1
|
Mazhari A, Shafieian M. Toward understanding the brain tissue behavior due to preconditioning: an experimental study and RVE approach. Front Bioeng Biotechnol 2024; 12:1462148. [PMID: 39439552 PMCID: PMC11493751 DOI: 10.3389/fbioe.2024.1462148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Brain tissue under preconditioning, as a complex issue, refers to repeated loading-unloading cycles applied in mechanical testing protocols. In previous studies, only the mechanical behavior of the tissue under preconditioning was investigated; However, the link between macrostructural mechanical behavior and microstructural changes in brain tissue remains underexplored. This study aims to bridge this gap by investigating bovine brain tissue responses both before and after preconditioning. We employed a dual approach: experimental mechanical testing and computational modeling. Experimental tests were conducted to observe microstructural changes in mechanical behavior due to preconditioning, with a focus on axonal damage. Concurrently, we developed multiscale models using statistically representative volume elements (RVE) to simulate the tissue's microstructural response. These RVEs, featuring randomly distributed axonal fibers within the extracellular matrix, provide a realistic depiction of the white matter microstructure. Our findings show that preconditioning induces significant changes in the mechanical properties of brain tissue and affects axonal integrity. The RVE models successfully captured localized stresses and facilitated the microscopic analysis of axonal injury mechanisms. These results underscore the importance of considering both macro and micro scales in understanding brain tissue behavior under mechanical loading. This comprehensive approach offers valuable insights into mechanotransduction processes and improves the analysis of microstructural phenomena in brain tissue.
Collapse
Affiliation(s)
| | - Mehdi Shafieian
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnique), Tehran, Iran
| |
Collapse
|
2
|
Sabetta Z, Krishna G, Curry-Koski T, Lopez M, Adelson PD, Thomas TC. Sex-dependent temporal changes in astrocyte-vessel interactions following diffuse traumatic brain injury in rats. Front Physiol 2024; 15:1469073. [PMID: 39387100 PMCID: PMC11461938 DOI: 10.3389/fphys.2024.1469073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Traumatic brain injury (TBI) is associated with diffuse axonal injury (DAI), a primary pathology linked to progressive neurodegeneration and neuroinflammation, including chronic astrogliosis, which influences long-term post-TBI recovery and morbidity. Sex-based differences in blood-brain barrier (BBB) permeability increases the risk of accelerated brain aging and early-onset neurodegeneration. However, few studies have evaluated chronic time course of astrocytic responses around cerebrovascular in the context of aging after TBI and sex dependence. We observed increased glial fibrillary acidic protein (GFAP)-labeled accessory processes branching near and connecting with GFAP-ensheathed cortical vessels, suggesting a critical nuance in astrocyte-vessel interactions after TBI. To quantify this observation, male and female Sprague Dawley rats (∼3 months old, n = 5-6/group) underwent either sham surgery or midline fluid percussion injury. Using immunohistochemical analysis, we quantified GFAP-labeled astrocyte primary and accessory processes that contacted GFAP-ensheathed vessels in the somatosensory barrel cortex at 7, 56, and 168 days post-injury (DPI). TBI significantly increased GFAP-positive primary processes at 7 DPI (P < 0.01) in both sexes. At 56 DPI, these vessel-process interactions remained significantly increased exclusively in males (P < 0.05). At 168 DPI, both sexes showed a significant reduction in vessel-process interactions compared to 7 DPI (P < 0.05); however, a modest but significant injury effect reemerged in females (P < 0.05). A similar sex-dependent pattern in the number of accessory processes provides novel evidence of long-term temporal changes in astrocyte-vessel interactions. TBI-induced changes in astrocyte-vessel interactions may indicate chronic BBB vulnerability and processes responsible for early onset vascular and neurodegenerative pathology.
Collapse
Affiliation(s)
- Zackary Sabetta
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- A.T. Still University Kirksville College of Osteopathic Medicine, Kirksville, MO, United States
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Tala Curry-Koski
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Mackenzie Lopez
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - P. David Adelson
- West Virginia University School of Medicine, Rockefeller Neuroscience Institute, Morgantown, WV, United States
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
- Phoenix VA Healthcare System, Phoenix, AZ, United States
| |
Collapse
|
3
|
Alkaslasi MR, Lloyd EYH, Gable AS, Silberberg H, Yarur HE, Tsai VS, Sohn M, Margolin G, Tejeda HA, Le Pichon CE. The transcriptional response of cortical neurons to concussion reveals divergent fates after injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.581939. [PMID: 38463961 PMCID: PMC10925231 DOI: 10.1101/2024.02.26.581939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Traumatic brain injury (TBI) is a risk factor for neurodegeneration, however little is known about how different neuron types respond to this kind of injury. In this study, we follow neuronal populations over several months after a single mild TBI (mTBI) to assess long ranging consequences of injury at the level of single, transcriptionally defined neuronal classes. We find that the stress responsive Activating Transcription Factor 3 (ATF3) defines a population of cortical neurons after mTBI. We show that neurons that activate ATF3 upregulate stress-related genes while repressing many genes, including commonly used markers for these cell types. Using an inducible reporter linked to ATF3, we genetically mark damaged cells to track them over time. Notably, we find that a population in layer V undergoes cell death acutely after injury, while another in layer II/III survives long term and retains the ability to fire action potentials. To investigate the mechanism controlling layer V neuron death, we genetically silenced candidate stress response pathways. We found that the axon injury responsive kinase MAP3K12, also known as dual leucine zipper kinase (DLK), is required for the layer V neuron death. This work provides a rationale for targeting the DLK signaling pathway as a therapeutic intervention for traumatic brain injury. Beyond this, our novel approach to track neurons after a mild, subclinical injury can inform our understanding of neuronal susceptibility to repeated impacts.
Collapse
Affiliation(s)
- Mor R. Alkaslasi
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Eliza Y. H. Lloyd
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Austin S. Gable
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hanna Silberberg
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hector E. Yarur
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Valerie S. Tsai
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Mira Sohn
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gennady Margolin
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hugo A. Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Claire E. Le Pichon
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Syzdykbayev M, Kazymov M, Aubakirov M, Kurmangazina A, Kairkhanov E, Kazangapov R, Bryzhakhina Z, Imangazinova S, Sheinin A. A Modern Approach to the Treatment of Traumatic Brain Injury. MEDICINES (BASEL, SWITZERLAND) 2024; 11:10. [PMID: 38786549 PMCID: PMC11123131 DOI: 10.3390/medicines11050010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/18/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
Background: Traumatic brain injury manifests itself in various forms, ranging from mild impairment of consciousness to severe coma and death. Traumatic brain injury remains one of the leading causes of morbidity and mortality. Currently, there is no therapy to reverse the effects associated with traumatic brain injury. New neuroprotective treatments for severe traumatic brain injury have not achieved significant clinical success. Methods: A literature review was performed to summarize the recent interdisciplinary findings on management of traumatic brain injury from both clinical and experimental perspective. Results: In the present review, we discuss the concepts of traditional and new approaches to treatment of traumatic brain injury. The recent development of different drug delivery approaches to the central nervous system is also discussed. Conclusions: The management of traumatic brain injury could be aimed either at the pathological mechanisms initiating the secondary brain injury or alleviating the symptoms accompanying the injury. In many cases, however, the treatment should be complex and include a variety of medical interventions and combination therapy.
Collapse
Affiliation(s)
- Marat Syzdykbayev
- Department of Hospital Surgery, Anesthesiology and Reanimatology, Semey Medical University, Semey 071400, Kazakhstan
| | - Maksut Kazymov
- Department of General Practitioners, Semey Medical University, Semey 071400, Kazakhstan
| | - Marat Aubakirov
- Department of Pediatric Surgery, Semey Medical University, Semey 071400, Kazakhstan
| | - Aigul Kurmangazina
- Committee for Medical and Pharmaceutical Control of the Ministry of Health of the Republic of Kazakhstan for East Kazakhstan Region, Ust-Kamenogorsk 070004, Kazakhstan
| | - Ernar Kairkhanov
- Pavlodar Branch of Semey Medical University, Pavlodar S03Y3M1, Kazakhstan
| | - Rustem Kazangapov
- Pavlodar Branch of Semey Medical University, Pavlodar S03Y3M1, Kazakhstan
| | - Zhanna Bryzhakhina
- Department Psychiatry and Narcology, Semey Medical University, Semey 071400, Kazakhstan
| | - Saule Imangazinova
- Department of Therapy, Astana Medical University, Astana 010000, Kazakhstan
| | - Anton Sheinin
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
5
|
Fesharaki-Zadeh A, Datta D. An overview of preclinical models of traumatic brain injury (TBI): relevance to pathophysiological mechanisms. Front Cell Neurosci 2024; 18:1371213. [PMID: 38682091 PMCID: PMC11045909 DOI: 10.3389/fncel.2024.1371213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of morbidity and mortality, affecting millions annually worldwide. Although the majority of TBI patients return to premorbid baseline, a subset of patient can develop persistent and often debilitating neurocognitive and behavioral changes. The etiology of TBI within the clinical setting is inherently heterogenous, ranging from sport related injuries, fall related injuries and motor vehicle accidents in the civilian setting, to blast injuries in the military setting. Objective Animal models of TBI, offer the distinct advantage of controlling for injury modality, duration and severity. Furthermore, preclinical models of TBI have provided the necessary temporal opportunity to study the chronic neuropathological sequelae of TBI, including neurodegenerative sequelae such as tauopathy and neuroinflammation within the finite experimental timeline. Despite the high prevalence of TBI, there are currently no disease modifying regimen for TBI, and the current clinical treatments remain largely symptom based. The preclinical models have provided the necessary biological substrate to examine the disease modifying effect of various pharmacological agents and have imperative translational value. Methods The current review will include a comprehensive survey of well-established preclinical models, including classic preclinical models including weight drop, blast injury, fluid percussion injury, controlled cortical impact injury, as well as more novel injury models including closed-head impact model of engineered rotational acceleration (CHIMERA) models and closed-head projectile concussive impact model (PCI). In addition to rodent preclinical models, the review will include an overview of other species including large animal models and Drosophila. Results There are major neuropathological perturbations post TBI captured in various preclinical models, which include neuroinflammation, calcium dysregulation, tauopathy, mitochondrial dysfunction and oxidative stress, axonopathy, as well as glymphatic system disruption. Conclusion The preclinical models of TBI continue to offer valuable translational insight, as well as essential neurobiological basis to examine specific disease modifying therapeutic regimen.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Department of Neurology and Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dibyadeep Datta
- Division of Aging and Geriatric Psychiatry, Alzheimer’s Disease Research Unit, Department of Psychiatry, New Haven, CT, United States
| |
Collapse
|
6
|
Katchur NJ, Notterman DA. Recent insights from non-mammalian models of brain injuries: an emerging literature. Front Neurol 2024; 15:1378620. [PMID: 38566857 PMCID: PMC10985199 DOI: 10.3389/fneur.2024.1378620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Traumatic brain injury (TBI) is a major global health concern and is increasingly recognized as a risk factor for neurodegenerative diseases including Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Repetitive TBIs (rTBIs), commonly observed in contact sports, military service, and intimate partner violence (IPV), pose a significant risk for long-term sequelae. To study the long-term consequences of TBI and rTBI, researchers have typically used mammalian models to recapitulate brain injury and neurodegenerative phenotypes. However, there are several limitations to these models, including: (1) lengthy observation periods, (2) high cost, (3) difficult genetic manipulations, and (4) ethical concerns regarding prolonged and repeated injury of a large number of mammals. Aquatic vertebrate model organisms, including Petromyzon marinus (sea lampreys), zebrafish (Danio rerio), and invertebrates, Caenorhabditis elegans (C. elegans), and Drosophila melanogaster (Drosophila), are emerging as valuable tools for investigating the mechanisms of rTBI and tauopathy. These non-mammalian models offer unique advantages, including genetic tractability, simpler nervous systems, cost-effectiveness, and quick discovery-based approaches and high-throughput screens for therapeutics, which facilitate the study of rTBI-induced neurodegeneration and tau-related pathology. Here, we explore the use of non-vertebrate and aquatic vertebrate models to study TBI and neurodegeneration. Drosophila, in particular, provides an opportunity to explore the longitudinal effects of mild rTBI and its impact on endogenous tau, thereby offering valuable insights into the complex interplay between rTBI, tauopathy, and neurodegeneration. These models provide a platform for mechanistic studies and therapeutic interventions, ultimately advancing our understanding of the long-term consequences associated with rTBI and potential avenues for intervention.
Collapse
Affiliation(s)
- Nicole J. Katchur
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
- Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Daniel A. Notterman
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| |
Collapse
|
7
|
Chen Q, Li L, Xu L, Yang B, Huang Y, Qiao D, Yue X. Proteomic analysis discovers potential biomarkers of early traumatic axonal injury in the brainstem. Int J Legal Med 2024; 138:207-227. [PMID: 37338605 DOI: 10.1007/s00414-023-03039-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/01/2023] [Indexed: 06/21/2023]
Abstract
OBJECTIVE Application of Tandem Mass Tags (TMT)-based LC-MS/MS analysis to screen for differentially expressed proteins (DEPs) in traumatic axonal injury (TAI) of the brainstem and to predict potential biomarkers and key molecular mechanisms of brainstem TAI. METHODS A modified impact acceleration injury model was used to establish a brainstem TAI model in Sprague-Dawley rats, and the model was evaluated in terms of both functional changes (vital sign measurements) andstructural changes (HE staining, silver-plating staining and β-APP immunohistochemical staining). TMT combined with LC-MS/MS was used to analyse the DEPs in brainstem tissues from TAI and Sham groups. The biological functions of DEPs and potential molecular mechanisms in the hyperacute phase of TAI were analysed by bioinformatics techniques, and candidate biomarkers were validated using western blotting and immunohistochemistry on brainstem tissues from animal models and humans. RESULTS Based on the successful establishment of the brainstem TAI model in rats, TMT-based proteomics identified 65 DEPs, and bioinformatics analysis indicated that the hyperacute phase of TAI involves multiple stages of biological processes including inflammation, oxidative stress, energy metabolism, neuronal excitotoxicity and apoptosis. Three DEPs, CBR1, EPHX2 and CYP2U1, were selected as candidate biomarkers and all three proteins were found to be significantly expressed in brainstem tissue 30 min-7 days after TAI in both animal models and humans. CONCLUSION Using TMT combined with LC-MS/MS analysis for proteomic study of early TAI in rat brainstem, we report for the first time that CBR1, EPHX2 and CYP2U1 can be used as biomarkers of early TAI in brainstem by means of western blotting and immunohistochemical staining, compensating for the limitations of silver-plating staining and β-APP immunohistochemical staining, especially in the case of very short survival time after TAI (shorter than 30 min). A number of other proteins that also have a potential marker role are also presented, providing new insights into the molecular mechanisms, therapeutic targets and forensic identification of early TAI in brainstem.
