is associated with blood-brain barrier dysfunction. Sci Transl Med 2020; 11:11/521/eaaw8954. [PMID: 31801888 DOI: 10.1126/scitranslmed.aaw8954] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/13/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022]
Abstract
A growing body of evidence shows that epileptic activity is frequent but often undiagnosed in patients with Alzheimer's disease (AD) and has major therapeutic implications. Here, we analyzed electroencephalogram (EEG) data from patients with AD and found an EEG signature of transient slowing of the cortical network that we termed paroxysmal slow wave events (PSWEs). The occurrence per minute of the PSWEs was correlated with level of cognitive impairment. Interictal (between seizures) PSWEs were also found in patients with epilepsy, localized to cortical regions displaying blood-brain barrier (BBB) dysfunction, and in three rodent models with BBB pathology: aged mice, young 5x familial AD model, and status epilepticus-induced epilepsy in young rats. To investigate the potential causative role of BBB dysfunction in network modifications underlying PSWEs, we infused the serum protein albumin directly into the cerebral ventricles of naïve young rats. Infusion of albumin, but not artificial cerebrospinal fluid control, resulted in high incidence of PSWEs. Our results identify PSWEs as an EEG manifestation of nonconvulsive seizures in patients with AD and suggest BBB pathology as an underlying mechanism and as a promising therapeutic target.
Collapse
Affiliation(s)
- Dan Z Milikovsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jonathan Ofer
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Vladimir V Senatorov
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Aaron R Friedman
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ofer Prager
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Liron Sheintuch
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Netta Elazari
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ronel Veksler
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Daniel Zelig
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Itai Weissberg
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Guy Bar-Klein
- Howard Hughes Medical Institute and the Institute of Genetic Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Evyatar Swissa
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Erez Hanael
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, P.O. Box 12, Rehovot 76100, Israel
| | - Gal Ben-Arie
- Department of Medical Imaging, Soroka University Medical Center, Beer-Sheva 84105, Israel
| | - Osnat Schefenbauer
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Lyna Kamintsky
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Rotem Saar-Ashkenazy
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.,Faculty of Social Work, Ashkelon Academic College, Ashkelon 78211, Israel
| | - Ilan Shelef
- Department of Medical Imaging, Soroka University Medical Center, Beer-Sheva 84105, Israel
| | - Merav H Shamir
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, P.O. Box 12, Rehovot 76100, Israel
| | - Ilan Goldberg
- Department of Neurology, Wolfson Medical Center, Holon 58100, Israel
| | - Amir Glik
- Department of Neurology, Rabin Medical Center, Beilinson Hospital, Petach Tikva 49100, Israel.,Cognitive Neurology Clinic, Rabin Medical Center, Beilinson Hospital, Petach Tikva 49100, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Felix Benninger
- Department of Neurology, Rabin Medical Center, Beilinson Hospital, Petach Tikva 49100, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel. .,Department of Medical Neuroscience, Dalhousie University, Halifax, NS B3H4R2, Canada
| |
Collapse
|
38
|
Wu Y, Wu H, Guo X, Pluimer B, Zhao Z. Blood-Brain Barrier Dysfunction in Mild Traumatic Brain Injury: Evidence From Preclinical Murine Models. Front Physiol 2020; 11:1030. [PMID: 32973558 PMCID: PMC7472692 DOI: 10.3389/fphys.2020.01030] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022] Open
Abstract
Mild traumatic brain injury (mTBI) represents more than 80% of total TBI cases and is a robust environmental risk factor for neurodegenerative diseases including Alzheimer’s disease (AD). Besides direct neuronal injury and neuroinflammation, blood–brain barrier (BBB) dysfunction is also a hallmark event of the pathological cascades after mTBI. However, the vascular link between BBB impairment caused by mTBI and subsequent neurodegeneration remains undefined. In this review, we focus on the preclinical evidence from murine models of BBB dysfunction in mTBI and provide potential mechanistic links between BBB disruption and the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yingxi Wu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Haijian Wu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinying Guo
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Brock Pluimer
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zhen Zhao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
39
|
Yoen H, Yoo RE, Choi SH, Kim E, Oh BM, Yang D, Hwang I, Kang KM, Yun TJ, Kim JH, Sohn CH. Blood-Brain Barrier Disruption in Mild Traumatic Brain Injury Patients with Post-Concussion Syndrome: Evaluation with Region-Based Quantification of Dynamic Contrast-Enhanced MR Imaging Parameters Using Automatic Whole-Brain Segmentation. Korean J Radiol 2020; 22:118-130. [PMID: 32783413 PMCID: PMC7772380 DOI: 10.3348/kjr.2020.0016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/05/2020] [Accepted: 05/24/2020] [Indexed: 12/29/2022] Open
Abstract
