1
|
Sun M, Baker TL, Wilson CT, Brady RD, Yamakawa GR, Wright DK, Mychasiuk R, Vo A, Wilson T, Allen J, McDonald SJ, Shultz SR. Treatment with the vascular endothelial growth factor-A antibody, bevacizumab, has sex-specific effects in a rat model of mild traumatic brain injury. J Cereb Blood Flow Metab 2024; 44:542-555. [PMID: 37933736 PMCID: PMC10981407 DOI: 10.1177/0271678x231212377] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023]
Abstract
Mild traumatic brain injury (mTBI) involves damage to the cerebrovascular system. Vascular endothelial growth factor-A (VEGF-A) is an important modulator of vascular health and VEGF-A promotes the brain's ability to recover after more severe forms of brain injury; however, the role of VEGF-A in mTBI remains poorly understood. Bevacizumab (BEV) is a monoclonal antibody that binds to VEGF-A and neutralises its actions. To better understand the role of VEGF-A in mTBI recovery, this study examined how BEV treatment affected outcomes in rats given a mTBI. Adult Sprague-Dawley rats were assigned to sham-injury + vehicle treatment (VEH), sham-injury + BEV treatment, mTBI + VEH treatment, mTBI + BEV treatment groups. Treatment was administered intracerebroventricularly via a cannula beginning at the time of injury and continuing until the end of the study. Rats underwent behavioral testing after injury and were euthanized on day 11. In both females and males, BEV had a negative impact on cognitive function. mTBI and BEV treatment increased the expression of inflammatory markers in females. In males, BEV treatment altered markers related to hypoxia and vascular health. These novel findings of sex-specific responses to BEV and mTBI provide important insights into the role of VEGF-A in mTBI.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Tamara L Baker
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Campbell T Wilson
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anh Vo
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Trevor Wilson
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Josh Allen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Health Sciences, Vancouver Island University, Nanaimo, BC, Canada
| |
Collapse
|
2
|
Sun M, Baker TL, Wilson CT, Brady RD, Mychasiuk R, Yamakawa GR, Vo A, Wilson T, McDonald SJ, Shultz SR. Treatment with vascular endothelial growth factor-A worsens cognitive recovery in a rat model of mild traumatic brain injury. Front Mol Neurosci 2022; 15:937350. [PMID: 36385769 PMCID: PMC9643175 DOI: 10.3389/fnmol.2022.937350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/29/2022] [Indexed: 09/08/2023] Open
Abstract
Mild traumatic brain injury (mTBI) is a common and unmet clinical issue, with limited treatments available to improve recovery. The cerebrovascular system is vital to provide oxygen and nutrition to the brain, and a growing body of research indicates that cerebrovascular injury contributes to mTBI symptomatology. Vascular endothelial growth factor-A (VEGF-A) is a potent promoter of angiogenesis and an important modulator of vascular health. While indirect evidence suggests that increased bioavailability of VEGF-A may be beneficial after mTBI, the direct therapeutic effects of VEGF-A in this context remains unknown. This study therefore aimed to determine whether intracerebroventricular administration of recombinant VEGF-A could improve recovery from mTBI in a rat model. Male and female Sprague-Dawley rats were assigned to four groups: sham + vehicle (VEH), sham + VEGF-A, mTBI + VEH, mTBI + VEGF-A. The mTBI was induced using the lateral impact model, and treatment began at the time of the injury and continued until the end of the study. Rats underwent behavioral testing between days 1 and 10 post-injury, and were euthanized on day 11 for post-mortem analysis. In males, the mTBI + VEGF-A group had significantly worse cognitive recovery in the water maze than all other groups. In females, the VEGF treatment worsened cognitive performance in the water maze regardless of mTBI or sham injury. Analysis of hippocampal tissue found that these cognitive deficits occurred in the presence of gene expression changes related to neuroinflammation and hypoxia in both male and female rats. These findings indicate that the VEGF-A treatment paradigm tested in this study failed to improve mTBI outcomes in either male or female rats.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Tamara L. Baker
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Campbell T. Wilson
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rhys D. Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Glenn R. Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anh Vo
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Trevor Wilson
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
- Health and Human Services, Vancouver Island University, Nanaimo, BC, Canada
| |
Collapse
|
3
|
Siwicka-Gieroba D, Dabrowski W. Credibility of the Neutrophil-to-Lymphocyte Count Ratio in Severe Traumatic Brain Injury. Life (Basel) 2021; 11:life11121352. [PMID: 34947883 PMCID: PMC8706648 DOI: 10.3390/life11121352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality worldwide. The consequences of a TBI generate the activation and accumulation of inflammatory cells. The peak number of neutrophils entering into an injured brain is observed after 24 h; however, cells infiltrate within 5 min of closed brain injury. Neutrophils release toxic molecules including free radicals, proinflammatory cytokines, and proteases that advance secondary damage. Regulatory T cells impair T cell infiltration into the central nervous system and elevate reactive astrogliosis and interferon-γ gene expression, probably inducing the process of healing. Therefore, the neutrophil-to-lymphocyte ratio (NLR) may be a low-cost, objective, and available predictor of inflammation as well as a marker of secondary injury associated with neutrophil activation. Recent studies have documented that an NLR value on admission might be effective for predicting outcome and mortality in severe brain injury patients.
Collapse
|
4
|
Blythe EN, Weaver LC, Brown A, Dekaban GA. β2 Integrin CD11d/CD18: From Expression to an Emerging Role in Staged Leukocyte Migration. Front Immunol 2021; 12:775447. [PMID: 34858434 PMCID: PMC8630586 DOI: 10.3389/fimmu.2021.775447] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
CD11d/CD18 is the most recently discovered and least understood β2 integrin. Known CD11d adhesive mechanisms contribute to both extravasation and mesenchymal migration – two key aspects for localizing peripheral leukocytes to sites of inflammation. Differential expression of CD11d induces differences in monocyte/macrophage mesenchymal migration including impacts on macrophage sub-set migration. The participation of CD11d/CD18 in leukocyte localization during atherosclerosis and following neurotrauma has sparked interest in the development of CD11d-targeted therapeutic agents. Whereas the adhesive properties of CD11d have undergone investigation, the signalling pathways induced by ligand binding remain largely undefined. Underlining each adhesive and signalling function, CD11d is under unique transcriptional control and expressed on a sub-set of predominately tissue-differentiated innate leukocytes. The following review is the first to capture the nearly three decades of CD11d research and discusses the emerging role of CD11d in leukocyte migration and retention during the progression of a staged immune response.
Collapse
Affiliation(s)
- Eoin N Blythe
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Lynne C Weaver
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Arthur Brown
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Gregory A Dekaban
- Molecular Medicine Research Laboratories, Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
5
|
Major B, Symons GF, Sinclair B, O'Brien WT, Costello D, Wright DK, Clough M, Mutimer S, Sun M, Yamakawa GR, Brady RD, O'Sullivan MJ, Mychasiuk R, McDonald SJ, O'Brien TJ, Law M, Kolbe S, Shultz SR. White and Gray Matter Abnormalities in Australian Footballers With a History of Sports-Related Concussion: An MRI Study. Cereb Cortex 2021; 31:5331-5338. [PMID: 34148076 DOI: 10.1093/cercor/bhab161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Sports-related concussion (SRC) is a form of mild traumatic brain injury that has been linked to long-term neurological abnormalities. Australian rules football is a collision sport with wide national participation and is growing in popularity worldwide. However, the chronic neurological consequences of SRC in Australian footballers remain poorly understood. This study investigated the presence of brain abnormalities in Australian footballers with a history of sports-related concussion (HoC) using multimodal MRI. Male Australian footballers with HoC (n = 26), as well as noncollision sport athletes with no HoC (n = 27), were recruited to the study. None of the footballers had sustained a concussion in the preceding 6 months, and all players were asymptomatic. Data were acquired using a 3T MRI scanner. White matter integrity was assessed using diffusion tensor imaging. Cortical thickness, subcortical volumes, and cavum septum pellucidum (CSP) were analyzed using structural MRI. Australian footballers had evidence of widespread microstructural white matter damage and cortical thinning. No significant differences were found regarding subcortical volumes or CSP. These novel findings provide evidence of persisting white and gray matter abnormalities in Australian footballers with HoC, and raise concerns related to the long-term neurological health of these athletes.
Collapse
Affiliation(s)
- Brendan Major
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Georgia F Symons
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Ben Sinclair
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - William T O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Daniel Costello
- Department of Medicine, The University of Melbourne, Parkville, VIC 3050, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Meaghan Clough
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Steven Mutimer
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC 3050, Australia
| | - Michael J O'Sullivan
- Department of Faculty of Medicine, UQ Centre for Clinical Research and Institute of Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC 3050, Australia
| | - Meng Law
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia.,Department of Radiology, Alfred Health, Melbourne, VIC 3004, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Melbourne, VIC 3800, Australia
| | - Scott Kolbe
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC 3050, Australia
| |
Collapse
|
6
|
Baker TL, Agoston DV, Brady RD, Major B, McDonald SJ, Mychasiuk R, Wright DK, Yamakawa GR, Sun M, Shultz SR. Targeting the Cerebrovascular System: Next-Generation Biomarkers and Treatment for Mild Traumatic Brain Injury. Neuroscientist 2021; 28:594-612. [PMID: 33966527 DOI: 10.1177/10738584211012264] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The diagnosis, prognosis, and treatment of mild traumatic brain injuries (mTBIs), such as concussions, are significant unmet medical issues. The kinetic forces that occur in mTBI adversely affect the cerebral vasculature, making cerebrovascular injury (CVI) a pathophysiological hallmark of mTBI. Given the importance of a healthy cerebrovascular system in overall brain function, CVI is likely to contribute to neurological dysfunction after mTBI. As such, CVI and related pathomechanisms may provide objective biomarkers and therapeutic targets to improve the clinical management and outcomes of mTBI. Despite this potential, until recently, few studies have focused on the cerebral vasculature in this context. This article will begin by providing a brief overview of the cerebrovascular system followed by a review of the literature regarding how mTBI can affect the integrity and function of the cerebrovascular system, and how this may ultimately contribute to neurological dysfunction and neurodegenerative conditions. We then discuss promising avenues of research related to mTBI biomarkers and interventions that target CVI, and conclude that a clinical approach that takes CVI into account could result in substantial improvements in the care and outcomes of patients with mTBI.
Collapse
Affiliation(s)
- Tamara L Baker
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Brendan Major
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
7
|
Xu X, Cowan M, Beraldo F, Schranz A, McCunn P, Geremia N, Brown Z, Patel M, Nygard KL, Khazaee R, Lu L, Liu X, Strong MJ, Dekaban GA, Menon R, Bartha R, Daley M, Mao H, Prado V, Prado MAM, Saksida L, Bussey T, Brown A. Repetitive mild traumatic brain injury in mice triggers a slowly developing cascade of long-term and persistent behavioral deficits and pathological changes. Acta Neuropathol Commun 2021; 9:60. [PMID: 33823944 PMCID: PMC8025516 DOI: 10.1186/s40478-021-01161-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
We have previously reported long-term changes in the brains of non-concussed varsity rugby players using magnetic resonance spectroscopy (MRS), diffusion tensor imaging (DTI) and functional magnetic imaging (fMRI). Others have reported cognitive deficits in contact sport athletes that have not met the diagnostic criteria for concussion. These results suggest that repetitive mild traumatic brain injuries (rmTBIs) that are not severe enough to meet the diagnostic threshold for concussion, produce long-term consequences. We sought to characterize the neuroimaging, cognitive, pathological and metabolomic changes in a mouse model of rmTBI. Using a closed-skull model of mTBI that when scaled to human leads to rotational and linear accelerations far below what has been reported for sports concussion athletes, we found that 5 daily mTBIs triggered two temporally distinct types of pathological changes. First, during the first days and weeks after injury, the rmTBI produced diffuse axonal injury, a transient inflammatory response and changes in diffusion tensor imaging (DTI) that resolved with time. Second, the rmTBI led to pathological changes that were evident months after the injury including: changes in magnetic resonance spectroscopy (MRS), altered levels of synaptic proteins, behavioural deficits in attention and spatial memory, accumulations of pathologically phosphorylated tau, altered blood metabolomic profiles and white matter ultrastructural abnormalities. These results indicate that exceedingly mild rmTBI, in mice, triggers processes with pathological consequences observable months after the initial injury.