Collapse
Affiliation(s)
- Qianling Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Lingyue Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Luyao Xu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Bin Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yuebing Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Dongfang Qiao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Xia Yue
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
8
|
Krieg JL, Leonard AV, Turner RJ, Corrigan F. Identifying the Phenotypes of Diffuse Axonal Injury Following Traumatic Brain Injury. Brain Sci 2023; 13:1607. [PMID: 38002566 PMCID: PMC10670443 DOI: 10.3390/brainsci13111607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Diffuse axonal injury (DAI) is a significant feature of traumatic brain injury (TBI) across all injury severities and is driven by the primary mechanical insult and secondary biochemical injury phases. Axons comprise an outer cell membrane, the axolemma which is anchored to the cytoskeletal network with spectrin tetramers and actin rings. Neurofilaments act as space-filling structural polymers that surround the central core of microtubules, which facilitate axonal transport. TBI has differential effects on these cytoskeletal components, with axons in the same white matter tract showing a range of different cytoskeletal and axolemma alterations with different patterns of temporal evolution. These require different antibodies for detection in post-mortem tissue. Here, a comprehensive discussion of the evolution of axonal injury within different cytoskeletal elements is provided, alongside the most appropriate methods of detection and their temporal profiles. Accumulation of amyloid precursor protein (APP) as a result of disruption of axonal transport due to microtubule failure remains the most sensitive marker of axonal injury, both acutely and chronically. However, a subset of injured axons demonstrate different pathology, which cannot be detected via APP immunoreactivity, including degradation of spectrin and alterations in neurofilaments. Furthermore, recent work has highlighted the node of Ranvier and the axon initial segment as particularly vulnerable sites to axonal injury, with loss of sodium channels persisting beyond the acute phase post-injury in axons without APP pathology. Given the heterogenous response of axons to TBI, further characterization is required in the chronic phase to understand how axonal injury evolves temporally, which may help inform pharmacological interventions.
Collapse
Affiliation(s)
| | | | | | - Frances Corrigan
- Translational Neuropathology Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (J.L.K.)
| |
Collapse
|
9
|
Denniss RJ, Barker LA. Brain Trauma and the Secondary Cascade in Humans: Review of the Potential Role of Vitamins in Reparative Processes and Functional Outcome. Behav Sci (Basel) 2023; 13:bs13050388. [PMID: 37232626 DOI: 10.3390/bs13050388] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
An estimated sixty-nine million people sustain a traumatic brain injury each year. Trauma to the brain causes the primary insult and initiates a secondary biochemical cascade as part of the immune and reparative response to injury. The secondary cascade, although a normal physiological response, may also contribute to ongoing neuroinflammation, oxidative stress and axonal injury, continuing in some cases years after the initial insult. In this review, we explain some of the biochemical mechanisms of the secondary cascade and their potential deleterious effects on healthy neurons including secondary cell death. The second part of the review focuses on the role of micronutrients to neural mechanisms and their potential reparative effects with regards to the secondary cascade after brain injury. The biochemical response to injury, hypermetabolism and excessive renal clearance of nutrients after injury increases the demand for most vitamins. Currently, most research in the area has shown positive outcomes of vitamin supplementation after brain injury, although predominantly in animal (murine) models. There is a pressing need for more research in this area with human participants because vitamin supplementation post-trauma is a potential cost-effective adjunct to other clinical and therapeutic treatments. Importantly, traumatic brain injury should be considered a lifelong process and better evaluated across the lifespan of individuals who experience brain injury.
Collapse
Affiliation(s)
- Rebecca J Denniss
- Department of Psychology, The University of Sheffield, Sheffield S10 2TN, UK
| | - Lynne A Barker
- Centre for Behavioural Science and Applied Psychology, Department of Psychology, Sociology and Politics, Sheffield Hallam University, Sheffield S1 1WB, UK
| |
Collapse
|
10
|
Katzenberger RJ, Ganetzky B, Wassarman DA. Lissencephaly-1 mutations enhance traumatic brain injury outcomes in Drosophila. Genetics 2023; 223:iyad008. [PMID: 36683334 PMCID: PMC9991514 DOI: 10.1093/genetics/iyad008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/14/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Traumatic brain injury (TBI) outcomes vary greatly among individuals, but most of the variation remains unexplained. Using a Drosophila melanogaster TBI model and 178 genetically diverse lines from the Drosophila Genetic Reference Panel (DGRP), we investigated the role that genetic variation plays in determining TBI outcomes. Following injury at 20-27 days old, DGRP lines varied considerably in mortality within 24 h ("early mortality"). Additionally, the disparity in early mortality resulting from injury at 20-27 vs 0-7 days old differed among DGRP lines. These data support a polygenic basis for differences in TBI outcomes, where some gene variants elicit their effects by acting on aging-related processes. Our genome-wide association study of DGRP lines identified associations between single nucleotide polymorphisms in Lissencephaly-1 (Lis-1) and Patronin and early mortality following injury at 20-27 days old. Lis-1 regulates dynein, a microtubule motor required for retrograde transport of many cargoes, and Patronin protects microtubule minus ends against depolymerization. While Patronin mutants did not affect early mortality, Lis-1 compound heterozygotes (Lis-1x/Lis-1y) had increased early mortality following injury at 20-27 or 0-7 days old compared with Lis-1 heterozygotes (Lis-1x/+), and flies that survived 24 h after injury had increased neurodegeneration but an unaltered lifespan, indicating that Lis-1 affects TBI outcomes independently of effects on aging. These data suggest that Lis-1 activity is required in the brain to ameliorate TBI outcomes through effects on axonal transport, microtubule stability, and other microtubule proteins, such as tau, implicated in chronic traumatic encephalopathy, a TBI-associated neurodegenerative disease in humans.
Collapse
Affiliation(s)
- Rebeccah J Katzenberger
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Barry Ganetzky
- Department of Genetics, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David A Wassarman
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
11
|
Abstract
Imaging of mild traumatic brain injury (TBI) using conventional techniques such as CT or MRI often results in no specific imaging correlation that would explain cognitive and clinical symptoms. Molecular imaging of mild TBI suggests that secondary events after injury can be detected using PET. However, no single specific pattern emerges that can aid in diagnosing the injury or determining the prognosis of the long-term behavioral profiles, indicating the heterogeneous and diffuse nature of TBI. Chronic traumatic encephalopathy, a primary tauopathy, has been shown to be strongly associated with repetitive TBI. In vivo data on the available tau PET tracers, however, have produced mixed results and overall low retention profiles in athletes with a history of repetitive mild TBI. Here, we emphasize that the lack of a mechanistic understanding of chronic TBI has posed a challenge when interpreting the results of molecular imaging biomarkers. We advocate for better target identification, improved analysis techniques such as machine learning or artificial intelligence, and novel tracer development.
Collapse
Affiliation(s)
- Gérard N. Bischof
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany;,Institute for Neuroscience and Medicine II–Molecular Organization of the Brain, Research Center Juelich, Juelich, Germany; and
| | - Donna J. Cross
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| |
Collapse
|
12
|
Chen Q, Chen X, Xu L, Zhang R, Li Z, Yue X, Qiao D. Traumatic axonal injury: neuropathological features, postmortem diagnostic methods, and strategies. Forensic Sci Med Pathol 2022; 18:530-544. [PMID: 36117238 DOI: 10.1007/s12024-022-00522-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) has high morbidity and poor prognosis and imposes a serious socioeconomic burden. Traumatic axonal injury (TAI), which is one of the common pathological changes in the primary injury of TBI, is often caused by the external force to the head that causes the white matter bundles to generate shear stress and tension; resulting in tissue damage and leading to the cytoskeletal disorder. At present, the forensic pathological diagnosis of TAI-caused death is still a difficult problem. Most of the TAI biomarkers studied are used for the prediction, evaluation, and prognosis of TAI in the living state. The research subjects are mainly humans in the living state or model animals, which are not suitable for the postmortem diagnosis of TAI. In addition, there is still a lack of recognized indicators for the autopsy pathological diagnosis of TAI. Different diagnostic methods and markers have their limitations, and there is a lack of systematic research and summary of autopsy diagnostic markers of TAI. Therefore, this study mainly summarizes the pathological mechanism, common methods, techniques of postmortem diagnosis, and corresponding biomarkers of TAI, and puts forward the strategies for postmortem diagnosis of TAI for forensic cases with different survival times, which is of great significance to forensic pathological diagnosis.
Collapse
Affiliation(s)
- Qianling Chen
- School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023, Guangzhou, 510515, Guangdong, China
| | - Xuebing Chen
- School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023, Guangzhou, 510515, Guangdong, China
| | - Luyao Xu
- School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023, Guangzhou, 510515, Guangdong, China
| | - Rui Zhang
- School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023, Guangzhou, 510515, Guangdong, China
| | - Zhigang Li
- Guangzhou Forensic Science Institute & Key Laboratory of Forensic Pathology, Ministry of Public Security, Guangzhou, 510442, China.
| | - Xia Yue
- School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023, Guangzhou, 510515, Guangdong, China.
| | - Dongfang Qiao
- School of Forensic Medicine, Southern Medical University, South Shaitai Road #1023, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
13
|
Honig MG, Del Mar NA, Moore BM, Reiner A. Raloxifene Mitigates Emotional Deficits after Mild Traumatic Brain Injury in Mice. Neurotrauma Rep 2022; 3:534-544. [DOI: 10.1089/neur.2022.0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Marcia G. Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Nobel A. Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Bob M. Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
14
|
Joyce JM, La PL, Walker R, Harris A. Magnetic resonance spectroscopy of traumatic brain injury and subconcussive hits: A systematic review and meta-analysis. J Neurotrauma 2022; 39:1455-1476. [PMID: 35838132 DOI: 10.1089/neu.2022.0125] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Magnetic resonance spectroscopy (MRS) is a non-invasive technique used to study metabolites in the brain. MRS findings in traumatic brain injury (TBI) and subconcussive hit literature have been mixed. The most common observation is a decrease in N-acetyl-aspartate (NAA), traditionally considered a marker of neuronal integrity. Other metabolites, however, such as creatine (Cr), choline (Cho), glutamate+glutamine (Glx) and myo-inositol (mI) have shown inconsistent changes in these populations. The objective of this systematic review and meta-analysis was to synthesize MRS literature in head injury and explore factors (brain region, injury severity, time since injury, demographic, technical imaging factors, etc.) that may contribute to differential findings. One hundred and thirty-eight studies met inclusion criteria for the systematic review and of those, 62 NAA, 24 Cr, 49 Cho, 18 Glx and 21 mI studies met inclusion criteria for meta-analysis. A random effects model was used for meta-analyses with brain region as a subgroup for each of the five metabolites studied. Meta-regression was used to examine the influence of potential moderators including injury severity, time since injury, age, sex, tissue composition and methodological factors. In this analysis of 1428 unique head-injured subjects and 1132 controls, the corpus callosum was identified as a brain region highly susceptible to metabolite alteration. NAA was consistently decreased in TBI of all severity, but not in subconcussive hits. Cho and mI were found to be increased in moderate-to-severe TBI but not mild TBI. Glx and Cr were largely unaffected, however did show alterations in certain conditions.
Collapse
Affiliation(s)
- Julie Michele Joyce
- University of Calgary, 2129, Radiology, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, 157742, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, 157744, Calgary, Alberta, Canada.,Integrated Concussion Research Program, Calgary, Alberta, Canada;
| | - Parker L La
- University of Calgary, 2129, Radiology, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, 157742, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, 157744, Calgary, Alberta, Canada.,Integrated Concussion Research Program, Calgary, Alberta, Canada;
| | - Robyn Walker
- University of Calgary, 2129, Radiology, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, 157742, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, 157744, Calgary, Alberta, Canada.,Integrated Concussion Research Program, Calgary, Alberta, Canada;
| | - Ashley Harris
- University of Calgary, Radiology, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, 157742, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, 157744, Calgary, Alberta, Canada.,Integrated Concussion Research Program, Calgary, Alberta, Canada;
| |
Collapse
|
15
|
Harris AC, Jin XT, Greer JE, Povlishock JT, Jacobs KM. Somatostatin interneurons exhibit enhanced functional output and resilience to axotomy after mild traumatic brain injury. Neurobiol Dis 2022; 171:105801. [PMID: 35753625 PMCID: PMC9383472 DOI: 10.1016/j.nbd.2022.105801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/01/2022] Open
Abstract
Mild traumatic brain injury (mTBI) gives rise to a remarkable breadth of pathobiological consequences, principal among which are traumatic axonal injury and perturbation of the functional integrity of neuronal networks that may arise secondary to the elimination of the presynaptic contribution of axotomized neurons. Because there exists a vast diversity of neocortical neuron subtypes, it is imperative to elucidate the relative vulnerability to axotomy among different subtypes. Toward this end, we exploited SOM-IRES-Cre mice to investigate the consequences of the central fluid percussion model of mTBI on the microanatomical integrity and the functional efficacy of the somatostatin (SOM) interneuron population, one of the principal subtypes of neocortical interneuron. We found that the SOM population is resilient to axotomy, representing only 10% of the global burden of inhibitory interneuron axotomy, a result congruous with past work demonstrating that parvalbumin (PV) interneurons bear most of the burden of interneuron axotomy. However, the intact structure of SOM interneurons after injury did not translate to normal cellular function. One day after mTBI, the SOM population is more intrinsically excitable and demonstrates enhanced synaptic efficacy upon post-synaptic layer 5 pyramidal neurons as measured by optogenetics, yet the global evoked inhibitory tone within layer 5 is stable. Simultaneously, there exists a significant increase in the frequency of miniature inhibitory post-synaptic currents within layer 5 pyramidal neurons. These results are consistent with a scheme in which 1 day after mTBI, SOM interneurons are stimulated to compensate for the release from inhibition of layer 5 pyramidal neurons secondary to the disproportionate axotomy of PV interneurons. The enhancement of SOM interneuron intrinsic excitability and synaptic efficacy may represent the initial phase of a dynamic process of attempted autoregulation of neocortical network homeostasis secondary to mTBI.