Objective This study aimed to investigate the blood-brain barrier (BBB) disruption in mild traumatic brain injury (mTBI) patients with post-concussion syndrome (PCS) using dynamic contrast-enhanced (DCE) magnetic resonance (MR) imaging and automatic whole brain segmentation. Materials and Methods Forty-two consecutive mTBI patients with PCS who had undergone post-traumatic MR imaging, including DCE MR imaging, between October 2016 and April 2018, and 29 controls with DCE MR imaging were included in this retrospective study. After performing three-dimensional T1-based brain segmentation with FreeSurfer software (Laboratory for Computational Neuroimaging), the mean Ktrans and vp from DCE MR imaging (derived using the Patlak model and extended Tofts and Kermode model) were analyzed in the bilateral cerebral/cerebellar cortex, bilateral cerebral/cerebellar white matter (WM), and brainstem. Ktrans values of the mTBI patients and controls were calculated using both models to identify the model that better reflected the increased permeability owing to mTBI (tendency toward higher Ktrans values in mTBI patients than in controls). The Mann-Whitney U test and Spearman rank correlation test were performed to compare the mean Ktrans and vp between the two groups and correlate Ktrans and vp with neuropsychological tests for mTBI patients. Results Increased permeability owing to mTBI was observed in the Patlak model but not in the extended Tofts and Kermode model. In the Patlak model, the mean Ktrans in the bilateral cerebral cortex was significantly higher in mTBI patients than in controls (p = 0.042). The mean vp values in the bilateral cerebellar WM and brainstem were significantly lower in mTBI patients than in controls (p = 0.009 and p = 0.011, respectively). The mean Ktrans of the bilateral cerebral cortex was significantly higher in patients with atypical performance in the auditory continuous performance test (commission errors) than in average or good performers (p = 0.041). Conclusion BBB disruption, as reflected by the increased Ktrans and decreased vp values from the Patlak model, was observed throughout the bilateral cerebral cortex, bilateral cerebellar WM, and brainstem in mTBI patients with PCS.
Collapse
Affiliation(s)
- Heera Yoen
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Roh Eul Yoo
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| | - Seung Hong Choi
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul National University, Seoul, Korea.,School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea
| | - Eunkyung Kim
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Byung Mo Oh
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul, Korea.,National Traffic Injury Rehabilitation Hospital, Yangpyeong, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dongjin Yang
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Inpyeong Hwang
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Tae Jin Yun
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Hoon Kim
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Chul Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
40
|
Abstract
With age, the presence of multiple neuropathologies in a single individual becomes increasingly common. Given that traumatic brain injury and the repetitive head impacts (RHIs) that occur in contact sports have been associated with the development of many neurodegenerative diseases, including chronic traumatic encephalopathy (CTE), Alzheimer's disease, Lewy body disease, and amyotrophic lateral sclerosis, it is becoming critical to understand the relationship and interactions between these pathologies. In fact, comorbid pathology is common in CTE and likely influenced by both age and the severity and type of exposure to RHI as well as underlying genetic predisposition. Here, we review the major comorbid pathologies seen with CTE and in former contact sports athletes and discuss what is known about the associations between RHI, age, and the development of neuropathologies. In addition, we examine the distinction between CTE and age-related pathology including primary age-related tauopathy and age-related tau astrogliopathy.
Collapse
Affiliation(s)
- Thor D. Stein
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts,Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts,Departments of Research and Pathology & Laboratory Medicine, VA Boston Healthcare System, Boston, Massachusetts,Department of Veterans Affairs Medical Center, Bedford, Massachusetts
| | - John F. Crary
- Department of Pathology, Neuropathology Brain Bank & Research Core, Ronald M. Loeb Center for Alzheimer’s Disease, Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
41
|
Janigro D, Kawata K, Silverman E, Marchi N, Diaz-Arrastia R. Is Salivary S100B a Biomarker of Traumatic Brain Injury? A Pilot Study. Front Neurol 2020; 11:528. [PMID: 32595592 PMCID: PMC7303321 DOI: 10.3389/fneur.2020.00528] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/13/2020] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) results in short and long-term disability neurodegeneration. Mild traumatic brain injury (mTBI) represents up to 85% of head injuries; diagnosis and early management is based on computed tomography (CT) or in-hospital observation, which are time- and cost- intensive. CT involves exposure to potentially harmful ionizing radiation and >90% of the scans are negative. Blood-brain barrier (BBB) damage is suspected pathological event post-TBI contributing to long-term sequelae and a reliable and rapid point-of-care test to screen those who can safely forego acute head CT would be of great help in evaluating patients with an acute mTBI. In this pilot study, 15 adult patients with suspected TBI (mean age = 47 years, range 18–79) and 15 control subjects (mean age = 33 years, range 23–53) were enrolled. We found that the average salivary S100B level was 3.9 fold higher than blood S100B, regardless of the presence of pathology. [S100B]saliva positively correlated with [S100B]serum (Pearson' coefficient = 0.79; p < 0.01). Salivary S100B levels were as effective in differentiating TBI patients from control subjects as serum levels (Control vs. TBI: p < 0.01; Serum ROCAUC = 0.94 and Saliva ROCAUC = 0.75). I These initial results suggest that measuring salivary S100B could represent an alternative to serum S100B in the diagnosis of TBI. Larger and confirmatory trials are needed to define salivary biomarker kinetics in relation to TBI severity and the possible roles of gender, ethnicity and age in influencing salivary S100B levels.