Collapse
|
8
|
McCarty MF, Lerner A. The second phase of brain trauma can be controlled by nutraceuticals that suppress DAMP-mediated microglial activation. Expert Rev Neurother 2021; 21:559-570. [PMID: 33749495 DOI: 10.1080/14737175.2021.1907182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION A delayed second wave of brain trauma is mediated in large part by microglia that are activated to a pro-inflammatory M1 phenotype by DAMP proteins released by dying neurons. These microglia can promote apoptosis or necrosis in neighboring neurons by producing a range of pro-inflammatory cytokines and the deadly oxidant peroxynitrite. This second wave could therefore be mitigated with agents that blunt the post-traumatic M1 activation of microglia and that preferentially promote a pro-healing M2 phenotype. AREAS COVERED The literature on nutraceuticals that might have clinical potential in this regard. EXPERT OPINION The chief signaling pathway whereby DAMPs promote M1 microglial activation involves activation of toll-like receptor 4 (TLR4), NADPH oxidase, NF-kappaB, and the stress activated kinases JNK and p38. The green tea catechin EGCG can suppress TLR4 expression. Phycocyanobilin can inhibit NOX2-dependent NADPH oxidase, ferulate and melatonin can oppose pro-inflammatory signal modulation by NADPH oxidase-derived oxidants. Long-chain omega-3 fatty acids, the soy isoflavone genistein, the AMPK activator berberine, glucosamine, and ketone bodies can down-regulate NF-kappaB activation. Vitamin D activity can oppose JNK/p38 activation. A sophisticated program of nutraceutical supplementation may have important potential for mitigating the second phase of neuronal death and aiding subsequent healing.
Collapse
Affiliation(s)
- Mark F McCarty
- Department of research, Catalytic Longevity Foundation, San Diego, California, USA
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| |
Collapse
|
9
|
O'Brien WT, Symons GF, Bain J, Major BP, Costello DM, Sun M, Kimpton JS, Chen Z, Brady RD, Mychasiuk R, O'Brien TJ, Monif M, Shultz SR, McDonald SJ. Elevated Serum Interleukin-1β Levels in Male, but not Female, Collision Sport Athletes with a Concussion History. J Neurotrauma 2021; 38:1350-1357. [PMID: 33308001 DOI: 10.1089/neu.2020.7479] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It is increasingly reported that a history of concussion may be associated with chronic deleterious consequences. While the pathophysiology that contributes to these consequences is not well understood, neuroinflammation is postulated to be critical. Activation of multi-protein complexes termed inflammasomes, a key component of this inflammatory response, has been reported in more severe TBIs; however, it has not been investigated in milder TBIs, such as concussion. This study investigated serum levels of interleukin (IL)-1β and IL-18 (key proteins activated downstream of these inflammasomes) at acute, sub-acute, and chronic time-points post-concussion. We recruited 105 Australian footballers (65 male, 40 female) during the pre-season, then prospectively followed these players for the occurrence of concussion during the season. At baseline, 58 footballers reported a previous concussion history, and 47 reported no previous concussion history. Additionally, 25 players sustained a mid-season concussion and were sampled at 2, 6, and 13 days post-concussion. Serum levels of IL-1β and IL-18 were quantified using highly sensitive Simoa HD-X Analyzer assays. At baseline, IL-1β levels were higher in male, but not female, footballers with a previous concussion history compared with footballers with no concussion history. There was also a positive correlation between years of collision sport participation and IL-18 levels in males. No evidence was found in males or females to indicate that IL-1β or IL-18 levels differed at 2, 6, or 13 days post-concussion. These findings provide novel insights into potential sex-specific physiological consequences of concussion, and suggest that neuroinflammation may be persistent chronically following concussion in male athletes.
Collapse
Affiliation(s)
- William T O'Brien
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Georgia F Symons
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Jesse Bain
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Brendan P Major
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Daniel M Costello
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Joshua S Kimpton
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Zhibin Chen
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Epidemiology, Monash University, Melbourne, Victoria, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Melbourne Health, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Mastura Monif
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Melbourne Health, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.,Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Huntemer-Silveira A, Patil N, Brickner MA, Parr AM. Strategies for Oligodendrocyte and Myelin Repair in Traumatic CNS Injury. Front Cell Neurosci 2021; 14:619707. [PMID: 33505250 PMCID: PMC7829188 DOI: 10.3389/fncel.2020.619707] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022] Open
Abstract
A major consequence of traumatic brain and spinal cord injury is the loss of the myelin sheath, a cholesterol-rich layer of insulation that wraps around axons of the nervous system. In the central nervous system (CNS), myelin is produced and maintained by oligodendrocytes. Damage to the CNS may result in oligodendrocyte cell death and subsequent loss of myelin, which can have serious consequences for functional recovery. Demyelination impairs neuronal function by decelerating signal transmission along the axon and has been implicated in many neurodegenerative diseases. After a traumatic injury, mechanisms of endogenous remyelination in the CNS are limited and often fail, for reasons that remain poorly understood. One area of research focuses on enhancing this endogenous response. Existing techniques include the use of small molecules, RNA interference (RNAi), and monoclonal antibodies that target specific signaling components of myelination for recovery. Cell-based replacement strategies geared towards replenishing oligodendrocytes and their progenitors have been utilized by several groups in the last decade as well. In this review article, we discuss the effects of traumatic injury on oligodendrocytes in the CNS, the lack of endogenous remyelination, translational studies in rodent models promoting remyelination, and finally human clinical studies on remyelination in the CNS after injury.
Collapse
Affiliation(s)
| | - Nandadevi Patil
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Megan A. Brickner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Ann M. Parr
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
11
|
Alexiou GA, Lianos GD, Tzima A, Sotiropoulos A, Nasios A, Metaxas D, Zigouris A, RN JZ, Mitsis M, Voulgaris S. Neutrophil to lymphocyte ratio as a predictive biomarker for computed tomography scan use in mild traumatic brain injury. Biomark Med 2020; 14:1085-1090. [DOI: 10.2217/bmm-2020-0150] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aim: Traumatic brain injury (TBI) is a serious health concern. We set out to investigate the role of neutrophil-to-lymphocytes ratio (NLR) at admission for predicting the need for computed tomography (CT) in mild-TBI. Materials & methods: A retrospective study of adult patients who presented with mild-TBI Results: One hundred and thirty patients met the inclusion criteria. Seventy-four patients had positive CT-findings. The mean NLR-levels at presentations were 5.6 ± 4.8. Patients with positive CT-findings had significant higher NLR-levels. Receiver operating characteristic curve analysis was conducted and the threshold of NLR-levels for detecting the cases with positive CT-findings was 2.5, with 78.1% sensitivity and 63% specificity Conclusion: To the best of our knowledge no previous study has assessed the value of NLR-levels for predicting the need for CT in mild-TBI.
Collapse
Affiliation(s)
- George A Alexiou
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | - Georgios D Lianos
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | - Aggeliki Tzima
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | | | - Anastasios Nasios
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | - Dimitris Metaxas
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | - Andreas Zigouris
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | - Jolanda Zika RN
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | - Michail Mitsis
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | - Spyridon Voulgaris
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| |
Collapse
|
12
|
Baker TL, Sun M, Semple BD, Tyebji S, Tonkin CJ, Mychasiuk R, Shultz SR. Catastrophic consequences: can the feline parasite Toxoplasma gondii prompt the purrfect neuroinflammatory storm following traumatic brain injury? J Neuroinflammation 2020; 17:222. [PMID: 32711529 PMCID: PMC7382044 DOI: 10.1186/s12974-020-01885-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/02/2020] [Indexed: 12/02/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality worldwide; however, treatment development is hindered by the heterogenous nature of TBI presentation and pathophysiology. In particular, the degree of neuroinflammation after TBI varies between individuals and may be modified by other factors such as infection. Toxoplasma gondii, a parasite that infects approximately one-third of the world’s population, has a tropism for brain tissue and can persist as a life-long infection. Importantly, there is notable overlap in the pathophysiology between TBI and T. gondii infection, including neuroinflammation. This paper will review current understandings of the clinical problems, pathophysiological mechanisms, and functional outcomes of TBI and T. gondii, before considering the potential synergy between the two conditions. In particular, the discussion will focus on neuroinflammatory processes such as microglial activation, inflammatory cytokines, and peripheral immune cell recruitment that occur during T. gondii infection and after TBI. We will present the notion that these overlapping pathologies in TBI individuals with a chronic T. gondii infection have the strong potential to exacerbate neuroinflammation and related brain damage, leading to amplified functional deficits. The impact of chronic T. gondii infection on TBI should therefore be investigated in both preclinical and clinical studies as the possible interplay could influence treatment strategies.
Collapse
Affiliation(s)
- Tamara L Baker
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Shiraz Tyebji
- Division of Infectious Diseases and Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Christopher J Tonkin
- Division of Infectious Diseases and Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia. .,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
13
|
Ah Kim H, Semple BD, Dill LK, Pham L, Dworkin S, Zhang SR, Lim R, Sobey CG, McDonald SJ. Systemic treatment with human amnion epithelial cells after experimental traumatic brain injury. Brain Behav Immun Health 2020; 5:100072. [PMID: 34589854 PMCID: PMC8474600 DOI: 10.1016/j.bbih.2020.100072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 10/26/2022] Open
Abstract
Systemic administration of human amnion epithelial cells (hAECs) was recently shown to reduce neuropathology and improve functional recovery following ischemic stroke in both mice and marmosets. Given the significant neuropathological overlap between ischemic stroke and traumatic brain injury (TBI), we hypothesized that a similar hAEC treatment regime would also improve TBI outcomes. Male mice (12 weeks old, n = 40) were given a sham injury or moderate severity TBI by controlled cortical impact. At 60 min post-injury, mice were given a single tail vein injection of either saline (vehicle) or 1 × 106 hAECs suspended in saline. At 24 h post-injury, mice were assessed for locomotion and anxiety using an open field, and sensorimotor ability using a rotarod. At 48 h post-injury, brains were collected for analysis of immune cells via flow cytometry, or histological evaluation of lesion volume and hAEC penetration. To assess the impact of TBI and hAECs on lymphoid organs, spleen and thymus weights were determined. Treatment with hAECs did not prevent TBI-induced sensorimotor deficits at 24 h post-injury. hAECs were detected in the injured brain parenchyma; however, lesion volume was not altered by hAEC treatment. Robust increases in several leukocyte populations in the ipsilateral hemisphere of TBI mice were found when compared to sham mice at 48 h post-injury; however, hAEC treatment did not alter brain immune cell numbers. Both TBI and hAEC treatment were found to increase spleen weight. Taken together, these findings indicate that-unlike in ischemic stroke-treatment with hAEC was unable to prevent immune cell infiltration and sensorimotor deficits in the acute stages following controlled cortical impact in mice. Although further investigations are required, our data suggests that the lack of hAEC-induced neuroprotection in the current study may be explained by the differential splenic contributions to neuropathology between these brain injury models.
Collapse
Affiliation(s)
- Hyun Ah Kim
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Larissa K Dill
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Alfred Health, Melbourne, VIC, Australia
| | - Louise Pham
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Shenpeng R Zhang
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.,Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia.,Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Zhang ZW, Liang J, Yan JX, Ye YC, Wang JJ, Chen C, Sun HT, Chen F, Tu Y, Li XH. TBHQ improved neurological recovery after traumatic brain injury by inhibiting the overactivation of astrocytes. Brain Res 2020; 1739:146818. [PMID: 32275911 DOI: 10.1016/j.brainres.2020.146818] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) is a major leading cause of death and long-term disability. Although astrocytes play a key role in neuroprotection after TBI in the early stage, the overactivation of astrocytes can lead to long-term functional deficits, and the underlying pathophysiological mechanisms remain unclear. In addition, it is unknown whether the nuclear factor erythroid 2-related factor2/haem oxygenase-1 (Nrf-2/HO-1) pathway could elicit a neuroprotective effect by decreasing astrocyte overactivation after TBI. We aimed to study the effects of tert-butylhydroquinone (TBHQ) in reducing astrocyte overactivation after TBI and explored the underlying mechanisms. We first established a controlled cortical impact (CCI) model in rats and performed Haematoxylin and eosin (H&E) staining to observe brain tissue damage. The cognitive function of rats was assessed by modified neurological severity scoring (mNSS) and Morris water maze (MWM) test. Astrocyte and microglia activation was detected by immunofluorescence staining. Oxidative stress conditions were investigated using Western blotting. An enzyme-linked immunosorbent assay (ELISA) was designed to assess the level of the proinflammatory factor tumour necrosis factor-alpha (TNF-α). Dihydroethidium (DHE) staining was used to detect reactive oxygen species (ROS). Apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The results showed that the administration of TBHQ ameliorated motor function and cognitive deficits and decreased the lesion volume. In addition, TBHQ significantly decreased astrocyte overactivation, diminished the pro-inflammatory phenotype M1 and inflammatory cytokines production after TBI, increased Nrf-2 nuclear accumulation, and enhanced the levels of the Nrf-2 downstream antioxidative genes HO-1 and NADPH-quinone oxidoreductase-1 (NQO-1). Furthermore, TBHQ treatment alleviated apoptosis and neuronal death in the cerebral cortex. Overall, our data indicated that the upregulation of Nrf-2 expression could enhance neuroprotection and decrease astrocyte overactivation and might represent a new theoretical basis for treating TBI.
