1
|
Whitehead B, Corbin D, Meadows E, Zhang N, Hollander JM, Karelina K, Weil ZM. Cerebral hypoperfusion exacerbates vascular dysfunction after traumatic brain injury. Exp Neurol 2024; 380:114907. [PMID: 39103029 DOI: 10.1016/j.expneurol.2024.114907] [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: 02/28/2024] [Revised: 06/17/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
Traumatic brain injuries are extremely common, and although most patients recover from their injuries many TBI patients suffer prolonged symptoms and remain at a higher risk for developing cardiovascular disease and neurodegeneration. Moreover, it remains challenging to identify predictors of poor long-term outcomes. Here, we tested the hypothesis that preexisting cerebrovascular impairment exacerbates metabolic and vascular dysfunction and leads to worse outcomes after TBI. Male mice underwent a mild surgical reduction in cerebral blood flow using a model of bilateral carotid artery stenosis (BCAS) wherein steel microcoils were implanted around the carotid arteries. Then, 30 days post coil implantation, mice underwent TBI or sham surgery. Gene expression profiles, cerebral blood flow, metabolic function, oxidative damage, vascular health and angiogenesis were assessed. Single nuclei RNA sequencing of endothelial cells isolated from mice after TBI showed differential gene expression profiles after TBI and BCAS, that were further altered when mice underwent both challenges. TBI but not BCAS increased mitochondrial oxidative metabolism. Both BCAS and TBI decreased cerebrovascular responses to repeated whisker stimulation. BCAS induced oxidative damage and inflammation in the vasculature as well as loss of vascular density, and reduced the numbers of angiogenic tip cells. Finally, intravascular protein accumulation was increased among mice that experienced both BCAS and TBI. Overall, our findings reveal that a prior vascular impairment significantly alters the profile of vascular health and function of the cerebrovasculature, and when combined with TBI may result in worsened outcomes.
Collapse
Affiliation(s)
- Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA.
| | - Deborah Corbin
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Ethan Meadows
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA; Department of Human Performance - Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Ning Zhang
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - John M Hollander
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA; Department of Human Performance - Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Zachary M Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
2
|
Adam CD, Mirzakhalili E, Gagnon KG, Cottone C, Arena JD, Ulyanova AV, Johnson VE, Wolf JA. Disrupted Hippocampal Theta-Gamma Coupling and Spike-Field Coherence Following Experimental Traumatic Brain Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596704. [PMID: 39314320 PMCID: PMC11418945 DOI: 10.1101/2024.05.30.596704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Traumatic brain injury (TBI) often results in persistent learning and memory deficits, likely due to disrupted hippocampal circuitry underlying these processes. Precise temporal control of hippocampal neuronal activity is important for memory encoding and retrieval and is supported by oscillations that dynamically organize single unit firing. Using high-density laminar electrophysiology, we discovered a loss of oscillatory power across CA1 lamina, with a profound, layer-specific reduction in theta-gamma phase amplitude coupling in injured rats. Interneurons from injured animals were less strongly entrained to theta and gamma oscillations, suggesting a mechanism for the loss of coupling, while pyramidal cells were entrained to a later phase of theta. During quiet immobility, we report decreased ripple amplitudes from injured animals during sharp-wave ripple events. These results reveal deficits in information encoding and retrieval schemes essential to cognition that likely underlie TBI-associated learning and memory impairments, and elucidate potential targets for future neuromodulation therapies.
Collapse
Affiliation(s)
- Christopher D Adam
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Ehsan Mirzakhalili
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Kimberly G Gagnon
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Carlo Cottone
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - John D Arena
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Alexandra V Ulyanova
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, USA
| | - Victoria E Johnson
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - John A Wolf
- Center for Brain Injury and Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, USA
| |
Collapse
|
3
|
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
|
4
|
Hubbard WB, Velmurugan GV, Sullivan PG. The role of mitochondrial uncoupling in the regulation of mitostasis after traumatic brain injury. Neurochem Int 2024; 174:105680. [PMID: 38311216 PMCID: PMC10922998 DOI: 10.1016/j.neuint.2024.105680] [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: 11/03/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
Mitostasis, the maintenance of healthy mitochondria, plays a critical role in brain health. The brain's high energy demands and reliance on mitochondria for energy production make mitostasis vital for neuronal function. Traumatic brain injury (TBI) disrupts mitochondrial homeostasis, leading to secondary cellular damage, neuronal degeneration, and cognitive deficits. Mild mitochondrial uncoupling, which dissociates ATP production from oxygen consumption, offers a promising avenue for TBI treatment. Accumulating evidence, from endogenous and exogenous mitochondrial uncoupling, suggests that mitostasis is closely regulating by mitochondrial uncoupling and cellular injury environments may be more sensitive to uncoupling. Mitochondrial uncoupling can mitigate calcium overload, reduce oxidative stress, and induce mitochondrial proteostasis and mitophagy, a process that eliminates damaged mitochondria. The interplay between mitochondrial uncoupling and mitostasis is ripe for further investigation in the context of TBI. These multi-faceted mechanisms of action for mitochondrial uncoupling hold promise for TBI therapy, with the potential to restore mitochondrial health, improve neurological outcomes, and prevent long-term TBI-related pathology.