Collapse
Affiliation(s)
- Damir Janigro
- FloTBI Inc., Cleveland, OH, United States.,Department of Physiology, Case Western Reserve University, Cleveland, OH, United States
| | - Keisuke Kawata
- Department of Kinesiology, School of Public Health, Program in Neuroscience, College of Arts and Sciences, Indiana University, Bloomington, IN, United States
| | - Erika Silverman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (CNRS/INSERM), University of Montpellier, Montpellier, France
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
42
|
Ware JB, Sandsmark DK, Diaz-Arrastia R. Unravelling the mechanisms of blood-brain barrier dysfunction in repetitive mild head injury. Brain 2020; 143:1625-1628. [PMID: 32543693 PMCID: PMC7296838 DOI: 10.1093/brain/awaa166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This scientific commentary refers to ‘Slow blood-to-brain transport underlies enduring barrier dysfunction in American football players’, by Veksler et al. (doi:10.1093/brain/awaa140).
Collapse
Affiliation(s)
- Jeffrey B Ware
- University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | | | | |
Collapse
|
43
|
Arena JD, Smith DH, Lee EB, Gibbons GS, Irwin DJ, Robinson JL, Lee VMY, Trojanowski JQ, Stewart W, Johnson VE. Tau immunophenotypes in chronic traumatic encephalopathy recapitulate those of ageing and Alzheimer's disease. Brain 2020; 143:1572-1587. [PMID: 32390044 PMCID: PMC7241956 DOI: 10.1093/brain/awaa071] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/21/2020] [Accepted: 02/02/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a risk factor for neurodegenerative disease, including chronic traumatic encephalopathy (CTE). Preliminary consensus criteria define the pathognomonic lesion of CTE as patchy tau pathology within neurons and astrocytes at the depths of cortical sulci. However, the specific tau isoform composition and post-translational modifications in CTE remain largely unexplored. Using immunohistochemistry, we performed tau phenotyping of CTE neuropathologies and compared this to a range of tau pathologies, including Alzheimer's disease, primary age-related tauopathy, ageing-related tau astrogliopathy and multiple subtypes of frontotemporal lobar degeneration with tau inclusions. Cases satisfying preliminary consensus diagnostic criteria for CTE neuropathological change (CTE-NC) were identified (athletes, n = 10; long-term survivors of moderate or severe TBI, n = 4) from the Glasgow TBI Archive and Penn Neurodegenerative Disease Brain Bank. In addition, material from a range of autopsy-proven ageing-associated and primary tauopathies in which there was no known history of exposure to TBI was selected as non-injured controls (n = 32). Each case was then stained with a panel of tau antibodies specific for phospho-epitopes (PHF1, CP13, AT100, pS262), microtubule-binding repeat domains (3R, 4R), truncation (Tau-C3) or conformation (GT-7, GT-38) and the extent and distribution of staining assessed. Cell types were confirmed with double immunofluorescent labelling. Results demonstrate that astroglial tau pathology in CTE is composed of 4R-immunoreactive thorn-shaped astrocytes, echoing the morphology and immunophenotype of astrocytes encountered in ageing-related tau astrogliopathy. In contrast, neurofibrillary tangles of CTE contain both 3R and 4R tau, with post-translational modifications and conformations consistent with Alzheimer's disease and primary age-related tauopathy. Our observations establish that the astroglial and neurofibrillary tau pathologies of CTE are phenotypically distinct from each other and recapitulate the tau immunophenotypes encountered in ageing and Alzheimer's disease. As such, the immunohistochemical distinction of CTE neuropathology from other mixed 3R/4R tauopathies of Alzheimer's disease and ageing may rest solely on the pattern and distribution of pathology.
Collapse
Affiliation(s)
- John D Arena
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas H Smith
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Translational Neuropathology Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Garrett S Gibbons
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John L Robinson
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Virginia M -Y Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - William Stewart
- Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Victoria E Johnson
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
44
|
O'Keeffe E, Kelly E, Liu Y, Giordano C, Wallace E, Hynes M, Tiernan S, Meagher A, Greene C, Hughes S, Burke T, Kealy J, Doyle N, Hay A, Farrell M, Grant GA, Friedman A, Veksler R, Molloy MG, Meaney JF, Pender N, Camarillo D, Doherty CP, Campbell M. Dynamic Blood-Brain Barrier Regulation in Mild Traumatic Brain Injury. J Neurotrauma 2020; 37:347-356. [PMID: 31702476 PMCID: PMC10331162 DOI: 10.1089/neu.2019.6483] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Whereas the diagnosis of moderate and severe traumatic brain injury (TBI) is readily visible on current medical imaging paradigms (magnetic resonance imaging [MRI] and computed tomography [CT] scanning), a far greater challenge is associated with the diagnosis and subsequent management of mild TBI (mTBI), especially concussion which, by definition, is characterized by a normal CT. To investigate whether the integrity of the blood-brain barrier (BBB) is altered in a high-risk population for concussions, we studied professional mixed martial arts (MMA) fighters and adolescent rugby players. Additionally, we performed the linear regression between the BBB disruption defined by increased gadolinium contrast extravasation on dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) on MRI and multiple biomechanical parameters indicating the severity of impacts recorded using instrumented mouthguards in professional MMA fighters. MMA fighters were examined pre-fight for a baseline and again within 120 h post-competitive fight, whereas rugby players were examined pre-season and again post-season or post-match in a subset of cases. DCE-MRI, serological analysis of BBB biomarkers, and an analysis of instrumented mouthguard data, was performed. Here, we provide pilot data that demonstrate disruption of the BBB in both professional MMA fighters and rugby players, dependent on the level of exposure. Our data suggest that biomechanical forces in professional MMA and adolescent rugby can lead to BBB disruption. These changes on imaging may serve as a biomarker of exposure of the brain to repetitive subconcussive forces and mTBI.