Collapse
Affiliation(s)
- Zhen-Wen Zhang
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jun Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jing-Xing Yan
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Yi-Chao Ye
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Chong Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Feng Chen
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yue Tu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China.
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
15
|
Wang M, Luo L. An Effective NADPH Oxidase 2 Inhibitor Provides Neuroprotection and Improves Functional Outcomes in Animal Model of Traumatic Brain Injury. Neurochem Res 2020; 45:1097-1106. [PMID: 32072445 DOI: 10.1007/s11064-020-02987-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/23/2022]
Abstract
Traumatic brain injury (TBI) has become a leading cause of death and disability all over the world. Pharmacological suppression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) can inhibit oxidative stress which is implicated in the pathology of TBI. GSK2795039 was reported to target NOX2 to inhibit [Formula: see text] and ROS production. The present study aimed to investigate the effect of GSK2795039 on NOX2 activity and neurological deficits in a TBI mouse model. TBI mouse model was established by a weight-drop to mouse skull. GSK2795039 at a dose of 100 mg/kg was administrated to mice 30 min before TBI. NOX2 expression and activity were detected by Western blot and biochemical method. Neurological damage and apoptosis were detected by behavioral test and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. GSK2795039 significantly inhibited NOX2 expression and activity in the TBI mouse model. It also attenuated TBI-induced neurological deficits, apoptosis, and neurological recovery. The results indicate that GSK2795039 can be used as a potential drug for TBI treatment.
Collapse
Affiliation(s)
- Mengwei Wang
- Department of Emergency, The Fourth Affiliated Hospital of China Medical University, No. 4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China.
| | - Le Luo
- Shanghai Zhuole Biotechnology Center, No. 2066 Wangyuan Road, Shanghai, 201499, China
| |
Collapse
|
16
|
Xu C, Diao YF, Wang J, Liang J, Xu HH, Zhao ML, Zheng B, Luan Z, Wang JJ, Yang XP, Wei MG, Duan JH, Wang KQ, Chen C, Chen F, Ming D, Zhang S, Sun HT, Li XH. Intravenously Infusing the Secretome of Adipose-Derived Mesenchymal Stem Cells Ameliorates Neuroinflammation and Neurological Functioning After Traumatic Brain Injury. Stem Cells Dev 2020; 29:222-234. [DOI: 10.1089/scd.2019.0173] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Chao Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Yun-Feng Diao
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Jing Wang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
- Department of Neurosurgery, Shanxi Dayi Hospital, Taiyuan, China
| | - Jun Liang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Hai-Huan Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Ming-Liang Zhao
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Zuo Luan
- Department of Pediatrics, Pediatric Surgery of Navy General Hospital, Beijing, China
| | - Jing-Jing Wang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Xi-Ping Yang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Meng-Guang Wei
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Jing-Hao Duan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Ke-Qiang Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Feng Chen
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Sai Zhang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Hong-Tao Sun
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| |
Collapse
|
17
|
de Azevedo-Quintanilha IG, Vieira-de-Abreu A, Ferreira AC, Reis PA, Silva TI, Nascimento DDO, Campbell RA, Estato V, Weyrich AS, Bozza PT, Zimmerman GA, Castro-Faria-Neto HC. Integrin αDβ2 influences cerebral edema, leukocyte accumulation and neurologic outcomes in experimental severe malaria. PLoS One 2019; 14:e0224610. [PMID: 31869339 PMCID: PMC6927624 DOI: 10.1371/journal.pone.0224610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 10/17/2019] [Indexed: 12/30/2022] Open
Abstract
Malaria is an infectious disease of major worldwide clinical importance that causes a variety of severe, or complicated, syndromes including cerebral malaria, which is often fatal. Leukocyte integrins are essential for host defense but also mediate physiologic responses of the innate and adaptive immune systems. We previously showed that targeted deletion of the αD subunit (αD-/-) of the αDβ2 integrin, which is expressed on key leukocyte subsets in mice and humans, leads to absent expression of the integrin heterodimer on murine macrophages and reduces mortality in mice infected with Plasmodium berghei ANKA (P. berghei ANKA). To further identify mechanisms involved in the protective effect of αD deletion in this model of severe malaria we examined wild type C57BL/6 (WT) and αD-/- mice after P. berghei ANKA infection and found that vessel plugging and leukocyte infiltration were significantly decreased in the brains of αD-/- animals. Intravital microscopy demonstrated decreased rolling and adhesion of leukocytes in cerebral vessels of αD-/- mice. Flow cytometry analysis showed decreased T-lymphocyte accumulation in the brains of infected αD-/- animals. Evans blue dye exclusion assays demonstrated significantly less dye extravasation in the brains of αD-/- mice, indicating preserved blood-brain barrier integrity. WT mice that were salvaged from P. berghei ANKA infection by treatment with chloroquine had impaired aversive memory, which was not observed in αD-/- mice. We conclude that deletion of integrin αDβ2 alters the natural course of experimental severe malaria, demonstrating previously unrecognized activities of a key leukocyte integrin in immune-inflammatory responses that mediate cerebral involvement.
Collapse
Affiliation(s)
| | - Adriana Vieira-de-Abreu
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - André C. Ferreira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia A. Reis
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tathiany I. Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle de O. Nascimento
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robert A. Campbell
- Department of Internal Medicine and Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - Vanessa Estato
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrew S. Weyrich
- Department of Internal Medicine and Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - Patrícia T. Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guy A. Zimmerman
- Department of Internal Medicine and Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - Hugo C. Castro-Faria-Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Wright DK, Brady RD, Kamnaksh A, Trezise J, Sun M, McDonald SJ, Mychasiuk R, Kolbe SC, Law M, Johnston LA, O'Brien TJ, Agoston DV, Shultz SR. Repeated mild traumatic brain injuries induce persistent changes in plasma protein and magnetic resonance imaging biomarkers in the rat. Sci Rep 2019; 9:14626. [PMID: 31602002 PMCID: PMC6787341 DOI: 10.1038/s41598-019-51267-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/28/2019] [Indexed: 01/05/2023] Open
Abstract
A single mild traumatic brain injury (mTBI) typically causes only transient symptoms, but repeated mTBI (RmTBI) is associated with cumulative and chronic neurological abnormalities. Clinical management of mTBI is challenging due to the heterogeneous, subjective and transient nature of symptoms, and thus would be aided by objective biomarkers. Promising biomarkers including advanced magnetic resonance imaging (MRI) and plasma levels of select proteins were examined here in a rat model of RmTBI. Rats received either two mild fluid percussion or sham injuries administered five days apart. Rats underwent MRI and behavioral testing 1, 3, 5, 7, and 30 days after the second injury and blood samples were collected on days 1, 7, and 30. Structural and diffusion-weighted MRI revealed that RmTBI rats had abnormalities in the cortex and corpus callosum. Proteomic analysis of plasma found that RmTBI rats had abnormalities in markers indicating axonal and vascular injury, metabolic and mitochondrial dysfunction, and glial reactivity. These changes occurred in the presence of ongoing cognitive and sensorimotor deficits in the RmTBI rats. Our findings demonstrate that RmTBI can result in chronic neurological abnormalities, provide insight into potential contributing pathophysiological mechanisms, and supports the use of MRI and plasma protein measures as RmTBI biomarkers.
Collapse
Affiliation(s)
- David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Alaa Kamnaksh
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Jack Trezise
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Scott C Kolbe
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Leigh A Johnston
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia.,Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia. .,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
19
|
Sun M, Brady RD, Casillas-Espinosa PM, Wright DK, Semple BD, Kim HA, Mychasiuk R, Sobey CG, O'Brien TJ, Vinh A, McDonald SJ, Shultz SR. Aged rats have an altered immune response and worse outcomes after traumatic brain injury. Brain Behav Immun 2019; 80:536-550. [PMID: 31039431 DOI: 10.1016/j.bbi.2019.04.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/28/2019] [Accepted: 04/26/2019] [Indexed: 12/14/2022] Open
Abstract
Initial studies suggest that increased age is associated with worse outcomes after traumatic brain injury (TBI), though the pathophysiological mechanisms responsible for this remain unclear. Immunosenescence (i.e., dysregulation of the immune system due to aging) may play a significant role in influencing TBI outcomes. This study therefore examined neurological outcomes and immune response in young-adult (i.e., 10 weeks old) compared to middle-aged (i.e., 1 year old) rats following a TBI (i.e., fluid percussion) or sham-injury. Rats were euthanized at either 24 h or one-week post-injury to analyze immune cell populations in the brain and periphery via flow cytometry, as well as telomere length (i.e., a biomarker of neurological health). Behavioral testing, as well as volumetric and diffusion-weighted MRI, were also performed in the one-week recovery rats to assess for functional deficits and brain damage. Middle-aged rats had worse sensorimotor deficits and shorter telomeres after TBI compared to young rats. Both aging and TBI independently worsened cognitive function and cortical volume. These changes occurred in the presence of fewer total leukocytes, fewer infiltrating myeloid cells, and fewer microglia in the brains of middle-aged TBI rats compared to young rats. These findings indicate that middle-aged rats have worse sensorimotor deficits and shorter telomeres after TBI than young rats, and this may be related to an altered neuroimmune response. Although further studies are required, these findings have important implications for understanding the pathophysiology and optimal treatment strategies in TBI patients across the life span.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Hyun Ah Kim
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Psychology, University of Calgary, Calgary, AB, Canada
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Antony Vinh
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
20
|
Sun M, Brady RD, van der Poel C, Apted D, Semple BD, Church JE, O'Brien TJ, McDonald SJ, Shultz SR. A Concomitant Muscle Injury Does Not Worsen Traumatic Brain Injury Outcomes in Mice. Front Neurol 2018; 9:1089. [PMID: 30619048 PMCID: PMC6297867 DOI: 10.3389/fneur.2018.01089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 11/28/2018] [Indexed: 01/20/2023] Open
Abstract
Traumatic brain injury (TBI) often involves multitrauma in which concurrent extracranial injury occurs. We previously demonstrated that a long bone fracture exacerbates neuroinflammation and functional outcomes in mice given a TBI. Whether other forms of concomitant peripheral trauma that are common in the TBI setting, such as skeletal muscle injury, have similar effects is unknown. As such, here we developed a novel mouse multitrauma model by combining a closed-skull TBI with a cardiotoxin (CTX)-induced muscle injury to investigate whether muscle injury affects TBI outcomes. Adult male mice were assigned to four groups: sham-TBI + sham-muscle injury (SHAM); sham-TBI + CTX-muscle injury (CTX); TBI + sham-muscle injury (TBI); TBI + CTX-muscle injury (MULTI). Some mice were euthanized at 24 h post-injury to assess neuroinflammation and cerebral edema. The remaining mice underwent behavioral testing after a 30-day recovery period, and were euthanized at 35 days post-injury for post-mortem analysis. At 24 h post-injury, both TBI and MULTI mice had elevated edema, increased expression of GFAP (i.e., a marker for reactive astrocytes), and increased mRNA levels of inflammatory chemokines. There was also an effect of injury on cytokine levels at 35 days post-injury. However, the TBI and MULTI mice did not significantly differ on any of the measures assessed. These initial findings suggest that a concomitant muscle injury does not significantly affect preclinical TBI outcomes. Future studies should investigate the combination of different injury models, additional outcomes, and other post-injury time points.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Rhys D Brady
- Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Chris van der Poel
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Danielle Apted
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Bridgette D Semple
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia.,Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jarrod E Church
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia.,Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia.,Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
21
|
Madathil SK, Wilfred BS, Urankar SE, Yang W, Leung LY, Gilsdorf JS, Shear DA. Early Microglial Activation Following Closed-Head Concussive Injury Is Dominated by Pro-Inflammatory M-1 Type. Front Neurol 2018; 9:964. [PMID: 30498469 PMCID: PMC6249371 DOI: 10.3389/fneur.2018.00964] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022] Open
Abstract
Microglial activation is a pathological hallmark of traumatic brain injury (TBI). Following brain injury, activated microglia/macrophages adopt different phenotypes, generally categorized as M-1, or classically activated, and M-2, or alternatively activated. While the M-1, or pro-inflammatory phenotype is detrimental to recovery, M-2, or the anti-inflammatory phenotype, aids in brain repair. Recent findings also suggest the existence of mixed phenotype following brain injury, where activated microglia simultaneously express both M-1 and M-2 markers. The present study sought to determine microglial activation states at early time points (6-72 h) following single or repeated concussive injury in rats. Closed-head concussive injury was modeled in rats using projectile concussive impact injury, with either single or repeated impacts (4 impacts, 1 h apart). Brain samples were examined using immunohistochemical staining, inflammatory gene profiling and real-time polymerase chain reaction analyses to detect concussive injury induced changes in microglial activation and phenotype in cortex and hippocampal regions. Our findings demonstrate robust microglial activation following concussive brain injury. Moreover, we show that multiple concussions induced a unique rod-shaped microglial morphology that was also observed in other diffuse brain injury models. Histological studies revealed a predominance of MHC-II positive M-1 phenotype in the post-concussive microglial milieu following multiple impacts. Although there was simultaneous expression of M-1 and M-2 markers, gene expression results indicate a clear dominance in M-1 pro-inflammatory markers following both single and repeated concussions. While the increase in M-1 markers quickly resolved after a single concussion, they persisted following repeated concussions, indicating a pro-inflammatory environment induced by multiple concussions that may delay recovery and contribute to long-lasting consequences of concussion.