Collapse
Affiliation(s)
- Alan C Harris
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - Xiao-Tao Jin
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - John E Greer
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - John T Povlishock
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - Kimberle M Jacobs
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| |
Collapse
|
16
|
Secondary Mechanisms of Neurotrauma: A Closer Look at the Evidence. Diseases 2022; 10:diseases10020030. [PMID: 35645251 PMCID: PMC9149951 DOI: 10.3390/diseases10020030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
Traumatic central nervous system injury is a leading cause of neurological injury worldwide. While initial neuroresuscitative efforts are focused on ameliorating the effects of primary injury through patient stabilization, secondary injury in neurotrauma is a potential cause of cell death, oxidative stress, and neuroinflammation. These secondary injuries lack defined therapy. The major causes of secondary injury in neurotrauma include endoplasmic reticular stress, mitochondrial dysfunction, and the buildup of reactive oxygen or nitrogenous species. Stress to the endoplasmic reticulum in neurotrauma results in the overactivation of the unfolded protein response with subsequent cell apoptosis. Mitochondrial dysfunction can lead to the release of caspases and the buildup of reactive oxygen species; several characteristics make the central nervous system particularly susceptible to oxidative damage. Together, endoplasmic reticulum, mitochondrial, and oxidative stress can have detrimental consequences, beginning moments and lasting days to months after the primary injury. Understanding these causative pathways has led to the proposal of various potential treatment options.
Collapse
|
17
|
Radomski KL, Zi X, Lischka FW, Noble MD, Galdzicki Z, Armstrong RC. Acute axon damage and demyelination are mitigated by 4-aminopyridine (4-AP) therapy after experimental traumatic brain injury. Acta Neuropathol Commun 2022; 10:67. [PMID: 35501931 PMCID: PMC9059462 DOI: 10.1186/s40478-022-01366-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
Damage to long axons in white matter tracts is a major pathology in closed head traumatic brain injury (TBI). Acute TBI treatments are needed that protect against axon damage and promote recovery of axon function to prevent long term symptoms and neurodegeneration. Our prior characterization of axon damage and demyelination after TBI led us to examine repurposing of 4-aminopyridine (4-AP), an FDA-approved inhibitor of voltage-gated potassium (Kv) channels. 4-AP is currently indicated to provide symptomatic relief for patients with chronic stage multiple sclerosis, which involves axon damage and demyelination. We tested clinically relevant dosage of 4-AP as an acute treatment for experimental TBI and found multiple benefits in corpus callosum axons. This randomized, controlled pre-clinical study focused on the first week after TBI, when axons are particularly vulnerable. 4-AP treatment initiated one day post-injury dramatically reduced axon damage detected by intra-axonal fluorescence accumulations in Thy1-YFP mice of both sexes. Detailed electron microscopy in C57BL/6 mice showed that 4-AP reduced pathological features of mitochondrial swelling, cytoskeletal disruption, and demyelination at 7 days post-injury. Furthermore, 4-AP improved the molecular organization of axon nodal regions by restoring disrupted paranode domains and reducing Kv1.2 channel dispersion. 4-AP treatment did not resolve deficits in action potential conduction across the corpus callosum, based on ex vivo electrophysiological recordings at 7 days post-TBI. Thus, this first study of 4-AP effects on axon damage in the acute period demonstrates a significant decrease in multiple pathological hallmarks of axon damage after experimental TBI.
Collapse
Affiliation(s)
- Kryslaine L. Radomski
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Xiaomei Zi
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Fritz W. Lischka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Mark D. Noble
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave, Box 633, Rochester, NY 14642 USA
| | - Zygmunt Galdzicki
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Regina C. Armstrong
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| |
Collapse
|
18
|
Honig MG, Del Mar NA, Henderson DL, O'Neal D, Yammanur M, Cox R, Li C, Perry AM, Moore BM, Reiner A. Raloxifene, a cannabinoid type-2 receptor inverse agonist, mitigates visual deficits and pathology and modulates microglia after ocular blast. Exp Eye Res 2022; 218:108966. [PMID: 35143834 DOI: 10.1016/j.exer.2022.108966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/04/2022] [Accepted: 01/24/2022] [Indexed: 11/19/2022]
Abstract
Visual deficits after ocular blast injury (OBI) are common, but pharmacological approaches to improve long-term outcomes have not been identified. Blast forces frequently damage the retina and optic nerves, and work on experimental animals has shown the pro-inflammatory actions of microglia can further exacerbate such injuries. Cannabinoid type-2 receptor (CB2) inverse agonists specifically target activated microglia, biasing them away from the harmful pro-inflammatory M1 state toward the helpful reparative M2 state. We previously found that treating mice with CB2 inverse agonists after traumatic brain injury, produced by either focal cranial air blast or dorsal cranial impact, greatly attenuated the visual deficits and pathology that otherwise resulted. Here we examined the consequences of single and repeat OBI and the benefit provided by raloxifene, an FDA-approved estrogen receptor drug that possesses noteworthy CB2 inverse agonism. After single OBI, although the amplitudes of the A- and B-waves of the electroretinogram and pupil light response appeared to be normal, the mice showed hints of deficits in contrast sensitivity and visual acuity, a trend toward optic nerve axon loss, and significantly increased light aversion, which were reversed by 2 weeks of daily treatment with raloxifene. Mice subjected to repeat OBI (5 blasts spaced 1 min apart), exhibited more severe visual deficits, including decreases in contrast sensitivity, visual acuity, the amplitudes of the A- and B-waves of the electroretinogram, light aversion, and resting pupil diameter (i.e. hyperconstriction), accompanied by the loss of photoreceptor cells and optic nerve axons, nearly all of which were mitigated by raloxifene. Interestingly, optic nerve axon abundance was strongly correlated with contrast sensitivity and visual acuity across all groups of experimental mice in the repeat OBI study, suggesting optic nerve axon loss with rOBI and its attenuation with raloxifene are associated with the extent of these two deficits while photoreceptor abundance was highly correlated with A-wave amplitude and resting pupil size, suggesting a prominent role for photoreceptors in these two deficits. Quantitative PCR (qPCR) showed levels of M1-type microglial markers (e.g. iNOS, IL1β, TNFα, and CD32) in retina, optic nerve, and thalamus were increased 3 days after repeat OBI. With raloxifene treatment, the overall expression of M1 markers was more similar to that in sham mice. Raloxifene treatment was also associated with the elevation of IL10 transcripts in all three tissues compared to repeat OBI alone, but the results for the three other M2 microglial markers we examined were more varied. Taken together, the qPCR results suggest that raloxifene benefit for visual function and pathology was associated with a lessening of the pro-inflammatory actions of microglia. The benefit we find for raloxifene following OBI provides a strong basis for phase-2 efficacy testing in human clinical trials for treating ocular injury.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Desmond L Henderson
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Dylan O'Neal
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Meghna Yammanur
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Rachel Cox
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Chunyan Li
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Aaron M Perry
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Bob M Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA; Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
19
|
Ogino Y, Bernas T, Greer JE, Povlishock JT. Axonal injury following mild traumatic brain injury is exacerbated by repetitive insult and is linked to the delayed attenuation of NeuN expression without concomitant neuronal death in the mouse. Brain Pathol 2021; 32:e13034. [PMID: 34729854 PMCID: PMC8877729 DOI: 10.1111/bpa.13034] [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: 06/06/2021] [Revised: 09/06/2021] [Accepted: 10/14/2021] [Indexed: 11/30/2022] Open
Abstract
Mild traumatic brain injury (mTBI) affects brain structure and function and can lead to persistent abnormalities. Repetitive mTBI exacerbates the acute phase response to injury. Nonetheless, its long‐term implications remain poorly understood, particularly in the context of traumatic axonal injury (TAI), a player in TBI morbidity via axonal disconnection, synaptic loss and retrograde neuronal perturbation. In contrast to the examination of these processes in the acute phase of injury, the chronic‐phase burden of TAI and/or its implications for retrograde neuronal perturbation or death have received little consideration. To critically assess this issue, murine neocortical tissue was investigated at acute (24‐h postinjury, 24hpi) and chronic time points (28‐days postinjury, 28dpi) after singular or repetitive mTBI induced by central fluid percussion injury (cFPI). Neurons were immunofluorescently labeled for NeuroTrace and NeuN (all neurons), p‐c‐Jun (axotomized neurons) and DRAQ5 (cell nuclei), imaged in 3D and quantified in automated manner. Single mTBI produced axotomy in 10% of neurons at 24hpi and the percentage increased after repetitive injury. The fraction of p‐c‐Jun+ neurons decreased at 28dpi but without neuronal loss (NeuroTrace), suggesting their reorganization and/or repair following TAI. In contrast, NeuN+ neurons decreased with repetitive injury at 24hpi while the corresponding fraction of NeuroTrace+ neurons decreased over 28dpi. Attenuated NeuN expression was linked exclusively to non‐axotomized neurons at 24hpi which extended to the axotomized at 28dpi, revealing a delayed response of the axotomized neurons. Collectively, we demonstrate an increased burden of TAI after repetitive mTBI, which is most striking in the acute phase response to the injury. Our finding of widespread axotomy in large fields of intact neurons contradicts the notion that repetitive mTBI elicits progressive neuronal death, rather, emphasizing the importance of axotomy‐mediated change.
Collapse
Affiliation(s)
- Yasuaki Ogino
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Tytus Bernas
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - John E Greer
- Department of Neurosurgery, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.,Department of Surgery, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, Virginia, USA
| | - John T Povlishock
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
20
|
Mi Z, Liu H, Rose ME, Ma J, Reay DP, Ma X, Henchir JJ, Dixon CE, Graham SH. Mutation of a Ubiquitin Carboxy Terminal Hydrolase L1 Lipid Binding Site Alleviates Cell Death, Axonal Injury, and Behavioral Deficits After Traumatic Brain Injury in Mice. Neuroscience 2021; 475:127-136. [PMID: 34508847 DOI: 10.1016/j.neuroscience.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022]
Abstract
Ubiquitin carboxy terminal hydrolase L1 (UCHL1) is a protein highly expressed in neurons that may play important roles in the ubiquitin proteasome pathway (UPP) in neurons, axonal integrity, and motor function after traumatic brain injury (TBI). Binding of reactive lipid species to cysteine 152 of UCHL1 results in unfolding, aggregation, and inactivation of the enzyme. To test the role of this mechanism in TBI, mice bearing a cysteine to alanine mutation at site 152 (C152A mice) that renders UCHL1 resistant to inactivation by reactive lipids were subjected to the controlled cortical impact model (CCI) of TBI and compared to wild type (WT) controls. Alterations in protein ubiquitination and activation of autophagy pathway markers in traumatized brain were detected by immunoblotting. Cell death and axonal injury were determined by histological assessment and anti-amyloid precursor protein (APP) immunohistochemistry. Behavioral outcomes were determined using the beam balance and Morris water maze tests. C152A mice had reduced accumulation of ubiquitinated proteins, decreased activation of the autophagy markers Beclin-1 and LC3B, a decreased number of abnormal axons, decreased CA1 cell death, and improved motor and cognitive function compared to WT controls after CCI; no significant change in spared tissue volume was observed. These results suggest that binding of lipid substrates to cysteine 152 of UCHL1 is important in the pathogenesis of injury and recovery after TBI and may be a novel target for future therapeutic approaches.
Collapse
Affiliation(s)
- Zhiping Mi
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Hao Liu
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA
| | - Marie E Rose
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Jie Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Daniel P Reay
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Xiecheng Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Jeremy J Henchir
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | - C Edward Dixon
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | - Steven H Graham
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
21
|
Vijayakumari AA, Parker D, Osmanlioglu Y, Alappatt JA, Whyte J, Diaz-Arrastia R, Kim JJ, Verma R. Free Water Volume Fraction: An Imaging Biomarker to Characterize Moderate-to-Severe Traumatic Brain Injury. J Neurotrauma 2021; 38:2698-2705. [PMID: 33913750 PMCID: PMC8590145 DOI: 10.1089/neu.2021.0057] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) is a major clinical and public health problem with few therapeutic interventions successfully translated to the clinic. Identifying imaging-based biomarkers characterizing injury severity and predicting long-term functional and cognitive outcomes in TBI patients is crucial for treatment. TBI results in white matter (WM) injuries, which can be detected using diffusion tensor imaging (DTI). Trauma-induced pathologies lead to accumulation of free water (FW) in brain tissue, and standard DTI is susceptible to the confounding effects of FW. In this study, we applied FW DTI to estimate free water volume fraction (FW-VF) in patients with moderate-to-severe TBI and demonstrated its association with injury severity and long-term outcomes. DTI scans and neuropsychological assessments were obtained longitudinally at 3, 6, and 12 months post-injury for 34 patients and once in 35 matched healthy controls. We observed significantly elevated FW-VF in 85 of 90 WM regions in patients compared to healthy controls (p < 0.05). We then presented a patient-specific summary score of WM regions derived using Mahalanobis distance. We observed that MVF at 3 months significantly predicted functional outcome (p = 0.008), executive function (p = 0.005), and processing speed (p = 0.01) measured at 12 months and was significantly correlated with injury severity (p < 0.001). Our findings are an important step toward implementing MVF as a biomarker for personalized therapy and rehabilitation planning for TBI patients.