Collapse
Affiliation(s)
- Eoin O'Keeffe
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Eoin Kelly
- Department of Neurology, Health Care Centre, Hospital 5, St. James's Hospital, Dublin, Ireland
| | - Yuzhe Liu
- Department of Mechanical Engineering, Stanford University, Stanford, California
| | - Chiara Giordano
- Department of Mechanical Engineering, Stanford University, Stanford, California
| | - Eugene Wallace
- Department of Neurology, Health Care Centre, Hospital 5, St. James's Hospital, Dublin, Ireland
| | - Mark Hynes
- Personal Health, Dublin, Dublin, Ireland
| | - Stephen Tiernan
- Department of Mechanical Engineering, Technological University Dublin, Dublin, Ireland
| | - Aidan Meagher
- Department of Mechanical Engineering, Technological University Dublin, Dublin, Ireland
| | - Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | | | - Tom Burke
- Academic Unit of Neurology, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - John Kealy
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Niamh Doyle
- Department of Psychology, Beaumont Hospital, Dublin, Ireland
| | - Alison Hay
- Department of Neurology, Health Care Centre, Hospital 5, St. James's Hospital, Dublin, Ireland
| | - Michael Farrell
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Gerald A. Grant
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Alon Friedman
- Department of Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ronel Veksler
- Department of Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - James F. Meaney
- Department of Radiology, St. James's Hospital, Dublin, Ireland
- Centre for Advanced Medical Imaging (CAMI), St. James' Hospital, Dublin, Ireland
| | - Niall Pender
- Department of Psychology, Beaumont Hospital, Dublin, Ireland
| | - David Camarillo
- Department of Mechanical Engineering, Stanford University, Stanford, California
| | - Colin P. Doherty
- Department of Neurology, Health Care Centre, Hospital 5, St. James's Hospital, Dublin, Ireland
- Academic Unit of Neurology, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
45
|
Serlin Y, Ofer J, Ben-Arie G, Veksler R, Ifergane G, Shelef I, Minuk J, Horev A, Friedman A. Blood-Brain Barrier Leakage: A New Biomarker in Transient Ischemic Attacks. Stroke 2020; 50:1266-1269. [PMID: 31009340 DOI: 10.1161/strokeaha.119.025247] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Background and Purpose- The diagnosis of transient ischemic attack is challenging. Evidence of acute ischemia on MRI diffusion-weighted imaging is highly variable and confirmed in only about one-third of patients. This study investigated the significance of blood-brain barrier dysfunction (BBBD) mapping in patients with transient neurological deficits, as a diagnostic and prognostic biomarker required for risk stratification and stroke prevention. Methods- We used dynamic contrast-enhanced MRI to quantitatively map BBBD in a prospective cohort study of 57 patients diagnosed with transient ischemic attack/minor stroke and 50 healthy controls. Results- Brain volume with BBBD was significantly higher in patients compared with controls ( P=0.002). BBBD localization corresponded with the clinical presentation in 41 patients (72%) and was more extensive in patients with acute infarct on diffusion-weighted imaging ( P=0.05). Patients who developed new stroke during follow-up had a significantly greater BBBD at the initial presentation ( P=0.03) with a risk ratio of 5.35 for recurrent stroke. Conclusions- This is the first description of the extent and localization of BBBD in patients with transient ischemic attack/minor stroke. We propose BBBD mapping as a valuable tool for detection of subtle brain ischemia and a promising predictive biomarker required for risk stratification and stroke prevention.