Collapse
Affiliation(s)
- Sindhu K Madathil
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Bernard S Wilfred
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Sarah E Urankar
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Weihong Yang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Lai Yee Leung
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Janice S Gilsdorf
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Deborah A Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
22
|
Chan HH, Wathen CA, Mathews ND, Hogue O, Modic JP, Kundalia R, Wyant C, Park HJ, Najm IM, Trapp BD, Machado AG, Baker KB. Lateral cerebellar nucleus stimulation promotes motor recovery and suppresses neuroinflammation in a fluid percussion injury rodent model. Brain Stimul 2018; 11:1356-1367. [PMID: 30061053 DOI: 10.1016/j.brs.2018.07.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/17/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Many traumatic brain injury (TBI) survivors live with persistent disability from chronic motor deficits despite contemporary rehabilitation services, underscoring the need for novel treatment. OBJECTIVE/HYPOTHESIS We have previously shown that deep brain stimulation (DBS) of the lateral cerebellar nucleus (LCN) can enhance post-stroke motor recovery and increase the expression of markers of long-term potentiation in perilesional cerebral cortex. We hypothesize that a similar beneficial effect will be for motor deficits induced by unilateral fluid percussion injury (FPI) in rodents through long-term potentiation- and anti-inflammatory based mechanisms. METHODS Male Long Evans rats with a DBS macroelectrode in the LCN underwent FPI over contralateral primary motor cortex. After 4 weeks of spontaneous recovery, DBS treatment was applied for 4 weeks, with the pasta matrix, cylinder, and horizontal ladder tests used to evaluate motor performance. All animals were euthanized and tissue harvested for further analysis by histology, immunohistochemistry, RNA microarray assay and Western Blot. RESULTS LCN DBS-treated animals experienced a significantly greater rate of motor recovery than untreated surgical controls, with treated animals showing enhanced expression of RNA and protein for excitability related genes, suppressed expression of pro-inflammatory genes, suppressed microglial and astrocytic activation, but proliferation of c-fos positive cells. Finally, our data suggest a possible role for anti-apoptotic effects with LCN DBS. CONCLUSION LCN DBS enhanced the motor recovery following TBI, possibly by elevating the neuronal excitability at the perilesional area and mediating anti-apoptotic and anti-inflammatory effects.
Collapse
Affiliation(s)
- Hugh H Chan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Connor A Wathen
- Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH, USA
| | - Nicole D Mathews
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olivia Hogue
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - James P Modic
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ronak Kundalia
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Cara Wyant
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hyun-Joo Park
- Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH, USA
| | - Imad M Najm
- Epilepsy Center, Cleveland Clinic, Cleveland, OH, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andre G Machado
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH, USA
| | - Kenneth B Baker
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
23
|
Swanton T, Cook J, Beswick JA, Freeman S, Lawrence CB, Brough D. Is Targeting the Inflammasome a Way Forward for Neuroscience Drug Discovery? SLAS DISCOVERY 2018; 23:991-1017. [PMID: 29969573 DOI: 10.1177/2472555218786210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuroinflammation is becoming increasingly recognized as a critical factor in the pathology of both acute and chronic neurological conditions. Inflammasomes such as the one formed by NACHT, LRR, and PYD domains containing protein 3 (NLRP3) are key regulators of inflammation due to their ability to induce the processing and secretion of interleukin 1β (IL-1β). IL-1β has previously been identified as a potential therapeutic target in a variety of conditions due to its ability to promote neuronal damage under conditions of injury. Thus, inflammasome inhibition has the potential to curtail inflammatory signaling, which could prove beneficial in certain diseases. In this review, we discuss the evidence for inflammasome contributions to the pathology of neurodegenerative conditions such as Alzheimer's disease and Parkinson's disease, epilepsy, and acute degeneration following brain trauma or stroke. In addition, we review the current landscape of drug development targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Tessa Swanton
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James Cook
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James A Beswick
- 2 Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sally Freeman
- 2 Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Catherine B Lawrence
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - David Brough
- 1 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
24
|
Orr MB, Gensel JC. Spinal Cord Injury Scarring and Inflammation: Therapies Targeting Glial and Inflammatory Responses. Neurotherapeutics 2018; 15:541-553. [PMID: 29717413 PMCID: PMC6095779 DOI: 10.1007/s13311-018-0631-6] [Citation(s) in RCA: 350] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Deficits in neuronal function are a hallmark of spinal cord injury (SCI) and therapeutic efforts are often focused on central nervous system (CNS) axon regeneration. However, secondary injury responses by astrocytes, microglia, pericytes, endothelial cells, Schwann cells, fibroblasts, meningeal cells, and other glia not only potentiate SCI damage but also facilitate endogenous repair. Due to their profound impact on the progression of SCI, glial cells and modification of the glial scar are focuses of SCI therapeutic research. Within and around the glial scar, cells deposit extracellular matrix (ECM) proteins that affect axon growth such as chondroitin sulfate proteoglycans (CSPGs), laminin, collagen, and fibronectin. This dense deposition of material, i.e., the fibrotic scar, is another barrier to endogenous repair and is a target of SCI therapies. Infiltrating neutrophils and monocytes are recruited to the injury site through glial chemokine and cytokine release and subsequent upregulation of chemotactic cellular adhesion molecules and selectins on endothelial cells. These peripheral immune cells, along with endogenous microglia, drive a robust inflammatory response to injury with heterogeneous reparative and pathological properties and are targeted for therapeutic modification. Here, we review the role of glial and inflammatory cells after SCI and the therapeutic strategies that aim to replace, dampen, or alter their activity to modulate SCI scarring and inflammation and improve injury outcomes.
Collapse
Affiliation(s)
- Michael B Orr
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky College of Medicine, 741 S. Limestone, B463 BBSRB, Lexington, Kentucky, 40536, USA
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky College of Medicine, 741 S. Limestone, B463 BBSRB, Lexington, Kentucky, 40536, USA.
| |
Collapse
|
25
|
Abstract
Mild traumatic brain injury (mTBI) represents a significant public healthcare concern, accounting for the majority of all head injuries. While symptoms are generally transient, some patients go on to experience long-term cognitive impairments and additional mild impacts can result in exacerbated and persisting negative outcomes. To date, studies using a range of experimental models have reported chronic behavioral deficits in the presence of axonal injury and inflammation following repeated mTBI; assessments of oxidative stress and myelin pathology have thus far been limited. However, some models employed induced acute focal damage more suggestive of moderate–severe brain injury and are therefore not relevant to repeated mTBI. Given that the nature of mechanical loading in TBI is implicated in downstream pathophysiological changes, the mechanisms of damage and chronic consequences of single and repeated closed-head mTBI remain to be fully elucidated. This review covers literature on potential mechanisms of damage following repeated mTBI, integrating known mechanisms of pathology underlying moderate–severe TBIs, with recent studies on adult rodent models relevant to direct impact injuries rather than blast-induced damage. Pathology associated with excitotoxicity and cerebral blood flow-metabolism uncoupling, oxidative stress, cell death, blood-brain barrier dysfunction, astrocyte reactivity, microglial activation, diffuse axonal injury, and dysmyelination is discussed, followed by a summary of functional deficits and preclinical assessments of therapeutic strategies. Comprehensive characterization of the pathology underlying delayed and persisting deficits following repeated mTBI is likely to facilitate further development of therapeutic strategies to limit long-term sequelae.
Collapse
Affiliation(s)
- Brooke Fehily
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Melinda Fitzgerald
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia.,2 Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,3 Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
26
|
Sun M, McDonald SJ, Brady RD, O'Brien TJ, Shultz SR. The influence of immunological stressors on traumatic brain injury. Brain Behav Immun 2018; 69:618-628. [PMID: 29355823 DOI: 10.1016/j.bbi.2018.01.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/13/2018] [Accepted: 01/14/2018] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide, and typically involves a robust immune response. Although a great deal of preclinical research has been conducted to identify an effective treatment, all phase III clinical trials have been unsuccessful to date. These translational shortcomings are in part due to a failure to recognize and account for the heterogeneity of TBI, including how extracranial factors can influence the aftermath of TBI. For example, most preclinical studies have utilized isolated TBI models in young adult males, while clinical trials typically involve highly heterogeneous patient populations (e.g., different mechanisms of injury, a range of ages, presence of polytrauma or infection). This paper will review the current, albeit limited literature related to how TBI is affected by common concomitant immunological stressors. In particular, discussion will focus on whether extracranial trauma (i.e., polytrauma), infection, and age/immunosenescence can influence TBI pathophysiology, and thereby may result in a different brain injury than what would have occurred in an isolated TBI. It is concluded that these immunological stressors are all likely to be TBI modifiers that should be further studied and could impact translational treatment strategies.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rhys D Brady
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Departments of Neuroscience and Medicine, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
27
|
Wright DK, O'Brien TJ, Mychasiuk R, Shultz SR. Telomere length and advanced diffusion MRI as biomarkers for repetitive mild traumatic brain injury in adolescent rats. Neuroimage Clin 2018; 18:315-324. [PMID: 29876252 PMCID: PMC5987845 DOI: 10.1016/j.nicl.2018.01.033] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/24/2018] [Accepted: 01/26/2018] [Indexed: 12/13/2022]
Abstract
Mild traumatic brain injuries (mTBI) are of worldwide concern in adolescents of both sexes, and repeated mTBI (RmTBI) may have serious long-term neurological consequences. As such, the study of RmTBI and discovery of objective biomarkers that can help guide medical decisions is an important undertaking. Diffusion-weighted MRI (DWI), which provides markers of axonal injury, and telomere length (TL) are two clinically relevant biomarkers that have been implicated in a number of neurological conditions, and may also be affected by RmTBI. Therefore, this study utilized the lateral impact injury model of RmTBI to investigate changes in diffusion MRI and TL, and how these changes relate to each other. Adolescent male and female rats received either three mTBIs or three sham injuries. The first injury was given on postnatal day 30 (P30), with the repeated injuries separated by four days each. Seven days after the final injury, a sample of ear tissue was collected for TL analysis. Rats were then euthanized and whole brains were collected and fixated for MRI analyses that included diffusion and high-resolution structural sequences. Compared to the sham-injured group, RmTBI rats had significantly shorter TL at seven days post-injury. Analysis of advanced DWI measures found that RmTBI rats had abnormalities in the corpus callosum and cortex at seven days post-injury. Notably, many of the DWI changes were correlated with TL. These findings demonstrate that TL and DWI measurements are changed by RmTBI and may represent clinically applicable biomarkers for this.