Collapse
Affiliation(s)
- Anupa Ambili Vijayakumari
- DiCIPHR (Diffusion and Connectomics in Precision Healthcare Research) Lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Drew Parker
- DiCIPHR (Diffusion and Connectomics in Precision Healthcare Research) Lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yusuf Osmanlioglu
- DiCIPHR (Diffusion and Connectomics in Precision Healthcare Research) Lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jacob A. Alappatt
- DiCIPHR (Diffusion and Connectomics in Precision Healthcare Research) Lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Whyte
- Moss Rehabilitation Research Institute, TBI Rehabilitation Research Laboratory, Einstein Medical Center Elkins Park, Philadelphia, Pennsylvania, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Junghoon J. Kim
- Department of Molecular, Cellular, and Biomedical Sciences, CUNY School of Medicine, The City College of New York, New York, New York, USA
| | - Ragini Verma
- DiCIPHR (Diffusion and Connectomics in Precision Healthcare Research) Lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Smith DH, Kochanek PM, Rosi S, Meyer R, Ferland-Beckham C, Prager EM, Ahlers ST, Crawford F. Roadmap for Advancing Pre-Clinical Science in Traumatic Brain Injury. J Neurotrauma 2021; 38:3204-3221. [PMID: 34210174 PMCID: PMC8820284 DOI: 10.1089/neu.2021.0094] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pre-clinical models of disease have long played important roles in the advancement of new treatments. However, in traumatic brain injury (TBI), despite the availability of numerous model systems, translation from bench to bedside remains elusive. Integrating clinical relevance into pre-clinical model development is a critical step toward advancing therapies for TBI patients across the spectrum of injury severity. Pre-clinical models include in vivo and ex vivo animal work-both small and large-and in vitro modeling. The wide range of pre-clinical models reflect substantial attempts to replicate multiple aspects of TBI sequelae in humans. Although these models reveal multiple putative mechanisms underlying TBI pathophysiology, failures to translate these findings into successful clinical trials call into question the clinical relevance and applicability of the models. Here, we address the promises and pitfalls of pre-clinical models with the goal of evolving frameworks that will advance translational TBI research across models, injury types, and the heterogenous etiology of pathology.
Collapse
Affiliation(s)
- Douglas H Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine; Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Rangos Research Center, Pittsburgh, Pennsylvania, USA
| | - Susanna Rosi
- Departments of Physical Therapy Rehabilitation Science, Neurological Surgery, Weill Institute for Neuroscience, University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Retsina Meyer
- Cohen Veterans Bioscience, New York, New York, USA.,Delix Therapeutics, Inc, Boston, Massachusetts, USA
| | | | | | - Stephen T Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate Naval Medical Research Center, Silver Spring, Maryland, USA
| | | |
Collapse
|
23
|
Progressive Neurodegeneration Across Chronic Stages of Severe Traumatic Brain Injury. J Head Trauma Rehabil 2021; 37:E144-E156. [PMID: 34145157 DOI: 10.1097/htr.0000000000000696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To examine the trajectory of structural gray matter changes across 2 chronic periods of recovery in individuals who have sustained severe traumatic brain injury (TBI), adding to the growing literature indicating that neurodegenerative processes occur in the months to years postinjury. PARTICIPANTS Patients who experienced posttraumatic amnesia of 1 hour or more, and/or scored 12 or less on the Glasgow Coma Scale at the emergency department or the scene of the accident, and/or had positive brain imaging findings were recruited while receiving inpatient care, resulting in 51 patients with severe TBI. METHODS Secondary analyses of gray matter changes across approximately 5 months, 1 year, and 2.5 years postinjury were undertaken, using an automated segmentation protocol with improved accuracy in populations with morphological anomalies. We compared patients and matched controls on regions implicated in poorer long-term clinical outcome (accumbens, amygdala, brainstem, hippocampus, thalamus). To model brain-wide patterns of change, we then conducted an exploratory principal component analysis (PCA) on the linear slopes of all regional volumes across the 3 time points. Finally, we assessed nonlinear trends across earlier (5 months-1 year) versus later (1-2.5 years) time-windows with PCA to compare degeneration rates across time. Chronic degeneration was predicted cortically and subcortically brain-wide, and within specific regions of interest. RESULTS (1) From 5 months to 1 year, patients showed significant degeneration in the accumbens, and marginal degeneration in the amygdala, brainstem, thalamus, and the left hippocampus when examined unilaterally, compared with controls. (2) PCA components representing subcortical and temporal regions, and regions from the basal ganglia, significantly differed from controls in the first time-window. (3) Progression occurred at the same rate across both time-windows, suggesting neither escalation nor attenuation of degeneration across time. CONCLUSION Localized yet progressive decline emphasizes the necessity of developing interventions to offset degeneration and improve long-term functioning.
Collapse
|
24
|
Edlow BL, Claassen J, Schiff ND, Greer DM. Recovery from disorders of consciousness: mechanisms, prognosis and emerging therapies. Nat Rev Neurol 2021; 17:135-156. [PMID: 33318675 PMCID: PMC7734616 DOI: 10.1038/s41582-020-00428-x] [Citation(s) in RCA: 291] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
Substantial progress has been made over the past two decades in detecting, predicting and promoting recovery of consciousness in patients with disorders of consciousness (DoC) caused by severe brain injuries. Advanced neuroimaging and electrophysiological techniques have revealed new insights into the biological mechanisms underlying recovery of consciousness and have enabled the identification of preserved brain networks in patients who seem unresponsive, thus raising hope for more accurate diagnosis and prognosis. Emerging evidence suggests that covert consciousness, or cognitive motor dissociation (CMD), is present in up to 15-20% of patients with DoC and that detection of CMD in the intensive care unit can predict functional recovery at 1 year post injury. Although fundamental questions remain about which patients with DoC have the potential for recovery, novel pharmacological and electrophysiological therapies have shown the potential to reactivate injured neural networks and promote re-emergence of consciousness. In this Review, we focus on mechanisms of recovery from DoC in the acute and subacute-to-chronic stages, and we discuss recent progress in detecting and predicting recovery of consciousness. We also describe the developments in pharmacological and electrophysiological therapies that are creating new opportunities to improve the lives of patients with DoC.
Collapse
Affiliation(s)
- Brian L Edlow
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jan Claassen
- Department of Neurology, Columbia University Medical Center, New York Presbyterian Hospital, New York, NY, USA
| | - Nicholas D Schiff
- Feil Family Brain Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - David M Greer
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
25
|
Jamjoom AAB, Rhodes J, Andrews PJD, Grant SGN. The synapse in traumatic brain injury. Brain 2021; 144:18-31. [PMID: 33186462 PMCID: PMC7880663 DOI: 10.1093/brain/awaa321] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide and is a risk factor for dementia later in life. Research into the pathophysiology of TBI has focused on the impact of injury on the neuron. However, recent advances have shown that TBI has a major impact on synapse structure and function through a combination of the immediate mechanical insult and the ensuing secondary injury processes, leading to synapse loss. In this review, we highlight the role of the synapse in TBI pathophysiology with a focus on the confluence of multiple secondary injury processes including excitotoxicity, inflammation and oxidative stress. The primary insult triggers a cascade of events in each of these secondary processes and we discuss the complex interplay that occurs at the synapse. We also examine how the synapse is impacted by traumatic axonal injury and the role it may play in the spread of tau after TBI. We propose that astrocytes play a crucial role by mediating both synapse loss and recovery. Finally, we highlight recent developments in the field including synapse molecular imaging, fluid biomarkers and therapeutics. In particular, we discuss advances in our understanding of synapse diversity and suggest that the new technology of synaptome mapping may prove useful in identifying synapses that are vulnerable or resistant to TBI.
Collapse
Affiliation(s)
- Aimun A B Jamjoom
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Jonathan Rhodes
- Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh EH16 4SA, UK
| | - Peter J D Andrews
- Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh EH16 4SA, UK
| | - Seth G N Grant
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
26
|
Belchev Z, Boulos ME, Rybkina J, Johns K, Jeffay E, Colella B, Ozubko J, Bray MJC, Di Genova N, Levi A, Changoor A, Worthington T, Gilboa A, Green R. Remotely delivered environmental enrichment intervention for traumatic brain injury: Study protocol for a randomised controlled trial. BMJ Open 2021; 11:e039767. [PMID: 33574141 PMCID: PMC7880099 DOI: 10.1136/bmjopen-2020-039767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/24/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Individuals with moderate-severe traumatic brain injury (m-sTBI) experience progressive brain and behavioural declines in the chronic stages of injury. Longitudinal studies found that a majority of patients with m-sTBI exhibit significant hippocampal atrophy from 5 to 12 months post-injury, associated with decreased cognitive environmental enrichment (EE). Encouragingly, engaging in EE has been shown to lead to neural improvements, suggesting it is a promising avenue for offsetting hippocampal neurodegeneration in m-sTBI. Allocentric spatial navigation (ie, flexible, bird's eye view approach), is a good candidate for EE in m-sTBI because it is associated with hippocampal activation and reduced ageing-related volume loss. Efficacy of EE requires intensive daily training, prohibitive within most current health delivery systems. The present protocol is a novel, remotely delivered and self-administered intervention designed to harness principles from EE and allocentric spatial navigation to offset hippocampal atrophy and potentially improve hippocampal functions such as navigation and memory for patients with m-sTBI. METHODS AND ANALYSIS Eighty-four participants with chronic m-sTBI are being recruited from an urban rehabilitation hospital and randomised into a 16-week intervention (5 hours/week; total: 80 hours) of either targeted spatial navigation or an active control group. The spatial navigation group engages in structured exploration of different cities using Google Street View that includes daily navigation challenges. The active control group watches and answers subjective questions about educational videos. Following a brief orientation, participants remotely self-administer the intervention on their home computer. In addition to feasibility and compliance measures, clinical and experimental cognitive measures as well as MRI scan data are collected pre-intervention and post-intervention to determine behavioural and neural efficacy. ETHICS AND DISSEMINATION Ethics approval has been obtained from ethics boards at the University Health Network and University of Toronto. Findings will be presented at academic conferences and submitted to peer-reviewed journals. TRIAL REGISTRATION NUMBER Version 3, ClinicalTrials.gov Registry (NCT04331392).
Collapse
Affiliation(s)
- Zorry Belchev
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Rotman Research Institute at Baycrest, Toronto, Ontario, Canada
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Mary Ellene Boulos
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
- Graduate Department of Rehabilitation Science, University of Toronto, Toronto, Ontario, Canada
| | - Julia Rybkina
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
- Graduate Department of Rehabilitation Science, University of Toronto, Toronto, Ontario, Canada
| | - Kadeen Johns
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Eliyas Jeffay
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Rotman Research Institute at Baycrest, Toronto, Ontario, Canada
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Brenda Colella
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Jason Ozubko
- Department of Psychology, The State University of New York, Geneseo, New York, USA
| | - Michael Johnathan Charles Bray
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
- Graduate Department of Rehabilitation Science, University of Toronto, Toronto, Ontario, Canada
| | - Nicholas Di Genova
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
- Department of Computing and Software, McMaster University, Hamilton, Ontario, Canada
| | - Adina Levi
- Rotman Research Institute at Baycrest, Toronto, Ontario, Canada
- Department of Psychology, York University, Toronto, Ontario, Canada
| | - Alana Changoor
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
- Global Health Program, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Thomas Worthington
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
- Department of Psychology, York University, Toronto, Ontario, Canada
| | - Asaf Gilboa
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Rotman Research Institute at Baycrest, Toronto, Ontario, Canada
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Robin Green
- KITE, Toronto Rehabilitation Institute, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Abu Hamdeh S, Ciuculete DM, Sarkisyan D, Bakalkin G, Ingelsson M, Schiöth HB, Marklund N. Differential DNA Methylation of the Genes for Amyloid Precursor Protein, Tau, and Neurofilaments in Human Traumatic Brain Injury. J Neurotrauma 2021; 38:1679-1688. [PMID: 33191850 DOI: 10.1089/neu.2020.7283] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is an established risk factor for neurodegenerative disorders and dementias. Epigenetic modifications, such as DNA methylation, may alter the expression of genes without altering the DNA sequence in response to environmental factors. We hypothesized that DNA methylation changes may occur in the injured human brain and be implicated in the neurodegenerative aftermath of TBI. The DNA methylation status of genes related to neurodegeneration; for example, amyloid beta precursor protein (APP), microtubule associated protein tau (MAPT), neurofilament heavy (NEFH), neurofilament medium (NEFM), and neurofilament light (NEFL), was analyzed in fresh, surgically resected human brain tissue from 17 severe TBI patients and compared with brain biopsy samples from 19 patients with idiopathic normal pressure hydrocephalus (iNPH). We also performed an epigenome-wide association study (EWAS) comparing TBI patients with iNPH controls. Thirty-eight CpG sites in the APP, MAPT, NEFH, and NEFL genes were differentially methylated by TBI. Among the top 20 differentially methylated CpG sites, 11 were in the APP gene. In addition, the EWAS evaluating 828,888 CpG sites revealed 308 differentially methylated CpG sites in genes related to cellular/anatomical structure development, cell differentiation, and anatomical morphogenesis. These preliminary findings provide the first evidence of an altered DNA methylome in the injured human brain, and may have implications for the neurodegenerative disorders associated with TBI.