Collapse
Affiliation(s)
- Yonatan Serlin
- From the Neurology Residency Training Program (Y.S.), McGill University, Montreal, QC, Canada
| | - Jonathan Ofer
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel (J.O., R.V., A.F.), Soroka Medical Center, Beer-Sheva, Israel
| | - Gal Ben-Arie
- Department of Medical Imaging (G.B.-A., I.S.), Soroka Medical Center, Beer-Sheva, Israel
| | - Ronel Veksler
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel (J.O., R.V., A.F.), Soroka Medical Center, Beer-Sheva, Israel
| | - Gal Ifergane
- Department of Neurology (G.I., A.H.), Soroka Medical Center, Beer-Sheva, Israel
| | - Ilan Shelef
- Department of Medical Imaging (G.B.-A., I.S.), Soroka Medical Center, Beer-Sheva, Israel
| | - Jeffrey Minuk
- Department of Neurology and Neurosurgery, Jewish General Hospital (J.M.), McGill University, Montreal, QC, Canada
| | - Anat Horev
- Department of Neurology (G.I., A.H.), Soroka Medical Center, Beer-Sheva, Israel
| | - Alon Friedman
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel (J.O., R.V., A.F.), Soroka Medical Center, Beer-Sheva, Israel.,Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada (A.F.)
| |
Collapse
|
46
|
Senatorov VV, Friedman AR, Milikovsky DZ, Ofer J, Saar-Ashkenazy R, Charbash A, Jahan N, Chin G, Mihaly E, Lin JM, Ramsay HJ, Moghbel A, Preininger MK, Eddings CR, Harrison HV, Patel R, Shen Y, Ghanim H, Sheng H, Veksler R, Sudmant PH, Becker A, Hart B, Rogawski MA, Dillin A, Friedman A, Kaufer D. Blood-brain barrier dysfunction in aging induces hyperactivation of TGFβ signaling and chronic yet reversible neural dysfunction. Sci Transl Med 2019; 11:eaaw8283. [PMID: 31801886 DOI: 10.1126/scitranslmed.aaw8283] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/15/2019] [Accepted: 11/07/2019] [Indexed: 12/16/2022]
Abstract
Aging involves a decline in neural function that contributes to cognitive impairment and disease. However, the mechanisms underlying the transition from a young-and-healthy to aged-and-dysfunctional brain are not well understood. Here, we report breakdown of the vascular blood-brain barrier (BBB) in aging humans and rodents, which begins as early as middle age and progresses to the end of the life span. Gain-of-function and loss-of-function manipulations show that this BBB dysfunction triggers hyperactivation of transforming growth factor-β (TGFβ) signaling in astrocytes, which is necessary and sufficient to cause neural dysfunction and age-related pathology in rodents. Specifically, infusion of the serum protein albumin into the young rodent brain (mimicking BBB leakiness) induced astrocytic TGFβ signaling and an aged brain phenotype including aberrant electrocorticographic activity, vulnerability to seizures, and cognitive impairment. Furthermore, conditional genetic knockdown of astrocytic TGFβ receptors or pharmacological inhibition of TGFβ signaling reversed these symptomatic outcomes in aged mice. Last, we found that this same signaling pathway is activated in aging human subjects with BBB dysfunction. Our study identifies dysfunction in the neurovascular unit as one of the earliest triggers of neurological aging and demonstrates that the aging brain may retain considerable latent capacity, which can be revitalized by therapeutic inhibition of TGFβ signaling.
Collapse
Affiliation(s)
- Vladimir V Senatorov
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Aaron R Friedman
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Dan Z Milikovsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jonathan Ofer
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Rotem Saar-Ashkenazy
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Adiel Charbash
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Naznin Jahan
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gregory Chin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Eszter Mihaly
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jessica M Lin
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Harrison J Ramsay
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ariana Moghbel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marcela K Preininger
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chelsy R Eddings
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Helen V Harrison
- School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rishi Patel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yizhuo Shen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hana Ghanim
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Huanjie Sheng
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ronel Veksler
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Peter H Sudmant
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Albert Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Barry Hart
- Innovation Pathways, Palo Alto, CA 94301, USA
| | - Michael A Rogawski
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Andrew Dillin
- Glenn Center for Aging Research, Howard Hughes Medical Institute, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- Department of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute and Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA.
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
47
|
Swissa E, Serlin Y, Vazana U, Prager O, Friedman A. Blood-brain barrier dysfunction in status epileptics: Mechanisms and role in epileptogenesis. Epilepsy Behav 2019; 101:106285. [PMID: 31711869 DOI: 10.1016/j.yebeh.2019.04.038] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB), a unique anatomical and physiological interface between the central nervous system (CNS) and the peripheral circulation, is essential for the function of neural circuits. Interactions between the BBB, cerebral blood vessels, neurons, astrocytes, microglia, and pericytes form a dynamic functional unit known as the neurovascular unit (NVU). The NVU-BBB crosstalk plays a key role in the regulation of blood flow, response to injury, neuronal firing, and synaptic plasticity. Blood-brain barrier dysfunction (BBBD), a hallmark of brain injury, is a prominent finding in status epilepticus. Blood-brain barrier dysfunction is observed within the first hour of status epilepticus, and in epileptogenic brain regions, may last for months. Blood-brain barrier dysfunction was shown to have a role in astroglial dysfunction, neuroinflammation, increasing neural excitability, reduction of seizure threshold, excitatory synaptogenesis, impaired plasticity, and epileptogenesis. A key signaling pathway associated with BBBD-induced neurovascular dysfunction is the transforming growth factor beta (TGF-β) proinflammatory pathway, activated by the extravasation of serum albumin into the brain when BBB functions are compromised. Specific small molecules blocking TGF-β, and the nonspecific, Food and Drug Administration (FDA) approved blocker and angiotensin antagonist losartan, were shown to reduce BBBD and block epileptogenesis. With these encouraging preclinical data, we have developed imaging approach to quantitatively assess BBBD as a diagnostic, predictive, and pharmacodynamic biomarker after brain injury. Clinical trials in the foreseen future are expected to test the feasibility of BBB-targeted diagnostic coupled therapy in status epileptics and seizure disorders. This article is part of the Special Issue "Proceedings of the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures".