Collapse
Affiliation(s)
- David K Wright
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC 3010, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3010, Australia
| | - Richelle Mychasiuk
- Alberta Children's Hospital Research Institute, University of Calgary, Department of Psychology, Calgary, AB, Canada
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
28
|
Madathil SK, Wilfred BS, Urankar SE, Yang W, Leung LY, Gilsdorf JS, Shear DA. Early Microglial Activation Following Closed-Head Concussive Injury Is Dominated by Pro-Inflammatory M-1 Type. Front Neurol 2018. [PMID: 30498469 DOI: 10.3389/fneur.2018.00964/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Microglial activation is a pathological hallmark of traumatic brain injury (TBI). Following brain injury, activated microglia/macrophages adopt different phenotypes, generally categorized as M-1, or classically activated, and M-2, or alternatively activated. While the M-1, or pro-inflammatory phenotype is detrimental to recovery, M-2, or the anti-inflammatory phenotype, aids in brain repair. Recent findings also suggest the existence of mixed phenotype following brain injury, where activated microglia simultaneously express both M-1 and M-2 markers. The present study sought to determine microglial activation states at early time points (6-72 h) following single or repeated concussive injury in rats. Closed-head concussive injury was modeled in rats using projectile concussive impact injury, with either single or repeated impacts (4 impacts, 1 h apart). Brain samples were examined using immunohistochemical staining, inflammatory gene profiling and real-time polymerase chain reaction analyses to detect concussive injury induced changes in microglial activation and phenotype in cortex and hippocampal regions. Our findings demonstrate robust microglial activation following concussive brain injury. Moreover, we show that multiple concussions induced a unique rod-shaped microglial morphology that was also observed in other diffuse brain injury models. Histological studies revealed a predominance of MHC-II positive M-1 phenotype in the post-concussive microglial milieu following multiple impacts. Although there was simultaneous expression of M-1 and M-2 markers, gene expression results indicate a clear dominance in M-1 pro-inflammatory markers following both single and repeated concussions. While the increase in M-1 markers quickly resolved after a single concussion, they persisted following repeated concussions, indicating a pro-inflammatory environment induced by multiple concussions that may delay recovery and contribute to long-lasting consequences of concussion.
Collapse
Affiliation(s)
- Sindhu K Madathil
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Bernard S Wilfred
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Sarah E Urankar
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Weihong Yang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Lai Yee Leung
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Janice S Gilsdorf
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Deborah A Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
29
|
Zawaski JA, Sabek OM, Voicu H, Eastwood Leung HC, Gaber MW. Effect of Brain Tumor Presence During Radiation on Tissue Toxicity: Transcriptomic and Metabolic Changes. Int J Radiat Oncol Biol Phys 2017; 99:983-993. [DOI: 10.1016/j.ijrobp.2017.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 07/02/2017] [Accepted: 07/06/2017] [Indexed: 10/19/2022]
|
30
|
Geremia NM, Hryciw T, Bao F, Streijger F, Okon E, Lee JHT, Weaver LC, Dekaban GA, Kwon BK, Brown A. The effectiveness of the anti-CD11d treatment is reduced in rat models of spinal cord injury that produce significant levels of intraspinal hemorrhage. Exp Neurol 2017; 295:125-134. [PMID: 28587875 DOI: 10.1016/j.expneurol.2017.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/11/2017] [Accepted: 06/01/2017] [Indexed: 11/30/2022]
Abstract
We have previously reported that administration of a CD11d monoclonal antibody (mAb) improves recovery in a clip-compression model of SCI. In this model the CD11d mAb reduces the infiltration of activated leukocytes into the injured spinal cord (as indicated by reduced intraspinal MPO). However not all anti-inflammatory strategies have reported beneficial results, suggesting that success of the CD11d mAb treatment may depend on the type or severity of the injury. We therefore tested the CD11d mAb treatment in a rat hemi-contusion model of cervical SCI. In contrast to its effects in the clip-compression model, the CD11d mAb treatment did not improve forelimb function nor did it significantly reduce MPO levels in the hemi-contused cord. To determine if the disparate results using the CD11d mAb were due to the biomechanical nature of the cord injury (compression SCI versus contusion SCI) or to the spinal level of the injury (12th thoracic level versus cervical) we further evaluated the CD11d mAb treatment after a T12 contusion SCI. In contrast to the T12 clip compression SCI, the CD11d mAb treatment did not improve locomotor recovery or significantly reduce MPO levels after T12 contusion SCI. Lesion analyses revealed increased levels of hemorrhage after contusion SCI compared to clip-compression SCI. SCI that is accompanied by increased intraspinal hemorrhage would be predicted to be refractory to the CD11d mAb therapy as this approach targets leukocyte diapedesis through the intact vasculature. These results suggest that the disparate results of the anti-CD11d treatment in contusion and clip-compression models of SCI are due to the different pathophysiological mechanisms that dominate these two types of spinal cord injuries.
Collapse
Affiliation(s)
- N M Geremia
- Molecular Medicine Research Group, Robarts Research Institute, The University of Western Ontario, 1151 Richmond Street North, London, Ontario N6A 5B7, Canada
| | - T Hryciw
- Molecular Medicine Research Group, Robarts Research Institute, The University of Western Ontario, 1151 Richmond Street North, London, Ontario N6A 5B7, Canada
| | - F Bao
- Molecular Medicine Research Group, Robarts Research Institute, The University of Western Ontario, 1151 Richmond Street North, London, Ontario N6A 5B7, Canada
| | - F Streijger
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, 818 West 10th Avenue, Vancouver, BC V5Z 1M9, Canada
| | - E Okon
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, 818 West 10th Avenue, Vancouver, BC V5Z 1M9, Canada
| | - J H T Lee
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, 818 West 10th Avenue, Vancouver, BC V5Z 1M9, Canada
| | - L C Weaver
- Molecular Medicine Research Group, Robarts Research Institute, The University of Western Ontario, 1151 Richmond Street North, London, Ontario N6A 5B7, Canada
| | - G A Dekaban
- Molecular Medicine Research Group, Robarts Research Institute, The University of Western Ontario, 1151 Richmond Street North, London, Ontario N6A 5B7, Canada
| | - B K Kwon
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, 818 West 10th Avenue, Vancouver, BC V5Z 1M9, Canada
| | - A Brown
- Molecular Medicine Research Group, Robarts Research Institute, The University of Western Ontario, 1151 Richmond Street North, London, Ontario N6A 5B7, Canada.
| |
Collapse
|
31
|
Wilson NM, Gurney ME, Dietrich WD, Atkins CM. Therapeutic benefits of phosphodiesterase 4B inhibition after traumatic brain injury. PLoS One 2017; 12:e0178013. [PMID: 28542295 PMCID: PMC5438188 DOI: 10.1371/journal.pone.0178013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/05/2017] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) initiates a deleterious inflammatory response that exacerbates pathology and worsens outcome. This inflammatory response is partially mediated by a reduction in cAMP and a concomitant upregulation of cAMP-hydrolyzing phosphodiesterases (PDEs) acutely after TBI. The PDE4B subfamily, specifically PDE4B2, has been found to regulate cAMP in inflammatory cells, such as neutrophils, macrophages and microglia. To determine if PDE4B regulates inflammation and subsequent pathology after TBI, adult male Sprague Dawley rats received sham surgery or moderate parasagittal fluid-percussion brain injury (2 ± 0.2 atm) and were then treated with a PDE4B - selective inhibitor, A33, or vehicle for up to 3 days post-surgery. Treatment with A33 reduced markers of microglial activation and neutrophil infiltration at 3 and 24 hrs after TBI, respectively. A33 treatment also reduced cortical contusion volume at 3 days post-injury. To determine whether this treatment paradigm attenuated TBI-induced behavioral deficits, animals were evaluated over a period of 6 weeks after surgery for forelimb placement asymmetry, contextual fear conditioning, water maze performance and spatial working memory. A33 treatment significantly improved contextual fear conditioning and water maze retention at 24 hrs post-training. However, this treatment did not rescue sensorimotor or working memory deficits. At 2 months after surgery, atrophy and neuronal loss were measured. A33 treatment significantly reduced neuronal loss in the pericontusional cortex and hippocampal CA3 region. This treatment paradigm also reduced cortical, but not hippocampal, atrophy. Overall, these results suggest that acute PDE4B inhibition may be a viable treatment to reduce inflammation, pathology and memory deficits after TBI.
Collapse
Affiliation(s)
- Nicole M. Wilson
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Mark E. Gurney
- Tetra Discovery Partners, Grand Rapids, Michigan, United States of America
| | - W. Dalton Dietrich
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Coleen M. Atkins
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| |
Collapse
|
32
|
Shultz SR, McDonald SJ, Vonder Haar C, Meconi A, Vink R, van Donkelaar P, Taneja C, Iverson GL, Christie BR. The potential for animal models to provide insight into mild traumatic brain injury: Translational challenges and strategies. Neurosci Biobehav Rev 2016; 76:396-414. [PMID: 27659125 DOI: 10.1016/j.neubiorev.2016.09.014] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 09/07/2016] [Accepted: 09/16/2016] [Indexed: 12/14/2022]
Abstract
Mild traumatic brain injury (mTBI) is a common health problem. There is tremendous variability and heterogeneity in human mTBI, including mechanisms of injury, biomechanical forces, injury severity, spatial and temporal pathophysiology, genetic factors, pre-injury vulnerability and resilience factors, and clinical outcomes. Animal models greatly reduce this variability and heterogeneity, and provide a means to study mTBI in a rigorous, controlled, and efficient manner. Rodent models, in particular, are time- and cost-efficient, and they allow researchers to measure morphological, cellular, molecular, and behavioral variables in a single study. However, inter-species differences in anatomy, morphology, metabolism, neurobiology, and lifespan create translational challenges. Although the term "mild" TBI is used often in the pre-clinical literature, clearly defined criteria for mild, moderate, and severe TBI in animal models have not been agreed upon. In this review, we introduce current issues facing the mTBI field, summarize the available research methodologies and previous studies in mTBI animal models, and discuss how a translational research approach may be useful in advancing our understanding and management of mTBI.
Collapse
Affiliation(s)
- Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Cole Vonder Haar
- Department of Psychology, The University of British Columbia, Vancouver, BC, Canada
| | - Alicia Meconi
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| | - Robert Vink
- Division of Health Sciences, The University of South Australia, Adelaide, SA, Australia
| | - Paul van Donkelaar
- School of Health and Exercise Sciences, The University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Chand Taneja
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| | - Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Home Base, A Red Sox Foundation and Massachusetts General Hospital Program, and MassGeneral Hospital for Children™ Sports Concussion Program, Boston, MA, USA
| | - Brian R Christie
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| |
Collapse
|
33
|
Sodium selenate, a protein phosphatase 2A activator, mitigates hyperphosphorylated tau and improves repeated mild traumatic brain injury outcomes. Neuropharmacology 2016; 108:382-93. [DOI: 10.1016/j.neuropharm.2016.05.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 04/26/2016] [Accepted: 05/03/2016] [Indexed: 12/14/2022]
|
34
|
Lifshitz J, Rowe RK, Griffiths DR, Evilsizor MN, Thomas TC, Adelson PD, McIntosh TK. Clinical relevance of midline fluid percussion brain injury: Acute deficits, chronic morbidities and the utility of biomarkers. Brain Inj 2016; 30:1293-1301. [PMID: 27712117 DOI: 10.1080/02699052.2016.1193628] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND After 30 years of characterisation and implementation, fluid percussion injury (FPI) is firmly recognised as one of the best-characterised reproducible and clinically relevant models of TBI, encompassing concussion through diffuse axonal injury (DAI). Depending on the specific injury parameters (e.g. injury site, mechanical force), FPI can model diffuse TBI with or without a focal component and may be designated as mild-to-severe according to the chosen mechanical forces and resulting acute neurological responses. Among FPI models, midline FPI may best represent clinical diffuse TBI, because of the acute behavioural deficits, the transition to late-onset behavioural morbidities and the absence of gross histopathology. REVIEW The goal here was to review acute and chronic physiological and behavioural deficits and morbidities associated with diffuse TBI induced by midline FPI. In the absence of neurodegenerative sequelae associated with focal injury, there is a need for biomarkers in the diagnostic, prognostic, predictive and therapeutic approaches to evaluate outcomes from TBI. CONCLUSIONS The current literature suggests that midline FPI offers a clinically-relevant, validated model of diffuse TBI to investigators wishing to evaluate novel therapeutic strategies in the treatment of TBI and the utility of biomarkers in the delivery of healthcare to patients with brain injury.