Collapse
Affiliation(s)
- Sami Abu Hamdeh
- Department of Neuroscience, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Diana-Maria Ciuculete
- Division of Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Daniil Sarkisyan
- Department of Pharmaceutical Biosciences, and Uppsala University, Uppsala, Sweden
| | - Georgy Bakalkin
- Department of Pharmaceutical Biosciences, and Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Division of Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden.,Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Niklas Marklund
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
28
|
The Protective Role of Nutraceuticals in Critically Ill Patients with Traumatic Brain Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1328:243-253. [DOI: 10.1007/978-3-030-73234-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
29
|
Bruggeman GF, Haitsma IK, Dirven CMF, Volovici V. Traumatic axonal injury (TAI): definitions, pathophysiology and imaging-a narrative review. Acta Neurochir (Wien) 2021; 163:31-44. [PMID: 33006648 PMCID: PMC7778615 DOI: 10.1007/s00701-020-04594-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/22/2020] [Indexed: 01/01/2023]
Abstract
Introduction Traumatic axonal injury (TAI) is a condition defined as multiple, scattered, small hemorrhagic, and/or non-hemorrhagic lesions, alongside brain swelling, in a more confined white matter distribution on imaging studies, together with impaired axoplasmic transport, axonal swelling, and disconnection after traumatic brain injury (TBI). Ever since its description in the 1980s and the grading system by Adams et al., our understanding of the processes behind this entity has increased. Methods We performed a scoping systematic, narrative review by interrogating Ovid MEDLINE, Embase, and Google Scholar on the pathophysiology, biomarkers, and diagnostic tools of TAI patients until July 2020. Results We underline the misuse of the Adams classification on MRI without proper validation studies, and highlight the hiatus in the scientific literature and areas needing more research. In the past, the theory behind the pathophysiology relied on the inertial force exerted on the brain matter after severe TBI inducing a primary axotomy. This theory has now been partially abandoned in favor of a more refined theory involving biochemical processes such as protein cleavage and DNA breakdown, ultimately leading to an inflammation cascade and cell apoptosis, a process now described as secondary axotomy. Conclusion The difference in TAI definitions makes the comparison of studies that report outcomes, treatments, and prognostic factors a daunting task. An even more difficult task is isolating the outcomes of isolated TAI from the outcomes of severe TBI in general. Targeted bench-to-bedside studies are required in order to uncover further pathways involved in the pathophysiology of TAI and, ideally, new treatments.
Collapse
Affiliation(s)
- Gavin F Bruggeman
- Department of Neurosurgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Iain K Haitsma
- Department of Neurosurgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Clemens M F Dirven
- Department of Neurosurgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Victor Volovici
- Department of Neurosurgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
30
|
Clark J, Zhu Z, Chuckowree J, Dickson T, Blizzard C. Efficacy of epothilones in central nervous system trauma treatment: what has age got to do with it? Neural Regen Res 2021; 16:618-620. [PMID: 33063710 PMCID: PMC8067923 DOI: 10.4103/1673-5374.295312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Central nervous system injury, specifically traumatic brain and spinal cord injury, can have significant long lasting effects. There are no comprehensive treatments to combat the injury and sequalae of events that occurring following a central nervous system trauma. Herein we discuss the potential for the epothilone family of microtubule stabilizing agents to improve outcomes following experimentally induced trauma. These drugs, which are able to cross the blood-brain barrier, may hold great promise for the treatment of central nervous system trauma and the current literature presents the extensive range of beneficial effects these drugs may have following trauma in animal models. Importantly, the effect of the epothilones can vary and our most recent contributions to this field indicate that the efficacy of epothilones following traumatic brain injury is dependent upon the age of the animals. Therefore, we present a case for a greater emphasis to be placed upon age when using an intervention aimed at neural regeneration and highlight the importance of tailoring the therapeutic regime in the clinic to the age of the patient to promote improved patient outcomes.
Collapse
Affiliation(s)
- Jayden Clark
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Zhendan Zhu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Jyoti Chuckowree
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Tracey Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Catherine Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| |
Collapse
|
31
|
Mohammadian M, Roine T, Hirvonen J, Kurki T, Posti JP, Katila AJ, Takala RSK, Tallus J, Maanpää HR, Frantzén J, Hutchinson PJ, Newcombe VF, Menon DK, Tenovuo O. Alterations in Microstructure and Local Fiber Orientation of White Matter Are Associated with Outcome after Mild Traumatic Brain Injury. J Neurotrauma 2020; 37:2616-2623. [PMID: 32689872 DOI: 10.1089/neu.2020.7081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Mild traumatic brain injury (mTBI) can have long-lasting consequences. We investigated white matter (WM) alterations at 6-12 months following mTBI using diffusion tensor imaging (DTI) and assessed if the alterations associate with outcome. Eighty-five patients with mTBI underwent diffusion-weighted magnetic resonance imaging (MRI) on average 8 months post-injury and patients' outcome was assessed at the time of imaging using the Glasgow Outcome Scale-Extended (GOS-E). Additionally, 30 age-matched patients with extracranial orthopedic injuries were used as control subjects. Voxel-wise analysis of the data was performed using a tract-based spatial statistics (TBSS) approach and differences in microstructural metrics between groups were investigated. Further, the susceptibility of the abnormalities to specific fiber orientations was investigated by analyzing the first eigenvector of the diffusion tensor in the voxels with significant differences. We found significantly lower fractional anisotropy (FA) and higher mean diffusivity (MD) and radial diffusivity (RD) in patients with mTBI compared with control subjects, whereas no significant differences were observed in axial diffusivity (AD) between the groups. The differences were present bilaterally in several WM regions and correlated with outcome. Moreover, multiple clusters were found in the principal fiber orientations of the significant voxels in anisotropy, and similar orientation patterns were found for the diffusivity metrics. These directional clusters correlated with patients' functional outcome. Our study showed that mTBI is associated with WM changes at the chronic stage and these alterations occur in several WM regions. In addition, several significant clusters of WM alterations in specific fiber orientations were found and these clusters were associated with outcome.
Collapse
Affiliation(s)
- Mehrbod Mohammadian
- Department of Clinical Neurosciences, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland.,Turku Brain Injury Center, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland
| | - Timo Roine
- Turku Brain and Mind Center, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland.,Department of Neuroscience and Biomedical Engineering, Aalto University School of Science, Espoo, Finland
| | - Jussi Hirvonen
- Department of Radiology, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland
| | - Timo Kurki
- Department of Clinical Neurosciences, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland.,Turku Brain Injury Center, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland.,Department of Radiology, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland
| | - Jussi P Posti
- Department of Clinical Neurosciences, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland.,Turku Brain Injury Center, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland.,Department of Neurosurgery, Division of Clinical Neurosciences, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland
| | - Ari J Katila
- Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland.,Anesthesiology, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland
| | - Riikka S K Takala
- Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland.,Anesthesiology, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland
| | - Jussi Tallus
- Department of Clinical Neurosciences, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland.,Turku Brain Injury Center, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland
| | - Henna-Riikka Maanpää
- Turku Brain Injury Center, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland.,Department of Neurosurgery, Division of Clinical Neurosciences, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland
| | - Janek Frantzén
- Department of Clinical Neurosciences, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland.,Department of Neurosurgery, Division of Clinical Neurosciences, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland
| | - Peter J Hutchinson
- Department of Clinical Neurosciences, Neurosurgery Unit, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | - David K Menon
- Division of Anesthesia, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Olli Tenovuo
- Department of Clinical Neurosciences, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland.,Turku Brain Injury Center, Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland
| |
Collapse
|
32
|
Kochanek PM, Jackson TC, Jha RM, Clark RS, Okonkwo DO, Bayır H, Poloyac SM, Wagner AK, Empey PE, Conley YP, Bell MJ, Kline AE, Bondi CO, Simon DW, Carlson SW, Puccio AM, Horvat CM, Au AK, Elmer J, Treble-Barna A, Ikonomovic MD, Shutter LA, Taylor DL, Stern AM, Graham SH, Kagan VE, Jackson EK, Wisniewski SR, Dixon CE. Paths to Successful Translation of New Therapies for Severe Traumatic Brain Injury in the Golden Age of Traumatic Brain Injury Research: A Pittsburgh Vision. J Neurotrauma 2020; 37:2353-2371. [PMID: 30520681 PMCID: PMC7698994 DOI: 10.1089/neu.2018.6203] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
New neuroprotective therapies for severe traumatic brain injury (TBI) have not translated from pre-clinical to clinical success. Numerous explanations have been suggested in both the pre-clinical and clinical arenas. Coverage of TBI in the lay press has reinvigorated interest, creating a golden age of TBI research with innovative strategies to circumvent roadblocks. We discuss the need for more robust therapies. We present concepts for traditional and novel approaches to defining therapeutic targets. We review lessons learned from the ongoing work of the pre-clinical drug and biomarker screening consortium Operation Brain Trauma Therapy and suggest ways to further enhance pre-clinical consortia. Biomarkers have emerged that empower choice and assessment of target engagement by candidate therapies. Drug combinations may be needed, and it may require moving beyond conventional drug therapies. Precision medicine may also link the right therapy to the right patient, including new approaches to TBI classification beyond the Glasgow Coma Scale or anatomical phenotyping-incorporating new genetic and physiologic approaches. Therapeutic breakthroughs may also come from alternative approaches in clinical investigation (comparative effectiveness, adaptive trial design, use of the electronic medical record, and big data). The full continuum of care must also be represented in translational studies, given the important clinical role of pre-hospital events, extracerebral insults in the intensive care unit, and rehabilitation. TBI research from concussion to coma can cross-pollinate and further advancement of new therapies. Misconceptions can stifle/misdirect TBI research and deserve special attention. Finally, we synthesize an approach to deliver therapeutic breakthroughs in this golden age of TBI research.
Collapse
Affiliation(s)
- Patrick M. Kochanek
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Travis C. Jackson
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ruchira M. Jha
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert S.B. Clark
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - David O. Okonkwo
- Department of Neurological Surgery, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania, USA
| | - Hülya Bayır
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Samuel M. Poloyac
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Amy K. Wagner
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Philip E. Empey
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Yvette P. Conley
- Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, Pennsylvania, USA
| | - Michael J. Bell
- Department of Critical Care Medicine, Children's National Medical Center, Washington, DC, USA
| | - Anthony E. Kline
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Corina O. Bondi
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dennis W. Simon
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shaun W. Carlson
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ava M. Puccio
- Department of Neurological Surgery, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania, USA
| | - Christopher M. Horvat
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Alicia K. Au
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jonathan Elmer
- Departments of Emergency Medicine and Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania, USA
| | - Amery Treble-Barna
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Milos D. Ikonomovic
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lori A. Shutter
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - D. Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew M. Stern
- Drug Discovery Institute, Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Steven H. Graham
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Valerian E. Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen R. Wisniewski
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - C. Edward Dixon
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
33
|
Beard K, Meaney DF, Issadore D. Clinical Applications of Extracellular Vesicles in the Diagnosis and Treatment of Traumatic Brain Injury. J Neurotrauma 2020; 37:2045-2056. [PMID: 32312151 PMCID: PMC7502684 DOI: 10.1089/neu.2020.6990] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) have emerged as key mediators of cell-cell communication during homeostasis and in pathology. Central nervous system (CNS)-derived EVs contain cell type-specific surface markers and intralumenal protein, RNA, DNA, and metabolite cargo that can be used to assess the biochemical and molecular state of neurons and glia during neurological injury and disease. The development of EV isolation strategies coupled with analysis of multi-plexed biomarker and clinical data have the potential to improve our ability to classify and treat traumatic brain injury (TBI) and resulting sequelae. Additionally, their ability to cross the blood-brain barrier (BBB) has implications for both EV-based diagnostic strategies and for potential EV-based therapeutics. In the present review, we discuss encouraging data for EV-based diagnostic, prognostic, and therapeutic strategies in the context of TBI monitoring and management.
Collapse
Affiliation(s)
- Kryshawna Beard
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David F. Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David Issadore
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
Edlow BL, Barra ME, Zhou DW, Foulkes AS, Snider SB, Threlkeld ZD, Chakravarty S, Kirsch JE, Chan ST, Meisler SL, Bleck TP, Fins JJ, Giacino JT, Hochberg LR, Solt K, Brown EN, Bodien YG. Personalized Connectome Mapping to Guide Targeted Therapy and Promote Recovery of Consciousness in the Intensive Care Unit. Neurocrit Care 2020; 33:364-375. [PMID: 32794142 PMCID: PMC8336723 DOI: 10.1007/s12028-020-01062-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/18/2020] [Indexed: 01/05/2023]
Abstract
There are currently no therapies proven to promote early recovery of consciousness in patients with severe brain injuries in the intensive care unit (ICU). For patients whose families face time-sensitive, life-or-death decisions, treatments that promote recovery of consciousness are needed to reduce the likelihood of premature withdrawal of life-sustaining therapy, facilitate autonomous self-expression, and increase access to rehabilitative care. Here, we present the Connectome-based Clinical Trial Platform (CCTP), a new paradigm for developing and testing targeted therapies that promote early recovery of consciousness in the ICU. We report the protocol for STIMPACT (Stimulant Therapy Targeted to Individualized Connectivity Maps to Promote ReACTivation of Consciousness), a CCTP-based trial in which intravenous methylphenidate will be used for targeted stimulation of dopaminergic circuits within the subcortical ascending arousal network (ClinicalTrials.gov NCT03814356). The scientific premise of the CCTP and the STIMPACT trial is that personalized brain network mapping in the ICU can identify patients whose connectomes are amenable to neuromodulation. Phase 1 of the STIMPACT trial is an open-label, safety and dose-finding study in 22 patients with disorders of consciousness caused by acute severe traumatic brain injury. Patients in Phase 1 will receive escalating daily doses (0.5-2.0 mg/kg) of intravenous methylphenidate over a 4-day period and will undergo resting-state functional magnetic resonance imaging and electroencephalography to evaluate the drug's pharmacodynamic properties. The primary outcome measure for Phase 1 relates to safety: the number of drug-related adverse events at each dose. Secondary outcome measures pertain to pharmacokinetics and pharmacodynamics: (1) time to maximal serum concentration; (2) serum half-life; (3) effect of the highest tolerated dose on resting-state functional MRI biomarkers of connectivity; and (4) effect of each dose on EEG biomarkers of cerebral cortical function. Predetermined safety and pharmacodynamic criteria must be fulfilled in Phase 1 to proceed to Phase 2A. Pharmacokinetic data from Phase 1 will also inform the study design of Phase 2A, where we will test the hypothesis that personalized connectome maps predict therapeutic responses to intravenous methylphenidate. Likewise, findings from Phase 2A will inform the design of Phase 2B, where we plan to enroll patients based on their personalized connectome maps. By selecting patients for clinical trials based on a principled, mechanistic assessment of their neuroanatomic potential for a therapeutic response, the CCTP paradigm and the STIMPACT trial have the potential to transform the therapeutic landscape in the ICU and improve outcomes for patients with severe brain injuries.