Collapse
Affiliation(s)
- Evyatar Swissa
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Inter-Faculty Brain Science School, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva Israel
| | - Yonatan Serlin
- Neurology Residency Training Program, McGill University, Montreal, QC, Canada
| | - Udi Vazana
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Inter-Faculty Brain Science School, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva Israel
| | - Ofer Prager
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Inter-Faculty Brain Science School, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva Israel
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Inter-Faculty Brain Science School, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva Israel; Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
48
|
Alosco ML, Stein TD, Tripodis Y, Chua AS, Kowall NW, Huber BR, Goldstein LE, Cantu RC, Katz DI, Palmisano JN, Martin B, Cherry JD, Mahar I, Killiany RJ, McClean MD, Au R, Alvarez V, Stern RA, Mez J, McKee AC. Association of White Matter Rarefaction, Arteriolosclerosis, and Tau With Dementia in Chronic Traumatic Encephalopathy. JAMA Neurol 2019; 76:1298-1308. [PMID: 31380975 PMCID: PMC6686769 DOI: 10.1001/jamaneurol.2019.2244] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/28/2019] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease associated with repetitive head impacts, including those from US football, that presents with cognitive and neuropsychiatric disturbances that can progress to dementia. Pathways to dementia in CTE are unclear and likely involve tau and nontau pathologic conditions. OBJECTIVE To investigate the association of white matter rarefaction and cerebrovascular disease with dementia in deceased men older than 40 years who played football and had CTE. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional study involves analyses of data from the ongoing Understanding Neurologic Injury and Traumatic Encephalopathy (UNITE) Study, which is conducted via and included brain donors from the Veterans Affairs-Boston University-Concussion Legacy Foundation brain bank between 2008 and 2017. An original sample of 224 men who had played football and were neuropathologically diagnosed with CTE was reduced after exclusion of those younger than 40 years and those missing data. EXPOSURES The number of years of football play as a proxy for repetitive head impacts. MAIN OUTCOMES AND MEASURES Neuropathological assessment of white matter rarefaction and arteriolosclerosis severity (on a scale of 0-3, where 3 is severe); number of infarcts, microinfarcts, and microbleeds; and phosphorylated tau accumulation determined by CTE stage and semiquantitative rating of dorsolateral frontal cortex (DLFC) neurofibrillary tangles (NFTs) (none or mild vs moderate or severe). Informant-based retrospective clinical interviews determined dementia diagnoses via diagnostic consensus conferences. RESULTS A total of 180 men were included. The mean (SD) age of the sample at death was 67.9 (12.7) years. Of 180, 120 [66.7%]) were found to have had dementia prior to death. Moderate to severe white matter rarefaction (84 of 180 [46.6%]) and arteriolosclerosis (85 of 180 [47.2%]) were common; infarcts, microinfarcts, and microbleeds were not. A simultaneous equations regression model controlling for age and race showed that more years of play was associated with more severe white matter rarefaction (β, 0.16 [95% CI, 0.02-0.29]; P = .03) and greater phosphorylated tau accumulation (DLFC NFTs: β, 0.15 [95% CI, 0.004-0.30]; P = .04; CTE stage: β, 0.27 [95% CI, 0.14-0.41]; P < .001). White matter rarefaction (β, 0.16 [95% CI, 0.02-0.29]; P = .03) and DLFC NFTs (β, 0.16 [95% CI, 0.03-0.28]; P = .01) were associated with dementia. Arteriolosclerosis and years of play were not associated, but arteriolosclerosis was independently associated with dementia (β, 0.21 [95% CI, 0.07-0.35]; P = .003). CONCLUSIONS AND RELEVANCE Among older men who had played football and had CTE, more years of football play were associated with more severe white matter rarefaction and greater DLFC NFT burden. White matter rarefaction, arteriolosclerosis, and DLFC NFTs were independently associated with dementia. Dementia in CTE is likely a result of neuropathologic changes, including white matter rarefaction and phosphorylated tau, associated with repetitive head impact and pathologic changes not associated with head trauma, such as arteriolosclerosis.