Collapse
Affiliation(s)
- Jonathan Lifshitz
- a Translational Neurotrauma Research Program , BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona, College of Medicine - Phoenix , Phoenix , AZ , USA.,c Phoenix VA Healthcare System , Phoenix , AZ , USA.,d Neuroscience Graduate Program , Arizona State University , Tempe , AZ , USA
| | - Rachel K Rowe
- a Translational Neurotrauma Research Program , BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona, College of Medicine - Phoenix , Phoenix , AZ , USA.,c Phoenix VA Healthcare System , Phoenix , AZ , USA
| | - Daniel R Griffiths
- a Translational Neurotrauma Research Program , BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona, College of Medicine - Phoenix , Phoenix , AZ , USA
| | - Megan N Evilsizor
- a Translational Neurotrauma Research Program , BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona, College of Medicine - Phoenix , Phoenix , AZ , USA
| | - Theresa C Thomas
- a Translational Neurotrauma Research Program , BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona, College of Medicine - Phoenix , Phoenix , AZ , USA.,c Phoenix VA Healthcare System , Phoenix , AZ , USA.,d Neuroscience Graduate Program , Arizona State University , Tempe , AZ , USA
| | - P David Adelson
- a Translational Neurotrauma Research Program , BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona, College of Medicine - Phoenix , Phoenix , AZ , USA.,d Neuroscience Graduate Program , Arizona State University , Tempe , AZ , USA
| | | |
Collapse
|
35
|
DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in the details. J Neurochem 2016; 139 Suppl 2:136-153. [PMID: 26990767 DOI: 10.1111/jnc.13607] [Citation(s) in RCA: 884] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/27/2016] [Accepted: 03/02/2016] [Indexed: 12/11/2022]
Abstract
There is significant interest in understanding inflammatory responses within the brain and spinal cord. Inflammatory responses that are centralized within the brain and spinal cord are generally referred to as 'neuroinflammatory'. Aspects of neuroinflammation vary within the context of disease, injury, infection, or stress. The context, course, and duration of these inflammatory responses are all critical aspects in the understanding of these processes and their corresponding physiological, biochemical, and behavioral consequences. Microglia, innate immune cells of the CNS, play key roles in mediating these neuroinflammatory responses. Because the connotation of neuroinflammation is inherently negative and maladaptive, the majority of research focus is on the pathological aspects of neuroinflammation. There are, however, several degrees of neuroinflammatory responses, some of which are positive. In many circumstances including CNS injury, there is a balance of inflammatory and intrinsic repair processes that influences functional recovery. In addition, there are several other examples where communication between the brain and immune system involves neuroinflammatory processes that are beneficial and adaptive. The purpose of this review is to distinguish different variations of neuroinflammation in a context-specific manner and detail both positive and negative aspects of neuroinflammatory processes. In this review, we will use brain and spinal cord injury, stress, aging, and other inflammatory events to illustrate the potential harm and benefits inherent to neuroinflammation. Context, course, and duration of the inflammation are highly important to the interpretation of these events, and we aim to provide insight into this by detailing several commonly studied insults. This article is part of the 60th anniversary supplemental issue.
Collapse
Affiliation(s)
- Damon J DiSabato
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| | - Ning Quan
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA. .,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
36
|
Haus DL, López-Velázquez L, Gold EM, Cunningham KM, Perez H, Anderson AJ, Cummings BJ. Transplantation of human neural stem cells restores cognition in an immunodeficient rodent model of traumatic brain injury. Exp Neurol 2016; 281:1-16. [PMID: 27079998 DOI: 10.1016/j.expneurol.2016.04.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/15/2016] [Accepted: 04/07/2016] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) in humans can result in permanent tissue damage and has been linked to cognitive impairment that lasts years beyond the initial insult. Clinically effective treatment strategies have yet to be developed. Transplantation of human neural stem cells (hNSCs) has the potential to restore cognition lost due to injury, however, the vast majority of rodent TBI/hNSC studies to date have evaluated cognition only at early time points, typically <1month post-injury and cell transplantation. Additionally, human cell engraftment and long-term survival in rodent models of TBI has been difficult to achieve due to host immunorejection of the transplanted human cells, which confounds conclusions pertaining to transplant-mediated behavioral improvement. To overcome these shortfalls, we have developed a novel TBI xenotransplantation model that utilizes immunodeficient athymic nude (ATN) rats as the host recipient for the post-TBI transplantation of human embryonic stem cell (hESC) derived NSCs and have evaluated cognition in these animals at long-term (≥2months) time points post-injury. We report that immunodeficient ATN rats demonstrate hippocampal-dependent spatial memory deficits (Novel Place, Morris Water Maze), but not non-spatial (Novel Object) or emotional/anxiety-related (Elevated Plus Maze, Conditioned Taste Aversion) deficits, at 2-3months post-TBI, confirming that ATN rats recapitulate some of the cognitive deficits found in immunosufficient animal strains. Approximately 9-25% of transplanted hNSCs survived for at least 5months post-transplantation and differentiated into mature neurons (NeuN, 18-38%), astrocytes (GFAP, 13-16%), and oligodendrocytes (Olig2, 11-13%). Furthermore, while this model of TBI (cortical impact) targets primarily cortex and the underlying hippocampus and generates a large lesion cavity, hNSC transplantation facilitated cognitive recovery without affecting either lesion volume or total spared cortical or hippocampal tissue volume. Instead, we have found an overall increase in host hippocampal neuron survival in hNSC transplanted animals and demonstrate that a correlation exists between hippocampal neuron survival and cognitive performance. Together, these findings support the use of immunodeficient rodents in models of TBI that involve the transplantation of human cells, and suggest that hNSC transplantation may be a viable, long-term therapy to restore cognition after brain injury.
Collapse
Affiliation(s)
- Daniel L Haus
- Sue & Bill Gross Stem Cell Center, University of California, Irvine,CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine,CA 92697-1750, USA
| | - Luci López-Velázquez
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA
| | - Eric M Gold
- Sue & Bill Gross Stem Cell Center, University of California, Irvine,CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine,CA 92697-1750, USA
| | - Kelly M Cunningham
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA
| | - Harvey Perez
- UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA
| | - Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, University of California, Irvine,CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine,CA 92697-1750, USA; Physical and Medical Rehabilitation, University of California, Irvine,CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA
| | - Brian J Cummings
- Sue & Bill Gross Stem Cell Center, University of California, Irvine,CA 92697-1750, USA; Anatomy & Neurobiology, University of California, Irvine,CA 92697-1750, USA; Physical and Medical Rehabilitation, University of California, Irvine,CA 92697-1750, USA; UCI Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine,CA 92697-1750, USA.
| |
Collapse
|
37
|
Kulbe JR, Geddes JW. Current status of fluid biomarkers in mild traumatic brain injury. Exp Neurol 2016; 275 Pt 3:334-352. [PMID: 25981889 PMCID: PMC4699183 DOI: 10.1016/j.expneurol.2015.05.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 05/05/2015] [Accepted: 05/08/2015] [Indexed: 01/07/2023]
Abstract
Mild traumatic brain injury (mTBI) affects millions of people annually and is difficult to diagnose. Mild injury is insensitive to conventional imaging techniques and diagnoses are often made using subjective criteria such as self-reported symptoms. Many people who sustain a mTBI develop persistent post-concussive symptoms. Athletes and military personnel are at great risk for repeat injury which can result in second impact syndrome or chronic traumatic encephalopathy. An objective and quantifiable measure, such as a serum biomarker, is needed to aid in mTBI diagnosis, prognosis, return to play/duty assessments, and would further elucidate mTBI pathophysiology. The majority of TBI biomarker research focuses on severe TBI with few studies specific to mild injury. Most studies use a hypothesis-driven approach, screening biofluids for markers known to be associated with TBI pathophysiology. This approach has yielded limited success in identifying markers that can be used clinically, additional candidate biomarkers are needed. Innovative and unbiased methods such as proteomics, microRNA arrays, urinary screens, autoantibody identification and phage display would complement more traditional approaches to aid in the discovery of novel mTBI biomarkers.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, 40536-0509, USA,; Department of Anatomy and Neurobiology, College of Medicine, University of Kentucky, Lexington, KY, 40536-0509, USA
| | - James W Geddes
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, 40536-0509, USA,; Department of Anatomy and Neurobiology, College of Medicine, University of Kentucky, Lexington, KY, 40536-0509, USA.
| |
Collapse
|
38
|
Osier N, Dixon CE. The Controlled Cortical Impact Model of Experimental Brain Trauma: Overview, Research Applications, and Protocol. Methods Mol Biol 2016; 1462:177-92. [PMID: 27604719 PMCID: PMC5271598 DOI: 10.1007/978-1-4939-3816-2_11] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Controlled cortical impact (CCI) is a commonly used and highly regarded model of brain trauma that uses a pneumatically or electromagnetically controlled piston to induce reproducible and well-controlled injury. The CCI model was originally used in ferrets and it has since been scaled for use in many other species. This chapter will describe the historical development of the CCI model, compare and contrast the pneumatic and electromagnetic models, and summarize key short- and long-term consequences of TBI that have been gleaned using this model. In accordance with the recent efforts to promote high-quality evidence through the reporting of common data elements (CDEs), relevant study details-that should be reported in CCI studies-will be noted.
Collapse
Affiliation(s)
- Nicole Osier
- Safar Center for Resuscitation Research, University of Pittsburgh, 201 Hill Building, 3434 Fifth Avenue, Pittsburgh, PA, 15213, USA
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - C Edward Dixon
- Safar Center for Resuscitation Research, University of Pittsburgh, 201 Hill Building, 3434 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
- V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
| |
Collapse
|
39
|
Turner RC, Lucke-Wold BP, Robson MJ, Lee JM, Bailes JE. Alzheimer's disease and chronic traumatic encephalopathy: Distinct but possibly overlapping disease entities. Brain Inj 2016; 30:1279-1292. [PMID: 27715315 PMCID: PMC5303562 DOI: 10.1080/02699052.2016.1193631] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE) have long been recognized as sharing some similar neuropathological features, mainly the presence of neurofibrilary tangles and hyperphosphorylated tau, but have generally been described as distinct entities. Evidence indicates that neurotrauma increases the risk of developing dementia and accelerates the progression of disease. Findings are emerging that CTE and AD may be present in the same patients. CLINICAL PRESENTATION This study presents a series of previously unpublished cases, with one case demonstrating possible neurotrauma-related AD, one pure CTE, and an example of a case exhibiting features of both AD and CTE. The future significance of this work lies not only in the confirmation of AD-CTE co-existence, but, more importantly, ways of generating a hypothesis about the possibility that CTE may accelerate AD development. Understanding the relationship between neurotrauma and neurodegenerative disease will help elucidate how distinct disease entities can co-exist in the same patient. It will ultimately require the use of pre-clinical animal models and repeat injury paradigms to investigate clinically relevant injury mechanisms. These models should produce a CTE-like phenotype that must be both neuropathologically and behaviourally similar to human disease. CONCLUSION This case series and review of the literature presents a discussion of AD and CTE in the context of neurotrauma. It highlights recent work from repetitive neurotrauma models with an emphasis on those exhibiting a CTE-like phenotype. Potential mechanisms of interest shared amongst AD and CTE are briefly addressed and future experiments are advocated for to enhance understanding of CTE pathophysiology and the relationship between CTE and AD.