Collapse
Affiliation(s)
- Brian L Edlow
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Megan E Barra
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pharmacy, Massachusetts General Hospital, Boston, MA, USA
| | - David W Zhou
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrea S Foulkes
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Samuel B Snider
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zachary D Threlkeld
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Sourish Chakravarty
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John E Kirsch
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Suk-Tak Chan
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Steven L Meisler
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas P Bleck
- Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Joseph J Fins
- Division of Medical Ethics and Consortium for the Advanced Study of Brain Injury (CASBI), Weill Cornell Medical College, New York, NY, USA
- The Rockefeller University, New York, NY, USA
- Solomon Center for Health Law and Policy, Yale Law School, New Haven, CT, USA
| | - Joseph T Giacino
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| | - Leigh R Hochberg
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- School of Engineering and Carney Institute for Brain Science, Brown University, Providence, RI, USA
- Veterans Affairs RR&D Center for Neurorestoration and Neurotechnology, VA Medical Center, Providence, RI, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Emery N Brown
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yelena G Bodien
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| |
Collapse
|
35
|
Pender SC, Smith AM, Finnoff JT, Huston J, Stuart MJ. Concussions in Ice Hockey - Moving Toward Objective Diagnoses and Point-of-care Treatment: A Review. Curr Sports Med Rep 2020; 19:380-386. [PMID: 32925378 DOI: 10.1249/jsr.0000000000000752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The incidence of sport-related concussion coupled with a doubling of the participation rate in youth hockey over the past two decades provides impetus for the review of the most promising concussion treatment options. This narrative review summarizes the future treatment options for sport-related concussions in ice hockey, while acknowledging their generalizability to concussion in all sports. Symptom assessment, sign observation, as well as cognitive and balance testing, have historically been used to diagnose a concussion. These methods continue to improve, but the need for effective treatments is clear. Pharmacologic, transcranial light, and nutritional supplement treatment options for concussion warranting further investigation have been identified. Dimethyl fumarate is an immunomodulatory compound thought to trigger antioxidant gene expression. Memantine reduces apoptosis and astrogliosis by inhibiting the calcium influx into cells normally caused by glutamate's activation of N-methyl-D-aspartate receptors. Thioredoxin-mimetic peptides and transcranial photobiomodulation temper the effects of the energy crisis by acting as free radical scavengers. In addition, seven neuroprotective nutritional supplements have been identified: berberine, creatine, curcumin, melatonin, omega-3 fatty acids, resveratrol, and vitamins. An estimated US $1.1 billion has been spent on unsuccessful traumatic brain injury clinical trials. As our ability to accurately diagnose concussion improves, dimethyl fumarate, memantine, thioredoxin-mimetic peptides, transcranial photobiomodulation, and nutritional supplements (berberine, creatine, curcumin, melatonin, omega-3 fatty acids, resveratrol, and vitamins) warrant further preclinical and clinical examination in advancing the treatment of sport-related concussions.
Collapse
Affiliation(s)
- Sara C Pender
- School of Medicine, University College Dublin, Dublin, IRELAND
| | | | - Jonathan T Finnoff
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - John Huston
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Michael J Stuart
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine and Science, Rochester, MN
| |
Collapse
|
36
|
Provencio JJ, Hemphill JC, Claassen J, Edlow BL, Helbok R, Vespa PM, Diringer MN, Polizzotto L, Shutter L, Suarez JI, Stevens RD, Hanley DF, Akbari Y, Bleck TP, Boly M, Foreman B, Giacino JT, Hartings JA, Human T, Kondziella D, Ling GSF, Mayer SA, McNett M, Menon DK, Meyfroidt G, Monti MM, Park S, Pouratian N, Puybasset L, Rohaut B, Rosenthal ES, Schiff ND, Sharshar T, Wagner A, Whyte J, Olson DM. The Curing Coma Campaign: Framing Initial Scientific Challenges-Proceedings of the First Curing Coma Campaign Scientific Advisory Council Meeting. Neurocrit Care 2020; 33:1-12. [PMID: 32578124 PMCID: PMC7392933 DOI: 10.1007/s12028-020-01028-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Coma and disordered consciousness are common manifestations of acute neurological conditions and are among the most pervasive and challenging aspects of treatment in neurocritical care. Gaps exist in patient assessment, outcome prognostication, and treatment directed specifically at improving consciousness and cognitive recovery. In 2019, the Neurocritical Care Society (NCS) launched the Curing Coma Campaign in order to address the "grand challenge" of improving the management of patients with coma and decreased consciousness. One of the first steps was to bring together a Scientific Advisory Council including coma scientists, neurointensivists, neurorehabilitationists, and implementation experts in order to address the current scientific landscape and begin to develop a framework on how to move forward. This manuscript describes the proceedings of the first Curing Coma Campaign Scientific Advisory Council meeting which occurred in conjunction with the NCS Annual Meeting in October 2019 in Vancouver. Specifically, three major pillars were identified which should be considered: endotyping of coma and disorders of consciousness, biomarkers, and proof-of-concept clinical trials. Each is summarized with regard to current approach, benefits to the patient, family, and clinicians, and next steps. Integration of these three pillars will be essential to the success of the Curing Coma Campaign as will expanding the "curing coma community" to ensure broad participation of clinicians, scientists, and patient advocates with the goal of identifying and implementing treatments to fundamentally improve the outcome of patients.
Collapse
Affiliation(s)
- J Javier Provencio
- Department of Neurology and Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - J Claude Hemphill
- Department of Neurology, Zuckerberg San Francisco General Hospital, University of California, San Francisco, Building 1, Room 101, 1001 Potrero Avenue, San Francisco, CA, 94110, USA.
| | - Jan Claassen
- Department of Neurology, Columbia University Irving Medical Center/New York Presbyterian Hospital, New York, NY, USA
| | - Brian L Edlow
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Raimund Helbok
- Department of Neurology, Neurocritical Care, Medical University of Innsbruck, Innsbruck, Austria
| | - Paul M Vespa
- Departments of Neurology and Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Michael N Diringer
- Department of Neurology, Washington University, Barnes-Jewish Hospital, St Louis, MO, USA
| | - Len Polizzotto
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Lori Shutter
- Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh/UPMC Health System, Pittsburgh, PA, USA
| | - Jose I Suarez
- Departments of Anesthesiology and Critical Care Medicine, Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert D Stevens
- Departments of Anesthesiology and Critical Care Medicine, Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel F Hanley
- Division of Brain Injury Outcomes, Johns Hopkins University, Baltimore, MD, USA
| | - Yama Akbari
- Departments of Neurology, Neurosurgery and the Beckman Laser Institute, University of California-Irvine, Irvine, CA, USA
| | - Thomas P Bleck
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Melanie Boly
- Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Brandon Foreman
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati Gardner Neuroscience Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joseph T Giacino
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Jed A Hartings
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Theresa Human
- Departments of Neurology and Neurosurgery, Washington University, Barnes-Jewish Hospital, St Louis, MO, USA
| | - Daniel Kondziella
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Geoffrey S F Ling
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephan A Mayer
- Departments of Neurology and Neurosurgery, New York Medical College, Valhalla, NY, USA
| | - Molly McNett
- College of Nursing, The Ohio State University, Columbus, OH, USA
| | - David K Menon
- Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | - Geert Meyfroidt
- Department and Laboratory of Intensive Care Medicine, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Martin M Monti
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Soojin Park
- Department of Neurology, Columbia University Irving Medical Center/New York Presbyterian Hospital, New York, NY, USA
| | - Nader Pouratian
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Louis Puybasset
- Department of Anesthesiology and Critical Care, Sorbonne University, GRC 29, AP-HP, DMU DREAM, Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - Benjamin Rohaut
- Department of Neurology, Neuro-ICU, Sorbonne University, Pitié-Salpêtrière Hospital, Paris, France
| | - Eric S Rosenthal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholas D Schiff
- Departments of Neurology, Neuroscience, and Medical Ethics, Weill Cornell Medicine, New York, NY, USA
| | - Tarek Sharshar
- Neuro-anesthesiology and Intensive Care Medicine, Sainte-Anne Hospital, Paris-Descartes University, Paris, France
- Experimental Neuropathology, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| | - Amy Wagner
- Department of Physical Medicine and Rehabilitation, Department of Neuroscience, Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - John Whyte
- Moss Rehabilitation Research Institute, Elkins Park, PA, USA
| | - DaiWai M Olson
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern, Dallas, TX, USA
| |
Collapse
|
37
|
Diamond BR, Donald CLM, Frau-Pascual A, Snider SB, Fischl B, Dams-O'Connor K, Edlow BL. Optimizing the accuracy of cortical volumetric analysis in traumatic brain injury. MethodsX 2020; 7:100994. [PMID: 32760659 PMCID: PMC7393399 DOI: 10.1016/j.mex.2020.100994] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/08/2020] [Indexed: 01/21/2023] Open
Abstract
Cortical volumetric analysis is widely used to study the anatomic basis of neurological deficits in patients with traumatic brain injury (TBI). However, patients with TBI-related lesions are often excluded from MRI analyses because cortical lesions may compromise the accuracy of reconstructed surfaces upon which volumetric measurements are based. We developed a FreeSurfer-based lesion correction method and tested its impact on cortical volume measures in 87 patients with chronic moderate-to-severe TBI. We reconstructed cortical surfaces from T1-weighted MRI scans, then manually labeled and removed vertices on the cortical surfaces where lesions caused inaccuracies. Next, we measured the surface area of lesion overlap with seven canonical brain networks and the percent volume of each network affected by lesions.The lesion correction method revealed that cortical lesions in patients with TBI are preferentially located in the limbic and default mode networks (95.7% each), with the limbic network also having the largest average surface area (4.4+/−3.7%) and percent volume affected by lesions (12.7+/−9.7%). The method has the potential to improve the accuracy of cortical volumetric measurements and permit inclusion of patients with lesioned brains in MRI analyses. The method also provides new opportunities to elucidate network-based mechanisms of neurological deficits in patients with TBI.
Collapse
Affiliation(s)
- Bram R Diamond
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | | | - Aina Frau-Pascual
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Samuel B Snider
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Bruce Fischl
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA.,Harvard-MIT Health Sciences and Technology, Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
| | - Kristen Dams-O'Connor
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Brian L Edlow
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| |
Collapse
|
38
|
Sanchez E, Arbour C, El-Khatib H, Marcotte K, Blais H, Baril AA, Bedetti C, Descoteaux M, Lina JM, Gilbert D, Carrier J, Gosselin N. Sleep spindles are resilient to extensive white matter deterioration. Brain Commun 2020; 2:fcaa071. [PMID: 32954326 PMCID: PMC7472897 DOI: 10.1093/braincomms/fcaa071] [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: 02/13/2020] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 12/29/2022] Open
Abstract
Sleep spindles are an essential part of non-rapid eye movement sleep, notably involved in sleep consolidation, cognition, learning and memory. These oscillatory waves depend on an interaction loop between the thalamus and the cortex, which relies on a structural backbone of thalamo-cortical white matter tracts. It is still largely unknown if the brain can properly produce sleep spindles when it underwent extensive white matter deterioration in these tracts, and we hypothesized that it would affect sleep spindle generation and morphology. We tested this hypothesis with chronic moderate to severe traumatic brain injury (n = 23; 30.5 ± 11.1 years old; 17 m/6f), a unique human model of extensive white matter deterioration, and a healthy control group (n = 27; 30.3 ± 13.4 years old; 21m/6f). Sleep spindles were analysed on a full night of polysomnography over the frontal, central and parietal brain regions, and we measured their density, morphology and sigma-band power. White matter deterioration was quantified using diffusion-weighted MRI, with which we performed both whole-brain voxel-wise analysis (Tract-Based Spatial Statistics) and probabilistic tractography (with High Angular Resolution Diffusion Imaging) to target the thalamo-cortical tracts. Group differences were assessed for all variables and correlations were performed separately in each group, corrected for age and multiple comparisons. Surprisingly, although extensive white matter damage across the brain including all thalamo-cortical tracts was evident in the brain-injured group, sleep spindles remained completely undisrupted when compared to a healthy control group. In addition, almost all sleep spindle characteristics were not associated with the degree of white matter deterioration in the brain-injured group, except that more white matter deterioration correlated with lower spindle frequency over the frontal regions. This study highlights the resilience of sleep spindles to the deterioration of all white matter tracts critical to their existence, as they conserve normal density during non-rapid eye movement sleep with mostly unaltered morphology. We show that even with such a severe traumatic event, the brain has the ability to adapt or to withstand alterations in order to conserve normal sleep spindles.