Collapse
Affiliation(s)
- Michael L. Alosco
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Thor D. Stein
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
- VA Boston Healthcare System, Boston, Massachusetts
- Bedford Veterans Affairs Medical Center, Bedford, Massachusetts
| | - Yorghos Tripodis
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Alicia S. Chua
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Neil W. Kowall
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
- VA Boston Healthcare System, Boston, Massachusetts
| | - Bertrand Russell Huber
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- VA Boston Healthcare System, Boston, Massachusetts
- National Center for Posttraumatic Stress Disorder, VA Boston Healthcare, Boston, Massachusetts
| | - Lee E. Goldstein
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Psychiatry, Boston University School of Medicine, Boston, Massachusetts
- Department of Electrical & Computer Engineering, Boston University College of Engineering, Boston, Massachusetts
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, Massachusetts
| | - Robert C. Cantu
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts
- Concussion Legacy Foundation, Boston, Massachusetts
- Department of Neurosurgery, Emerson Hospital, Concord, Massachusetts
| | - Douglas I. Katz
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Braintree Rehabilitation Hospital, Braintree, Massachusetts
| | - Joseph N. Palmisano
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, Massachusetts
| | - Brett Martin
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, Massachusetts
| | - Jonathan D. Cherry
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
- VA Boston Healthcare System, Boston, Massachusetts
| | - Ian Mahar
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Ronald J. Killiany
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts
- Center for Biomedical Imaging, Boston University School of Medicine, Boston, Massachusetts
| | - Michael D. McClean
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts
| | - Rhoda Au
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Boston, Massachusetts
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| | - Victor Alvarez
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- VA Boston Healthcare System, Boston, Massachusetts
| | - Robert A. Stern
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts
| | - Jesse Mez
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Ann C. McKee
- Boston University Alzheimer’s Disease Center and CTE Center, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
- VA Boston Healthcare System, Boston, Massachusetts
- Bedford Veterans Affairs Medical Center, Bedford, Massachusetts
| |
Collapse
|
49
|
Kamintsky L, Cairns KA, Veksler R, Bowen C, Beyea SD, Friedman A, Calkin C. Blood-brain barrier imaging as a potential biomarker for bipolar disorder progression. Neuroimage Clin 2019; 26:102049. [PMID: 31718955 PMCID: PMC7229352 DOI: 10.1016/j.nicl.2019.102049] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/05/2019] [Accepted: 10/21/2019] [Indexed: 11/29/2022]
Abstract
Bipolar disorder affects approximately 2% of the population and is typically characterized by recurrent episodes of mania and depression. While some patients achieve remission using mood-stabilizing treatments, a significant proportion of patients show progressive changes in symptomatology over time. Bipolar progression is diverse in nature and may include a treatment-resistant increase in the frequency and severity of episodes, worse psychiatric and functional outcomes, and a greater risk of suicide. The mechanisms underlying bipolar disorder progression remain poorly understood and there are currently no biomarkers for identifying patients at risk. The objective of this study was to explore the potential of blood-brain barrier (BBB) imaging as such a biomarker, by acquiring the first imaging data of BBB leakage in bipolar patients, and evaluating the potential association between BBB dysfunction and bipolar symptoms. To this end, a cohort of 36 bipolar patients was recruited through the Mood Disorders Clinic (Nova Scotia Health Authority, Canada). All patients, along with 14 control subjects (matched for sex, age and metabolic status), underwent contrast-enhanced dynamic MRI scanning for quantitative assessment of BBB leakage as well as clinical and psychiatric evaluations. Outlier analysis has identified a group of 10 subjects with significantly higher percentages of brain volume with BBB leakage (labeled the "extensive BBB leakage" group). This group consisted exclusively of bipolar patients, while the "normal BBB leakage" group included the entire control cohort and the remaining 26 bipolar subjects. Among the bipolar cohort, patients with extensive BBB leakage were found to have more severe depression and anxiety, and a more chronic course of illness. Furthermore, all bipolar patients within this group were also found to have co-morbid insulin resistance, suggesting that insulin resistance may increase the risk of BBB dysfunction in bipolar patients. Our findings demonstrate a clear link between BBB leakage and greater psychiatric morbidity in bipolar patients and highlight the potential of BBB imaging as a mechanism-based biomarker for bipolar disorder progression.