Collapse
Affiliation(s)
- Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Matthew J. Robson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John M. Lee
- NorthShore Neurological Institute, NorthShore University Health System, Evanston, IL 60201
| | - Julian E. Bailes
- NorthShore Neurological Institute, NorthShore University Health System, Evanston, IL 60201
| |
Collapse
|
40
|
Webster KM, Wright DK, Sun M, Semple BD, Ozturk E, Stein DG, O'Brien TJ, Shultz SR. Progesterone treatment reduces neuroinflammation, oxidative stress and brain damage and improves long-term outcomes in a rat model of repeated mild traumatic brain injury. J Neuroinflammation 2015; 12:238. [PMID: 26683475 PMCID: PMC4683966 DOI: 10.1186/s12974-015-0457-7] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 12/13/2015] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Repeated mild traumatic brain injuries, such as concussions, may result in cumulative brain damage, neurodegeneration and other chronic neurological impairments. There are currently no clinically available treatment options known to prevent these consequences. However, growing evidence implicates neuroinflammation and oxidative stress in the pathogenesis of repetitive mild brain injuries; thus, these may represent potential therapeutic targets. Progesterone has been demonstrated to have potent anti-inflammatory and anti-oxidant properties after brain insult; therefore, here, we examined progesterone treatment in rats given repetitive mild brain injuries via the repeated mild fluid percussion injury model. METHODS Male Long-Evans rats were assigned into four groups: sham injury + vehicle treatment, sham injury + progesterone treatment (8 mg/kg/day), repeated mild fluid percussion injuries + vehicle treatment, and repeated mild fluid percussion injuries + progesterone treatment. Rats were administered a total of three injuries, with each injury separated by 5 days. Treatment was initiated 1 h after the first injury, then administered daily for a total of 15 days. Rats underwent behavioural testing at 12-weeks post-treatment to assess cognition, motor function, anxiety and depression. Brains were then dissected for analysis of markers for neuroinflammation and oxidative stress. Ex vivo MRI was conducted in order to examine structural brain damage and white matter integrity. RESULTS Repeated mild fluid percussion injuries + progesterone treatment rats showed significantly reduced cognitive and sensorimotor deficits compared to their vehicle-treated counterparts at 12-weeks post-treatment. Progesterone treatment significantly attenuated markers of neuroinflammation and oxidative stress in rats given repeated mild fluid percussion injuries, with concomitant reductions in grey and white matter damage as indicated by MRI. CONCLUSIONS These findings implicate neuroinflammation and oxidative stress in the pathophysiological aftermath of mild brain injuries and suggest that progesterone may be a viable treatment option to mitigate these effects and their detrimental consequences.
Collapse
Affiliation(s)
- Kyria M Webster
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - David K Wright
- Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Mujun Sun
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Bridgette D Semple
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Ezgi Ozturk
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia.
| |
Collapse
|
41
|
Osier ND, Carlson SW, DeSana A, Dixon CE. Chronic Histopathological and Behavioral Outcomes of Experimental Traumatic Brain Injury in Adult Male Animals. J Neurotrauma 2015; 32:1861-82. [PMID: 25490251 PMCID: PMC4677114 DOI: 10.1089/neu.2014.3680] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The purpose of this review is to survey the use of experimental animal models for studying the chronic histopathological and behavioral consequences of traumatic brain injury (TBI). The strategies employed to study the long-term consequences of TBI are described, along with a summary of the evidence available to date from common experimental TBI models: fluid percussion injury; controlled cortical impact; blast TBI; and closed-head injury. For each model, evidence is organized according to outcome. Histopathological outcomes included are gross changes in morphology/histology, ventricular enlargement, gray/white matter shrinkage, axonal injury, cerebrovascular histopathology, inflammation, and neurogenesis. Behavioral outcomes included are overall neurological function, motor function, cognitive function, frontal lobe function, and stress-related outcomes. A brief discussion is provided comparing the most common experimental models of TBI and highlighting the utility of each model in understanding specific aspects of TBI pathology. The majority of experimental TBI studies collect data in the acute postinjury period, but few continue into the chronic period. Available evidence from long-term studies suggests that many of the experimental TBI models can lead to progressive changes in histopathology and behavior. The studies described in this review contribute to our understanding of chronic TBI pathology.
Collapse
Affiliation(s)
- Nicole D. Osier
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shaun W. Carlson
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anthony DeSana
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Seton Hill University, Greensburg, Pennsylvania
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- V.A. Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
42
|
Balança B, Bapteste L, Lieutaud T, Ressnikoff D, Guy R, Bezin L, Marinesco S. Neuronal loss as evidenced by automated quantification of neuronal density following moderate and severe traumatic brain injury in rats. J Neurosci Res 2015; 94:39-49. [PMID: 26451689 DOI: 10.1002/jnr.23676] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/10/2015] [Accepted: 09/14/2015] [Indexed: 11/11/2022]
Abstract
Traumatic brain injury causes widespread neurological lesions that can be reproduced in animals with the lateral fluid percussion (LFP) model. The characterization of the pattern of neuronal death generated in this model remains unclear, involving both cortical and subcortical brain regions. Here, 7 days after moderate (3 atmospheres absolute [ATA]) or severe (3.8 ATA) LFP, we estimated neuronal loss by using immunohistochemistry together with a computer-assisted automated method for quantifying neuronal density in brain sections. Neuronal counts were performed ipsilateral to the impact, in the parietal cortex ventral to the site of percussion, in the temporal cortex, in the dorsal thalamus, and in the hippocampus. These results were compared with the counts observed at similar areas in sham animals. We found that neuronal density was severely decreased in the temporal cortex (-60%), in the dorsal thalamus (-63%), and in area CA3 of the hippocampus (-36%) of injured animals compared with controls but was not significantly modified in the cortices located immediately ventral to the impact. Total cellular density increased in brain structures displaying neuronal death, suggesting the presence of gliosis. The increase in the severity of LFP did not change the pattern of neuronal injury. This automated method simplified the study of neuronal loss following traumatic brain injury and allowed the identification of a pattern of neuronal loss that spreads from the dorsal thalamus to the temporal cortex, with the most severe lesions being in brain structures remote from the site of impact.
Collapse
Affiliation(s)
- Baptiste Balança
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team WAKING, Lyon, France.,Department of Anesthesiology and Intensive Care, P. Wertheimer Neurological Hospital, Hospices Civils de Lyon, Lyon, France.,University Claude Bernard Lyon 1, Lyon, France
| | - Lionel Bapteste
- Department of Anesthesiology and Intensive Care, P. Wertheimer Neurological Hospital, Hospices Civils de Lyon, Lyon, France.,University Claude Bernard Lyon 1, Lyon, France
| | - Thomas Lieutaud
- University Claude Bernard Lyon 1, Lyon, France.,INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
| | - Denis Ressnikoff
- University Claude Bernard Lyon 1, Lyon, France.,Centre d'Imagerie Quantitative Lyon Est, Lyon, France
| | - Rainui Guy
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team WAKING, Lyon, France.,University Claude Bernard Lyon 1, Lyon, France
| | - Laurent Bezin
- University Claude Bernard Lyon 1, Lyon, France.,INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team TIGER, Lyon, France
| | - Stéphane Marinesco
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team WAKING, Lyon, France.,University Claude Bernard Lyon 1, Lyon, France.,INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, AniRA-Neurochem Technological Platform, Lyon, France
| |
Collapse
|
43
|
Astrocytes Are Primed by Chronic Neurodegeneration to Produce Exaggerated Chemokine and Cell Infiltration Responses to Acute Stimulation with the Cytokines IL-1β and TNF-α. J Neurosci 2015; 35:8411-22. [PMID: 26041910 DOI: 10.1523/jneurosci.2745-14.2015] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Microgliosis and astrogliosis are standard pathological features of neurodegenerative disease. Microglia are primed by chronic neurodegeneration such that toll-like receptor agonists, such as LPS, drive exaggerated cytokine responses on this background. However, sterile inflammatory insults are more common than direct CNS infection in the degenerating brain and these insults drive robust IL-1β and TNF-α responses. It is unclear whether these pro-inflammatory cytokines can directly induce exaggerated responses in the degenerating brain. We hypothesized that glial cells in the hippocampus of animals with chronic neurodegenerative disease (ME7 prion disease) would display exaggerated responses to central cytokine challenges. TNF-α or IL-1β were administered intrahippocampally to ME7-inoculated mice and normal brain homogenate-injected (NBH) controls. Both IL-1β and TNF-α produced much more robust IL-1β synthesis in ME7 than in NBH animals and this occurred exclusively in microglia. However, there was strong nuclear localization of the NFκB subunit p65 in the astrocyte population, associated with marked astrocytic synthesis of the chemokines CXCL1 and CCL2 in response to both cytokine challenges in ME7 animals. Conversely, very limited expression of these chemokines was apparent in NBH animals similarly challenged. Thus, astrocytes are primed in the degenerating brain to produce exaggerated chemokine responses to acute stimulation with pro-inflammatory cytokines. Furthermore, this results in markedly increased neutrophil, T-cell, and monocyte infiltration in the diseased brain. These data have significant implications for acute sterile inflammatory insults such as stroke and traumatic brain injury occurring on a background of aging or neurodegeneration.
Collapse
|
44
|
Tibial fracture exacerbates traumatic brain injury outcomes and neuroinflammation in a novel mouse model of multitrauma. J Cereb Blood Flow Metab 2015; 35:1339-47. [PMID: 25853909 PMCID: PMC4528010 DOI: 10.1038/jcbfm.2015.56] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/26/2015] [Accepted: 03/05/2015] [Indexed: 11/08/2022]
Abstract
Multitrauma is a common medical problem worldwide, and often involves concurrent traumatic brain injury (TBI) and bone fracture. Despite the high incidence of combined TBI and fracture, preclinical TBI research commonly employs independent injury models that fail to incorporate the pathophysiologic interactions occurring in multitrauma. Here, we developed a novel mouse model of multitrauma, and investigated whether bone fracture worsened TBI outcomes. Male mice were assigned into four groups: sham-TBI+sham-fracture (SHAM); sham-TBI+fracture (FX); TBI+sham-fracture (TBI); and TBI+fracture (MULTI). The injury methods included a closed-skull weight-drop TBI model and a closed tibial fracture. After a 35-day recovery, mice underwent behavioral testing and magnetic resonance imaging (MRI). MULTI mice displayed abnormal behaviors in the open-field compared with all other groups. On MRI, MULTI mice had enlarged ventricles and diffusion abnormalities compared with all other groups. These changes occurred in the presence of heightened neuroinflammation in MULTI mice at 24 hours and 35 days after injury, and elevated edema and blood-brain barrier disruption at 24 hours after injury. Together, these findings indicate that tibial fracture worsens TBI outcomes, and that exacerbated neuroinflammation may be an important factor that contributes to these effects, which warrants further investigation.
Collapse
|
45
|
Weaver LC, Bao F, Dekaban GA, Hryciw T, Shultz SR, Cain DP, Brown A. CD11d integrin blockade reduces the systemic inflammatory response syndrome after traumatic brain injury in rats. Exp Neurol 2015; 271:409-22. [PMID: 26169930 DOI: 10.1016/j.expneurol.2015.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 07/04/2015] [Indexed: 12/15/2022]
Abstract
Traumatic CNS injury triggers a systemic inflammatory response syndrome (SIRS), in which circulating inflammatory cells invade body organs causing local inflammation and tissue damage. We have shown that the SIRS caused by spinal cord injury is greatly reduced by acute intravenous treatment with an antibody against the CD11d subunit of the CD11d/CD18 integrin expressed by neutrophils and monocyte/macrophages, a treatment that reduces their efflux from the circulation. Traumatic brain injury (TBI) is a frequently occurring injury after motor vehicle accidents, sporting and military injuries, and falls. Our studies have shown that the anti-CD11d treatment diminishes brain inflammation and oxidative injury after moderate or mild TBI, improving neurological outcomes. Accordingly, we examined the impact of this treatment on the SIRS triggered by TBI. The anti-CD11d treatment was given at 2h after a single moderate (2.5-3.0 atm) or 2 and 24h after each of three consecutive mild (1.0-1.5 atm) fluid percussion TBIs. Sham-injured, saline-treated rats served as controls. At 24h, 72 h, and 4 or 8 weeks after the single TBI and after the third of three TBIs, lungs of rats were examined histochemically, immunocytochemically and biochemically for downstream effects of SIRS including inflammation, tissue damage and expression of oxidative enzymes. Lung sections revealed that both the single moderate and repeated mild TBI caused alveolar disruption, thickening of inter-alveolar tissue, hemorrhage into the parenchyma and increased density of intra-and peri-alveolar macrophages. The anti-CD11d treatment decreased the intrapulmonary influx of neutrophils and the density of activated macrophages and the activity of myeloperoxidase after these TBIs. Moreover, Western blotting studies showed that the treatment decreased lung protein levels of oxidative enzymes gp91(phox), inducible nitric oxide synthase and cyclooxygenase-2, as well as the apoptotic pathway enzyme caspase-3 and levels of 4-hydroxynonenal-bound proteins (an indicator of lipid peroxidation). Decreased expression of the cytoprotective transcription factor Nrf2 reflected decreased lung oxidative stress. Anti-CD11d treatment also diminished the lung concentration of free radicals and tissue aldehydes. In conclusion, the substantial lung component of the SIRS after single or repeated TBIs is significantly decreased by a simple, minimally invasive and short-lasting anti-inflammatory treatment.