Collapse
Affiliation(s)
- Erlan Sanchez
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada.,Department of Neuroscience, Université de Montréal, Montreal H3T 1J4, Canada
| | - Caroline Arbour
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada.,Faculty of Nursing, Université de Montréal, Montreal H3T 1A8, Canada
| | - Héjar El-Khatib
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada.,Department of Psychology, Université de Montréal, Montreal H2V 2S9, Canada
| | - Karine Marcotte
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada.,School of Speech Language Pathology and Audiology, Université de Montréal, Montreal H3N 1X7, Canada
| | - Hélène Blais
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada
| | - Andrée-Ann Baril
- The Framingham Heart Study, Boston University School of Medicine, Boston 02118, USA
| | - Christophe Bedetti
- Department of Psychology, Université de Montréal, Montreal H2V 2S9, Canada
| | - Maxime Descoteaux
- Computer Science Department, Université de Sherbrooke, Sherbrooke J1K 2R1, Canada
| | - Jean-Marc Lina
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada.,Department of electrical engineering, École de Technologie Supérieure, Montreal H3C 1K3, Canada
| | - Danielle Gilbert
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada
| | - Julie Carrier
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada.,Department of Psychology, Université de Montréal, Montreal H2V 2S9, Canada
| | - Nadia Gosselin
- Research Center of the Centre Intégré Universitaire de Santé et de Services Sociaux du Nord de l'Île-de-Montréal, Montreal H4J 1C5, Canada.,Department of Psychology, Université de Montréal, Montreal H2V 2S9, Canada
| |
Collapse
|
39
|
Abstract
Traumatic brain injury is a calamity of various causes, pathologies, and extremely varied and often complex clinical presentations. Because of its predilection for brain systems underlying cognitive and complex behavioral operations, it may cause chronic and severe psychiatric illness that requires expert management. This is more so for the modern epidemic of athletic and military brain injuries which are dominated by psychiatric symptoms. Past medical, including psychiatric, history, and comorbidities are important and relevant for formulation and management. Traumatic brain injury is a model for other neuropsychiatric disorders and may serve as an incubator of new ideas for neurodegenerative disease.
Collapse
Affiliation(s)
- Vassilis E Koliatsos
- Department of Pathology (Neuropathology), Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Neuropsychiatry Program, Sheppard Pratt Health System, Baltimore, MD, USA.
| | - Vani Rao
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
Cross DJ, Meabon JS, Cline MM, Richards TL, Stump AJ, Cross CG, Minoshima S, Banks WA, Cook DG. Paclitaxel Reduces Brain Injury from Repeated Head Trauma in Mice. J Alzheimers Dis 2020; 67:859-874. [PMID: 30664506 DOI: 10.3233/jad-180871] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Repetitive mild traumatic brain injury (rmTBI) is known to disturb axonal integrity and may play an important role in the pathogenic cascades leading to neurodegeneration. One critical approach to reduce the future onset of neurodegeneration is to intervene in this process at an early stage following a brain injury. Previously we showed that direct application of the microtubule-stabilizing drug, paclitaxel, on the brain following controlled cortical impact improved motor function and reduced lesion size. Herein, we extended these findings to a model of mild brain injury induced by repeated closed-skull impacts. Paclitaxel was administered intranasally to circumvent its poor transport across the blood-brain barrier. Mice received five mild closed-skull impacts (one per day for five days). Intranasal paclitaxel was administered once only, immediately after the first impact. We found that paclitaxel prevented injury-induced deficits in a spatial memory task in a water tread maze. In vivo magnetic resonance imaging (MRI) and positron emission tomography with 18F-flurodeoxyglucose (FDG-PET) revealed that paclitaxel prevented structural injury and hypometabolism. On MRI, apparent, injury-induced microbleeds were observed in 100% of vehicle-treated rmTBI mice, but not in paclitaxel-treated subjects. FDG-PET revealed a 42% increase in whole brain glucose metabolism in paclitaxel-treated mice as compared to vehicle-treated rmTBI. Immunohistochemistry found reduced evidence of axonal injury and synaptic loss. Our results indicate that intranasal paclitaxel administration imparts neuroprotection against brain injury and cognitive impairment in mice. The results from this study support the idea that microtubule-stabilization strategies hold therapeutic promise in mitigating traumatic brain injury.
Collapse
Affiliation(s)
- Donna J Cross
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - James S Meabon
- The Mental Illness Research Education and Clinical Center (MIRECC), and VA Puget Sound Health Care System, Seattle, WA, USA.,Department of Psychiatry, University of Washington, Seattle, WA, USA
| | - Marcella M Cline
- Geriatric Research Education and Clinical Center (GRECC) and VA Puget Sound Health Care System, Seattle, WA, USA.,Department of Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Todd L Richards
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Amanda J Stump
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Chloe G Cross
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center (GRECC) and VA Puget Sound Health Care System, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - David G Cook
- Geriatric Research Education and Clinical Center (GRECC) and VA Puget Sound Health Care System, Seattle, WA, USA.,Department of Pharmacology, University of Washington, Seattle, WA, USA
| |
Collapse
|
41
|
Hill CS, Sreedharan J, Loreto A, Menon DK, Coleman MP. Loss of highwire Protects Against the Deleterious Effects of Traumatic Brain Injury in Drosophila Melanogaster. Front Neurol 2020; 11:401. [PMID: 32477254 PMCID: PMC7235382 DOI: 10.3389/fneur.2020.00401] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/17/2020] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury is a major global cause of death and disability. Axonal injury is a major underlying mechanism of TBI and could represent a major therapeutic target. We provide evidence that targeting the axonal death pathway known as Wallerian degeneration improves outcome in a Drosophila Melanogaster model of high impact trauma. This cell-autonomous neurodegenerative pathway is initiated following axon injury, and in Drosophila, involves activity of the E3 ubiquitin ligase highwire. We demonstrate that a loss-of-function mutation in the highwire gene rescues deleterious effects of a traumatic injury, including-improved functional outcomes, lifespan, survival of dopaminergic neurons, and retention of synaptic proteins. This data suggests that highwire represents a potential therapeutic target in traumatic injury.
Collapse
Affiliation(s)
- Ciaran S. Hill
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
- The Babraham Institute, Cambridge, United Kingdom
| | - Jemeen Sreedharan
- The Babraham Institute, Cambridge, United Kingdom
- Institute of Psychiatry, King's College London, London, United Kingdom
| | - Andrea Loreto
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - David K. Menon
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Clinical Neurosciences, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Michael P. Coleman
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
- The Babraham Institute, Cambridge, United Kingdom
| |
Collapse
|
42
|
Frank D, Melamed I, Gruenbaum BF, Grinshpun J, Kuts R, Shvartsur R, Azab AN, Assadi MH, Vinokur M, Boyko M. Induction of Diffuse Axonal Brain Injury in Rats Based on Rotational Acceleration. J Vis Exp 2020. [PMID: 32449735 DOI: 10.3791/61198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability. Diffuse axonal injury (DAI) is the predominant mechanism of injury in a large percentage of TBI patients requiring hospitalization. DAI involves widespread axonal damage from shaking, rotation or blast injury, leading to rapid axonal stretch injury and secondary axonal changes that are associated with a long-lasting impact on functional recovery. Historically, experimental models of DAI without focal injury have been difficult to design. Here we validate a simple, reproducible and reliable rodent model of DAI that causes widespread white matter damage without skull fractures or contusions.
Collapse
Affiliation(s)
- Dmitry Frank
- Division of Anesthesia and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev
| | - Israel Melamed
- Department of Neurosurgery, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev
| | | | - Julia Grinshpun
- Division of Anesthesia and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev
| | - Ruslan Kuts
- Division of Anesthesia and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev
| | - Rachel Shvartsur
- Recanati School for Community Health Professions, Faculty of Health Sciences, Ben-Gurion University of the Negev
| | - Abed N Azab
- Recanati School for Community Health Professions, Faculty of Health Sciences, Ben-Gurion University of the Negev
| | - Mohamad H Assadi
- Department of microbiology and immunology, Faculty of Health Sciences, Ben-Gurion University of the Negev
| | - Max Vinokur
- Division of Anesthesia and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev
| | - Matthew Boyko
- Division of Anesthesia and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev;
| |
Collapse
|
43
|
Honig MG, Dorian CC, Worthen JD, Micetich AC, Mulder IA, Sanchez KB, Pierce WF, Del Mar NA, Reiner A. Progressive long-term spatial memory loss following repeat concussive and subconcussive brain injury in mice, associated with dorsal hippocampal neuron loss, microglial phenotype shift, and vascular abnormalities. Eur J Neurosci 2020; 54:5844-5879. [PMID: 32090401 PMCID: PMC7483557 DOI: 10.1111/ejn.14711] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022]
Abstract
There is considerable concern about the long‐term deleterious effects of repeat head trauma on cognition, but little is known about underlying mechanisms and pathology. To examine this, we delivered four air blasts to the left side of the mouse cranium, a week apart, with an intensity that causes deficits when delivered singly and considered “concussive,” or an intensity that does not yield significant deficits when delivered singly and considered “subconcussive.” Neither repeat concussive nor subconcussive blast produced spatial memory deficits at 4 months, but both yielded deficits at 14 months, and dorsal hippocampal neuron loss. Hierarchical cluster analysis of dorsal hippocampal microglia across the three groups based on morphology and expression of MHCII, CX3CR1, CD68 and IBA1 revealed five distinct phenotypes. Types 1A and 1B microglia were more common in sham mice, linked to better neuron survival and memory, and appeared mildly activated. By contrast, 2B and 2C microglia were more common in repeat concussive and subconcussive mice, linked to poorer neuron survival and memory, and characterized by low expression levels and attenuated processes, suggesting they were de‐activated and dysfunctional. In addition, endothelial cells in repeat concussive mice exhibited reduced CD31 and eNOS expression, which was correlated with the prevalence of type 2B and 2C microglia. Our findings suggest that both repeat concussive and subconcussive head injury engender progressive pathogenic processes, possibly through sustained effects on microglia that over time lead to increased prevalence of dysfunctional microglia, adversely affecting neurons and blood vessels, and thereby driving neurodegeneration and memory decline.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Conor C Dorian
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - John D Worthen
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anthony C Micetich
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Isabelle A Mulder
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Katelyn B Sanchez
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - William F Pierce
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
44
|
Li Y, Li C, Gan C, Zhao K, Chen J, Song J, Lei T. A Precise, Controllable in vitro Model for Diffuse Axonal Injury Through Uniaxial Stretch Injury. Front Neurosci 2019; 13:1063. [PMID: 31680808 PMCID: PMC6811664 DOI: 10.3389/fnins.2019.01063] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 09/23/2019] [Indexed: 01/10/2023] Open
Abstract
Regarding the determination of the biomechanical parameters in a reliable in vitro cell model for diffuse axonal injury (DAI), our study aimed to demonstrate connections between those parameters and secondary axotomy through examination of morphological alterations under a variety of traumatic conditions. An in vitro cell model for DAI was established in primary cultured mouse neurons by uniaxial mechanical stretching of non-myelinated axons under various traumatic conditions: strain (ε) = 5, 10, 20, and 50%; strain time (t) = 500, 100, and 20 ms; strain rate ranging between 0.1 and 25 s-1. Axonal real strains (strainaxon) were measured as 4.53 ± 0.27, 9.02 ± 0.91, 17.75 ± 1.65, and 41.8 ± 4.4%. Axonal real strain rates (SRaxon) ranged between 0.096 ± 0.0054 and 20.9 ± 2.2 s-1. Results showed there was no obvious abnormality of axons with a lower strain condition (strainaxon < 17.75 ± 1.65%) during the acute phase within 30 min after injury. In contrast, acute axonal degeneration (AAD) was observed in the axons following injury with a higher strain condition (SRaxon > 17.75 ± 1.65%). In addition, the incidence and degree of AAD were closely correlated with strain rate. Specifically, AAD occurred to all axons that were examined, when ε = 50% (strainaxon = 41.8 ± 4.4%) for 20 ms, while no spontaneous rupture was observed in those axons. Besides, the concentration of Ca2+ within the axonal process was significantly increased under such traumatic conditions. Moreover, the continuity of axon cytoskeleton was interrupted, eventually resulting in neuronal death during subacute stage following injury. In this study, we found that there is a minimum strain threshold for the occurrence of AAD in non-myelinated axons of primary cultured mouse neurons, which ranges between 9.02 ± 0.91 and 17.75 ± 1.65%. Basically, the severity of axonal secondary axotomy post DAI is strain rate dependent under a higher strain above the threshold. Hence, a reliable and reproducible in vitro cell model for DAI was established, when ε = 50% (strainaxon = 41.8 ± 4.4%) for 20 ms.
Collapse
Affiliation(s)
- Yu Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| | - Chaoxi Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Gan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianbin Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinning Song
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Li HJ, Sun ZL, Pan YB, Xu MH, Feng DF. Effect of α7nAChR on learning and memory dysfunction in a rat model of diffuse axonal injury. Exp Cell Res 2019; 383:111546. [PMID: 31398352 DOI: 10.1016/j.yexcr.2019.111546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 11/30/2022]
Abstract
Diffuse axonal injury (DAI) is the predominant effect of severe traumatic brain injury and significantly contributes to cognitive deficits. The mechanisms that underlie these cognitive deficits are often associated with complex molecular alterations. α7nAChR, one of the abundant and widespread nicotinic acetylcholine receptors (nAChRs) in the brain, plays important physiological functions in the central nervous system. However, the relationship between temporospatial alterations in the α7nAChR and DAI-related learning and memory dysfunction are not completely understood. Our study detected temporospatial alterations of α7nAChR in vulnerable areas (hippocampus, internal capsule, corpus callosum and brain stem) of DAI rats and evaluated the development and progression of learning and memory dysfunction via the Morris water maze (MWM). We determined that α7nAChR expression in vulnerable areas was mainly reduced at the recovery of DAI in rats. Moreover, the escape latency of the injured group increased significantly and the percentages of the distance travelled and time spent in the target quadrant were significantly decreased after DAI. Furthermore, α7nAChR expression in the vulnerable area was significantly positively correlated with MWM performance after DAI according to regression analysis. In addition, we determined that a selective α7nAChR agonist significantly improved learning and memory dysfunction. Rats in the α7nAChR agonist group showed better learning and memory performance than those in the antagonist group. These results demonstrate that microstructural injury-induced alterations of α7nAChR in the vulnerable area are significantly correlated with learning and memory dysfunctions after DAI and that augmentation of the α7nAChR level by its agonist contributes to the improvement of learning and memory function.