Collapse
Affiliation(s)
- Lyna Kamintsky
- Department of Medical Neuroscience, Dalhousie University, Sir Charles Tupper Building, 5850 College Street, Halifax, NS, B3H 4R2, Canada
| | - Kathleen A Cairns
- Nova Scotia Health Authority, Mood Disorders Clinic, 5909 Veterans Memorial Lane, Halifax, NS, B3H 2E2, Canada
| | - Ronel Veksler
- Department of Physiology and Cell Biology, Medicine, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Chris Bowen
- Biomedical Translational Imaging Centre (BIOTIC), QEII Health Sciences Centre, and Department of Diagnostic Radiology, Dalhousie University, 1796 Summer Street, Halifax, NS, B3H 3A7, Canada
| | - Steven D Beyea
- Biomedical Translational Imaging Centre (BIOTIC), QEII Health Sciences Centre, and Department of Diagnostic Radiology, Dalhousie University, 1796 Summer Street, Halifax, NS, B3H 3A7, Canada
| | - Alon Friedman
- Department of Medical Neuroscience, Dalhousie University, Sir Charles Tupper Building, 5850 College Street, Halifax, NS, B3H 4R2, Canada; Department of Physiology and Cell Biology, Medicine, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Cynthia Calkin
- Departments of Psychiatry and Medical Neuroscience, Dalhousie University, Mood Disorders Clinic, 5909 Veterans Memorial Lane, Halifax, NS, B3H 2E2, Canada
| |
Collapse
|
50
|
Standring OJ, Friedberg J, Tripodis Y, Chua AS, Cherry JD, Alvarez VE, Huber BR, Xia W, Mez J, Alosco ML, Nicks R, Mahar I, Pothast MJ, Gardner HM, Meng G, Palmisano JN, Martin BM, Dwyer B, Kowall NW, Cantu RC, Goldstein LE, Katz DI, Stern RA, McKee AC, Stein TD. Contact sport participation and chronic traumatic encephalopathy are associated with altered severity and distribution of cerebral amyloid angiopathy. Acta Neuropathol 2019; 138:401-413. [PMID: 31183671 DOI: 10.1007/s00401-019-02031-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/24/2019] [Accepted: 05/26/2019] [Indexed: 12/14/2022]
Abstract
Cerebral amyloid angiopathy (CAA) consists of beta-amyloid deposition in the walls of the cerebrovasculature and is commonly associated with Alzheimer's disease (AD). However, the association of CAA with repetitive head impacts (RHI) and with chronic traumatic encephalopathy (CTE) is unknown. We evaluated the relationship between RHI from contact sport participation, CTE, and CAA within a group of deceased contact sport athletes (n = 357), a community-based cohort (n = 209), and an AD cohort from Boston University AD Center (n = 241). Unsupervised hierarchal cluster analysis demonstrated a unique cluster (n = 11) with increased CAA in the leptomeningeal vessels compared to the intracortical vessels (p < 0.001) comprised of participants with significantly greater frequencies of CTE (7/11) and history of RHI. Overall, participants with CTE (n = 251) had more prevalent (p < 0.001) and severe (p = 0.010) CAA within the frontal leptomeningeal vessels compared to intracortical vessels. Compared to those with AD, participants with CTE had more severe CAA in frontal than parietal lobes (p < 0.001) and more severe CAA in leptomeningeal than intracortical vessels (p = 0.002). The overall frequency of CAA in participants with CTE was low, and there was no significant association between contact sport participation and the presence of CAA. However, in those with CAA, a history of contact sports was associated with increased CAA severity in the frontal leptomeningeal vessels (OR = 4.01, 95% CI 2.52-6.38, p < 0.001) adjusting for AD, APOE ε4 status, and age. Participants with CAA had increased levels of sulcal tau pathology and decreased levels of the synaptic marker PSD-95 (p's < 0.05), and CAA was a predictor of dementia (OR = 1.75, 95% CI 1.02-2.99, p = 0.043) adjusting for age, sex, and comorbid pathology. Overall, contact sport participation and CTE were associated with more severe frontal and leptomeningeal CAA, and CAA was independently associated with worse pathological and clinical outcomes.
Collapse
Affiliation(s)
- Oliver J Standring
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
| | - Jacob Friedberg
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
| | - Yorghos Tripodis
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, 20118, USA
| | - Alicia S Chua
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, 20118, USA
| | - Jonathan D Cherry
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Victor E Alvarez
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- Department of Veterans Affairs Medical Centers, Bedford, MA, 01730, USA
| | - Bertrand R Huber
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Weiming Xia
- Department of Veterans Affairs Medical Centers, Bedford, MA, 01730, USA
| | - Jesse Mez
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Michael L Alosco
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Raymond Nicks
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Department of Veterans Affairs Medical Centers, Bedford, MA, 01730, USA
| | - Ian Mahar
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Morgan J Pothast
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Veterans Affairs Medical Centers, Bedford, MA, 01730, USA
| | - Hannah M Gardner
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
| | - Gaoyuan Meng
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Veterans Affairs Medical Centers, Bedford, MA, 01730, USA
| | - Joseph N Palmisano
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, USA
| | - Brett M Martin
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, USA
| | - Brigid Dwyer
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- Braintree Rehabilitation Hospital, Braintree, MA, 02118, USA
| | - Neil W Kowall
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Robert C Cantu
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, 20119, USA
- Concussion Legacy Foundation, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Neurosurgery, Emerson Hospital, Concord, MA, 01742, USA
| | - Lee E Goldstein
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Departments of Psychiatry, Ophthalmology, Boston University School of Medicine, Boston, USA
- Departments of Biomedical, Electrical and Computer Engineering, Boston University College of Engineering, Boston, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Douglas I Katz
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- Braintree Rehabilitation Hospital, Braintree, MA, 02118, USA
| | - Robert A Stern
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, 20119, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Ann C McKee
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- Department of Veterans Affairs Medical Centers, Bedford, MA, 01730, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Thor D Stein
- Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston University, Boston, MA, 02118, USA.
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA.
- Department of Veterans Affairs Medical Centers, Bedford, MA, 01730, USA.
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|