Collapse
Affiliation(s)
- Lynne C Weaver
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario N6A 5B7, Canada.
| | - Feng Bao
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Gregory A Dekaban
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Todd Hryciw
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Sandy R Shultz
- Program in Neuroscience, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Psychology, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Donald P Cain
- Program in Neuroscience, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Psychology, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Arthur Brown
- Spinal Cord Injury Team, Molecular Medicine, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Program in Neuroscience, The University of Western Ontario, London, Ontario N6A 5B7, Canada; Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| |
Collapse
|
46
|
Skendelas JP, Muccigrosso M, Eiferman DS, Godbout JP. Chronic Inflammation After TBI and Associated Behavioral Sequelae. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2015. [DOI: 10.1007/s40141-015-0091-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Johnstone VPA, Wright DK, Wong K, O'Brien TJ, Rajan R, Shultz SR. Experimental Traumatic Brain Injury Results in Long-Term Recovery of Functional Responsiveness in Sensory Cortex but Persisting Structural Changes and Sensorimotor, Cognitive, and Emotional Deficits. J Neurotrauma 2015; 32:1333-46. [PMID: 25739059 DOI: 10.1089/neu.2014.3785] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death worldwide. In recent studies, we have shown that experimental TBI caused an immediate (24-h post) suppression of neuronal processing, especially in supragranular cortical layers. We now examine the long-term effects of experimental TBI on the sensory cortex and how these changes may contribute to a range of TBI morbidities. Adult male Sprague-Dawley rats received either a moderate lateral fluid percussion injury (n=14) or a sham surgery (n=12) and 12 weeks of recovery before behavioral assessment, magnetic resonance imaging, and electrophysiological recordings from the barrel cortex. TBI rats demonstrated sensorimotor deficits, cognitive impairments, and anxiety-like behavior, and this was associated with significant atrophy of the barrel cortex and other brain structures. Extracellular recordings from ipsilateral barrel cortex revealed normal neuronal responsiveness and diffusion tensor MRI showed increased fractional anisotropy, axial diffusivity, and tract density within this region. These findings suggest that long-term recovery of neuronal responsiveness is owing to structural reorganization within this region. Therefore, it is likely that long-term structural and functional changes within sensory cortex post-TBI may allow for recovery of neuronal responsiveness, but that this recovery does not remediate all behavioral deficits.
Collapse
Affiliation(s)
| | - David K Wright
- 2 Anatomy and Neuroscience, The University of Melbourne, Parkville VIC, Australia, and The Florey Institute of Neuroscience and Mental Health , Parkville, VIC, Australia
| | - Kendrew Wong
- 3 Department of Medicine, The Royal Melbourne Hospital, The Melbourne Brain Center, The University of Melbourne , Parkville, VIC, Australia
| | - Terence J O'Brien
- 3 Department of Medicine, The Royal Melbourne Hospital, The Melbourne Brain Center, The University of Melbourne , Parkville, VIC, Australia
| | - Ramesh Rajan
- 4 Department of Physiology, Monash University , Clayton VIC, Australia
| | - Sandy R Shultz
- 3 Department of Medicine, The Royal Melbourne Hospital, The Melbourne Brain Center, The University of Melbourne , Parkville, VIC, Australia
| |
Collapse
|
48
|
Rathbone ATL, Tharmaradinam S, Jiang S, Rathbone MP, Kumbhare DA. A review of the neuro- and systemic inflammatory responses in post concussion symptoms: Introduction of the "post-inflammatory brain syndrome" PIBS. Brain Behav Immun 2015; 46:1-16. [PMID: 25736063 DOI: 10.1016/j.bbi.2015.02.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 12/22/2022] Open
Abstract
Post-concussion syndrome is an aggregate of symptoms that commonly present together after head injury. These symptoms, depending on definition, include headaches, dizziness, neuropsychiatric symptoms, and cognitive impairment. However, these symptoms are common, occurring frequently in non-head injured controls, leading some to question the existence of post-concussion syndrome as a unique syndrome. Therefore, some have attempted to explain post-concussion symptoms as post-traumatic stress disorder, as they share many similar symptoms and post-traumatic stress disorder does not require head injury. This explanation falls short as patients with post-concussion syndrome do not necessarily experience many key symptoms of post-traumatic stress disorder. Therefore, other explanations must be sought to explain the prevalence of post-concussion like symptoms in non-head injury patients. Many of the situations in which post-concussion syndrome like symptoms may be experienced such as infection and post-surgery are associated with systemic inflammatory responses, and even neuroinflammation. Post-concussion syndrome itself has a significant neuroinflammatory component. In this review we examine the evidence of neuroinflammation in post-concussion syndrome and the potential role systemic inflammation plays in post-concussion syndrome like symptoms. We conclude that given the overlap between these conditions and the role of inflammation in their etiologies, a new term, post-inflammatory brain syndromes (PIBS), is necessary to describe the common outcomes of many different inflammatory insults. The concept of post-concussion syndrome is in its evolution therefore, the new term post-inflammatory brain syndromes provides a better understanding of etiology of its wide-array of symptoms and the wide array of conditions they can be seen in.
Collapse
Affiliation(s)
| | - Surejini Tharmaradinam
- Division of Pediatric Neurology, Department of Pediatrics, McMaster Children's Hospital, Pediatric Neurology, MUMC 3A, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Shucui Jiang
- Division of Neurosurgery, Department of Surgery, and Hamilton Neurorestorative Group, McMaster University, HSC 4E15, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Michel P Rathbone
- Department of Medicine, Division of Neurology, McMaster University - Juravinski Hospital, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada.
| | - Dinesh A Kumbhare
- Division of Physical Medicine and Rehabilitation, Department of Medicine, University of Toronto, University Health Network - Toronto Rehab - University Centre, 550 University Ave, Toronto, Ontario M5G 2A2, Canada
| |
Collapse
|
49
|
Shultz SR, Wright DK, Zheng P, Stuchbery R, Liu SJ, Sashindranath M, Medcalf RL, Johnston LA, Hovens CM, Jones NC, O'Brien TJ. Sodium selenate reduces hyperphosphorylated tau and improves outcomes after traumatic brain injury. Brain 2015; 138:1297-313. [PMID: 25771151 DOI: 10.1093/brain/awv053] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 01/10/2015] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury is a common and serious neurodegenerative condition that lacks a pharmaceutical intervention to improve long-term outcome. Hyperphosphorylated tau is implicated in some of the consequences of traumatic brain injury and is a potential pharmacological target. Protein phosphatase 2A is a heterotrimeric protein that regulates key signalling pathways, and protein phosphatase 2A heterotrimers consisting of the PR55 B-subunit represent the major tau phosphatase in the brain. Here we investigated whether traumatic brain injury in rats and humans would induce changes in protein phosphatase 2A and phosphorylated tau, and whether treatment with sodium selenate-a potent PR55 activator-would reduce phosphorylated tau and improve traumatic brain injury outcomes in rats. Ninety young adult male Long-Evans rats were administered either a fluid percussion injury or sham-injury. A proportion of rats were killed at 2, 24, and 72 h post-injury to assess acute changes in protein phosphatase 2A and tau. Other rats were given either sodium selenate or saline-vehicle treatment that was continuously administered via subcutaneous osmotic pump for 12 weeks. Serial magnetic resonance imaging was acquired prior to, and at 1, 4, and 12 weeks post-injury to assess evolving structural brain damage and axonal injury. Behavioural impairments were assessed at 12 weeks post-injury. The results showed that traumatic brain injury in rats acutely reduced PR55 expression and protein phosphatase 2A activity, and increased the expression of phosphorylated tau and the ratio of phosphorylated tau to total tau. Similar findings were seen in post-mortem brain samples from acute human traumatic brain injury patients, although many did not reach statistical significance. Continuous sodium selenate treatment for 12 weeks after sham or fluid percussion injury in rats increased protein phosphatase 2A activity and PR55 expression, and reduced the ratio of phosphorylated tau to total tau, attenuated brain damage, and improved behavioural outcomes in rats given a fluid percussion injury. Notably, total tau levels were decreased in rats 12 weeks after fluid percussion injury, and several other factors, including the use of anaesthetic, the length of recovery time, and that some brain injury and behavioural dysfunction still occurred in rats treated with sodium selenate must be considered in the interpretation of this study. However, taken together these data suggest protein phosphatase 2A and hyperphosphorylated tau may be involved in the neurodegenerative cascade of traumatic brain injury, and support the potential use of sodium selenate as a novel traumatic brain injury therapy.
Collapse
Affiliation(s)
- Sandy R Shultz
- 1 Melbourne Brain Centre, Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - David K Wright
- 2 Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ping Zheng
- 1 Melbourne Brain Centre, Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Ryan Stuchbery
- 3 Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Shi-Jie Liu
- 1 Melbourne Brain Centre, Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Maithili Sashindranath
- 4 Australian Centre for Blood Disease, Monash University, Melbourne, Victoria, 3004, Australia
| | - Robert L Medcalf
- 4 Australian Centre for Blood Disease, Monash University, Melbourne, Victoria, 3004, Australia
| | - Leigh A Johnston
- 5 Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Christopher M Hovens
- 3 Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Nigel C Jones
- 1 Melbourne Brain Centre, Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Terence J O'Brien
- 1 Melbourne Brain Centre, Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| |
Collapse
|
50
|
Prasad KN, Bondy SC. Common biochemical defects linkage between post-traumatic stress disorders, mild traumatic brain injury (TBI) and penetrating TBI. Brain Res 2014; 1599:103-14. [PMID: 25553619 DOI: 10.1016/j.brainres.2014.12.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/18/2014] [Accepted: 12/19/2014] [Indexed: 12/29/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a complex mental disorder with psychological and emotional components, caused by exposure to single or repeated extreme traumatic events found in war, terrorist attacks, natural or man-caused disasters, and by violent personal assaults and accidents. Mild traumatic brain injury (TBI) occurs when the brain is violently rocked back and forth within the skull following a blow to the head or neck as in contact sports, or when in close proximity to a blast pressure wave following detonation of explosives in the battlefield. Penetrating TBI occurs when an object penetrates the skull and damages the brain, and is caused by vehicle crashes, gunshot wound to the head, and exposure to solid fragments in the proximity of explosions, and other combat-related head injuries. Despite clinical studies and improved understanding of the mechanisms of cellular damage, prevention and treatment strategies for patients with PTSD and TBI remain unsatisfactory. To develop an improved plan for treating and impeding progression of PTSD and TBI, it is important to identify underlying biochemical changes that may play key role in the initiation and progression of these disorders. This review identifies three common biochemical events, namely oxidative stress, chronic inflammation and excitotoxicity that participate in the initiation and progression of these conditions. While these features are separately discussed, in many instances, they overlap. This review also addresses the goal of developing novel treatments and drug regimens, aimed at combating this triad of events common to, and underlying, injury to the brain.
Collapse
Affiliation(s)
- Kedar N Prasad
- Antioxidant Research Institute, Premier Micronutrient Corporation, 14 Galli Drive, suite 200, Novato, CA 94949, USA.
| | - Stephen C Bondy
- Center for Occupational and Environmental Health, Department of Medicine, University of California, Irvine, CA 92697-1830, USA.
| |
Collapse
|