Collapse
Affiliation(s)
- Hong-Jiang Li
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China; Institute of Traumatic Medicine, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Zhao-Liang Sun
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China; Institute of Traumatic Medicine, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Yuan-Bo Pan
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Mang-Hua Xu
- Institute of Traumatic Medicine, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Dong-Fu Feng
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China; Institute of Traumatic Medicine, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China.
| |
Collapse
|
46
|
Omelchenko A, Shrirao AB, Bhattiprolu AK, Zahn JD, Schloss RS, Dickson S, Meaney DF, Boustany NN, Yarmush ML, Firestein BL. Dynamin and reverse-mode sodium calcium exchanger blockade confers neuroprotection from diffuse axonal injury. Cell Death Dis 2019; 10:727. [PMID: 31562294 PMCID: PMC6765020 DOI: 10.1038/s41419-019-1908-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 08/20/2019] [Accepted: 08/27/2019] [Indexed: 12/18/2022]
Abstract
Mild traumatic brain injury (mTBI) is a frequently overlooked public health concern that is difficult to diagnose and treat. Diffuse axonal injury (DAI) is a common mTBI neuropathology in which axonal shearing and stretching induces breakdown of the cytoskeleton, impaired axonal trafficking, axonal degeneration, and cognitive dysfunction. DAI is becoming recognized as a principal neuropathology of mTBI with supporting evidence from animal model, human pathology, and neuroimaging studies. As mitochondrial dysfunction and calcium overload are critical steps in secondary brain and axonal injury, we investigated changes in protein expression of potential targets following mTBI using an in vivo controlled cortical impact model. We show upregulated expression of sodium calcium exchanger1 (NCX1) in the hippocampus and cortex at distinct time points post-mTBI. Expression of dynamin-related protein1 (Drp1), a GTPase responsible for regulation of mitochondrial fission, also changes differently post-injury in the hippocampus and cortex. Using an in vitro model of DAI previously reported by our group, we tested whether pharmacological inhibition of NCX1 by SN-6 and of dynamin1, dynamin2, and Drp1 by dynasore mitigates secondary damage. Dynasore and SN-6 attenuate stretch injury-induced swelling of axonal varicosities and mitochondrial fragmentation. In addition, we show that dynasore, but not SN-6, protects against H2O2-induced damage in an organotypic oxidative stress model. As there is currently no standard treatment to mitigate cell damage induced by mTBI and DAI, this work highlights two potential therapeutic targets for treatment of DAI in multiple models of mTBI and DAI.
Collapse
Affiliation(s)
- Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Anil B Shrirao
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Atul K Bhattiprolu
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Jeffrey D Zahn
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Samantha Dickson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104-6391, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104-6391, USA
| | - Nada N Boustany
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA.
| |
Collapse
|
47
|
Erythropoietin Does Not Alter Serum Profiles of Neuronal and Axonal Biomarkers After Traumatic Brain Injury: Findings From the Australian EPO-TBI Clinical Trial. Crit Care Med 2019; 46:554-561. [PMID: 29278529 DOI: 10.1097/ccm.0000000000002938] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To determine profiles of serum ubiquitin carboxy-terminal hydrolase L1 and phosphorylated neurofilament heavy-chain, examine whether erythropoietin administration reduce their concentrations, and whether biomarkers discriminate between erythropoietin and placebo treatment groups. DESIGN Single-center, prospective observational study. SETTING A sub-study of the erythropoietin-traumatic brain injury clinical trial, conducted at the Alfred Hospital, Melbourne, Australia. PATIENTS Forty-four patients with moderate-to-severe traumatic brain injury. INTERVENTIONS Epoetin alfa 40,000 IU or 1 mL sodium chloride 0.9 as subcutaneous injection within 24 hours of traumatic brain injury. MEASUREMENTS AND MAIN RESULTS Ubiquitin carboxy-terminal hydrolase L1, phosphorylated neurofilament heavy-chain, and erythropoietin concentrations were measured in serum by enzyme-linked immunosorbent assay from D0 (within 24 hr of injury, prior to erythropoietin/vehicle administration) to D5. Biomarker concentrations were compared between injury severities, diffuse versus focal traumatic brain injury and erythropoietin or placebo treatment groups. Ubiquitin carboxy-terminal hydrolase L1 peaked at 146.0 ng/mL on D0, significantly decreased to 84.30 ng/mL on D1, and declined thereafter. Phosphorylated neurofilament heavy-chain levels were lowest at D0 and peaked on D5 at 157.9 ng/mL. D0 ubiquitin carboxy-terminal hydrolase L1 concentrations were higher in diffuse traumatic brain injury. Peak phosphorylated neurofilament heavy-chain levels on D3 and D4 correlated with Glasgow Outcome Score-Extended, predicting poor outcome. Erythropoietin did not reduce concentrations of ubiquitin carboxy-terminal hydrolase L1 or phosphorylated neurofilament heavy-chain. CONCLUSIONS Serum ubiquitin carboxy-terminal hydrolase L1 and phosphorylated neurofilament heavy-chain increase after traumatic brain injury reflecting early neuronal and progressive axonal injury. Consistent with lack of improved outcome in traumatic brain injury patients treated with erythropoietin, biomarker concentrations and profiles were not affected by erythropoietin. Pharmacokinetics of erythropoietin suggest that the dose given was possibly too low to exert neuroprotection.
Collapse
|
48
|
Marion CM, McDaniel DP, Armstrong RC. Sarm1 deletion reduces axon damage, demyelination, and white matter atrophy after experimental traumatic brain injury. Exp Neurol 2019; 321:113040. [PMID: 31445042 DOI: 10.1016/j.expneurol.2019.113040] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) often damages axons in white matter tracts and causes corpus callosum (CC) atrophy in chronic TBI patients. Injured axons encounter irreversible damage if transected, or alternatively may maintain continuity and subsequently either recover or degenerate. Secondary mechanisms can cause further axon damage, myelin pathology, and neuroinflammation. Molecular mechanisms regulating the progression of white matter pathology indicate potential therapeutic targets. SARM1 is essential for execution of the conserved axon death pathway. We examined white matter pathology following mild TBI with CC traumatic axonal injury in mice with Sarm1 gene deletion (Sarm1-/-). High resolution ultrastructural analysis at 3 days post-TBI revealed dramatically reduced axon damage in Sarm1-/- mice, as compared to Sarm1+/+ wild-type controls. Sarm1 deletion produced larger axons with thinner myelin, and attenuated TBI induced demyelination, i.e. myelin loss along apparently intact axons. At 6 weeks post-TBI, Sarm1-/- mice had less demyelination and thinner myelin than Sarm1+/+ mice, but axonal protection was no longer observed. We next used Thy1-YFP crosses to assess Sarm1 involvement in white matter neurodegeneration and neuroinflammation at 8 weeks post-TBI, when significant CC atrophy indicates chronic pathology. Thy1-YFP expression demonstrated continued CC axon damage yet absence of overt cortical pathology. Importantly, significant CC atrophy in Thy1-YFP/Sarm1+/+ mice was associated with reduced neurofilament immunolabeling of axons. Both effects were attenuated in Thy1-YFP/Sarm1-/- mice. Surprisingly, Thy1-YFP/Sarm1-/- mice had increased CC astrogliosis. This study demonstrates that Sarm1 inactivation reduces demyelination, and white matter atrophy after TBI, while the post-injury stage impacts when axon protection is effective.
Collapse
Affiliation(s)
- Christina M Marion
- Center for Neuroscience and Regenerative Medicine, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Program in Neuroscience, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Dennis P McDaniel
- Biomedical Instrumentation Center, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Regina C Armstrong
- Center for Neuroscience and Regenerative Medicine, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Program in Neuroscience, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| |
Collapse
|
49
|
Bodien YG, McCrea M, Dikmen S, Temkin N, Boase K, Joan M, Taylor SR, Sherer M, Levin H, Kramer JH, Corrigan JD, McAllister TW, Whyte J, Manley GT, Giacino JT. Optimizing Outcome Assessment in Multicenter TBI Trials: Perspectives From TRACK-TBI and the TBI Endpoints Development Initiative. J Head Trauma Rehabil 2019; 33:147-157. [PMID: 29385010 PMCID: PMC5940527 DOI: 10.1097/htr.0000000000000367] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Traumatic brain injury (TBI) is a global public health problem that affects the long-term cognitive, physical, and psychological health of patients, while also having a major impact on family and caregivers. In stark contrast to the effective trials that have been conducted in other neurological diseases, nearly 30 studies of interventions employed during acute hospital care for TBI have failed to identify treatments that improve outcome. Many factors may confound the ability to detect true and meaningful treatment effects. One promising area for improving the precision of intervention studies is to optimize the validity of the outcome assessment battery by using well-designed tools and data collection strategies to reduce variability in the outcome data. The Transforming Research and Clinical Knowledge in TBI (TRACK-TBI) study, conducted at 18 sites across the United States, implemented a multidimensional outcome assessment battery with 22 measures aimed at characterizing TBI outcome up to 1 year postinjury. In parallel, through the TBI Endpoints Development (TED) Initiative, federal agencies and investigators have partnered to identify the most valid, reliable, and sensitive outcome assessments for TBI. Here, we present lessons learned from the TRACK-TBI and TED initiatives aimed at optimizing the validity of outcome assessment in TBI.
Collapse
Affiliation(s)
- Yelena G. Bodien
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA
| | - Michael McCrea
- Departments of Neurosurgery and Neurology, Medical College of Wisconsin, Milwaukee, WI
| | - Sureyya Dikmen
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA Departments of
| | - Nancy Temkin
- Neurological Surgery and Biostatistics, University of Washington, Seattle, WA
| | - Kim Boase
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA Departments of
| | - Machamer Joan
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA Departments of
| | - Sabrina R. Taylor
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California, San Francisco, CA
| | - Mark Sherer
- TIRR Memorial Hermann, Houston, TX
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX
| | - Harvey Levin
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX
| | - Joel H. Kramer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA
| | - John D. Corrigan
- Department of Physical Medicine & Rehabilitation, The Ohio State University
| | - Thomas W. McAllister
- Department of Psychiatry, University of Indiana School of Medicine, Indianapolis, IN
| | - John Whyte
- Moss Rehabilitation Research Institute, Elkins Park, PA
| | - Geoffrey T. Manley
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California, San Francisco, CA
| | - Joseph T. Giacino
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA
| | | |
Collapse
|
50
|
Hajiaghamemar M, Seidi M, Oeur RA, Margulies SS. Toward development of clinically translatable diagnostic and prognostic metrics of traumatic brain injury using animal models: A review and a look forward. Exp Neurol 2019; 318:101-123. [PMID: 31055005 PMCID: PMC6612432 DOI: 10.1016/j.expneurol.2019.04.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/11/2019] [Accepted: 04/30/2019] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury is a leading cause of cognitive and behavioral deficits in children in the US each year. There is an increasing interest in both clinical and pre-clinical studies to discover biomarkers to accurately diagnose traumatic brain injury (TBI), predict its outcomes, and monitor its progression especially in the developing brain. In humans, the heterogeneity of TBI in terms of clinical presentation, injury causation, and mechanism has contributed to the many challenges associated with finding unifying diagnosis, treatment, and management practices. In addition, findings from adult human research may have little application to pediatric TBI, as age and maturation levels affect the injury biomechanics and neurophysiological consequences of injury. Animal models of TBI are vital to address the variability and heterogeneity of TBI seen in human by isolating the causation and mechanism of injury in reproducible manner. However, a gap between the pre-clinical findings and clinical applications remains in TBI research today. To take a step toward bridging this gap, we reviewed several potential TBI tools such as biofluid biomarkers, electroencephalography (EEG), actigraphy, eye responses, and balance that have been explored in both clinical and pre-clinical studies and have shown potential diagnostic, prognostic, or monitoring utility for TBI. Each of these tools measures specific deficits following TBI, is easily accessible, non/minimally invasive, and is potentially highly translatable between animals and human outcomes because they involve effort-independent and non-verbal tasks. Especially conspicuous is the fact that these biomarkers and techniques can be tailored for infants and toddlers. However, translation of preclinical outcomes to clinical applications of these tools necessitates addressing several challenges. Among the challenges are the heterogeneity of clinical TBI, age dependency of some of the biomarkers, different brain structure, life span, and possible variation between temporal profiles of biomarkers in human and animals. Conducting parallel clinical and pre-clinical research, in addition to the integration of findings across species from several pre-clinical models to generate a spectrum of TBI mechanisms and severities is a path toward overcoming some of these challenges. This effort is possible through large scale collaborative research and data sharing across multiple centers. In addition, TBI causes dynamic deficits in multiple domains, and thus, a panel of biomarkers combining these measures to consider different deficits is more promising than a single biomarker for TBI. In this review, each of these tools are presented along with the clinical and pre-clinical findings, advantages, challenges and prospects of translating the pre-clinical knowledge into the human clinical setting.
Collapse
Affiliation(s)
- Marzieh Hajiaghamemar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| | - Morteza Seidi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - R Anna Oeur
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Susan S Margulies
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|