Collapse
Affiliation(s)
- W Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA; Department of Physiology, University of Kentucky, Lexington, KY, USA; Lexington Veterans' Affairs Healthcare System, Lexington, KY, USA.
| | - Gopal V Velmurugan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA; Lexington Veterans' Affairs Healthcare System, Lexington, KY, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
5
|
Aqel S, Al-Thani N, Haider MZ, Abdelhady S, Al Thani AA, Kobeissy F, Shaito AA. Biomaterials in Traumatic Brain Injury: Perspectives and Challenges. BIOLOGY 2023; 13:21. [PMID: 38248452 PMCID: PMC10813103 DOI: 10.3390/biology13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and long-term impairment globally. TBI has a dynamic pathology, encompassing a variety of metabolic and molecular events that occur in two phases: primary and secondary. A forceful external blow to the brain initiates the primary phase, followed by a secondary phase that involves the release of calcium ions (Ca2+) and the initiation of a cascade of inflammatory processes, including mitochondrial dysfunction, a rise in oxidative stress, activation of glial cells, and damage to the blood-brain barrier (BBB), resulting in paracellular leakage. Currently, there are no FDA-approved drugs for TBI, but existing approaches rely on delivering micro- and macromolecular treatments, which are constrained by the BBB, poor retention, off-target toxicity, and the complex pathology of TBI. Therefore, there is a demand for innovative and alternative therapeutics with effective delivery tactics for the diagnosis and treatment of TBI. Tissue engineering, which includes the use of biomaterials, is one such alternative approach. Biomaterials, such as hydrogels, including self-assembling peptides and electrospun nanofibers, can be used alone or in combination with neuronal stem cells to induce neurite outgrowth, the differentiation of human neural stem cells, and nerve gap bridging in TBI. This review examines the inclusion of biomaterials as potential treatments for TBI, including their types, synthesis, and mechanisms of action. This review also discusses the challenges faced by the use of biomaterials in TBI, including the development of biodegradable, biocompatible, and mechanically flexible biomaterials and, if combined with stem cells, the survival rate of the transplanted stem cells. A better understanding of the mechanisms and drawbacks of these novel therapeutic approaches will help to guide the design of future TBI therapies.
Collapse
Affiliation(s)
- Sarah Aqel
- Medical Research Center, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
| | - Najlaa Al-Thani
- Research and Development Department, Barzan Holdings, Doha P.O. Box 7178, Qatar
| | - Mohammad Z. Haider
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria 21544, Egypt;
| | - Asmaa A. Al Thani
- Biomedical Research Center and Department of Biomedical Sciences, College of Health Science, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310, USA
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
6
|
Hoogenboom WS, Rubin TG, Ambadipudi K, Cui MH, Ye K, Foster H, Elkouby E, Liu J, Branch CA, Lipton ML. Evolving brain and behaviour changes in rats following repetitive subconcussive head impacts. Brain Commun 2023; 5:fcad316. [PMID: 38046094 PMCID: PMC10691880 DOI: 10.1093/braincomms/fcad316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/26/2023] [Accepted: 11/19/2023] [Indexed: 12/05/2023] Open
Abstract
There is growing concern that repetitive subconcussive head impacts, independent of concussion, alter brain structure and function, and may disproportionately affect the developing brain. Animal studies of repetitive subconcussive head impacts are needed to begin to characterize the pathological basis and mechanisms underlying imaging and functional effects of repetitive subconcussive head impacts seen in humans. Since repetitive subconcussive head impacts have been largely unexplored in animals, we aimed to characterize the evolution of imaging, behavioural and pathological effects of repetitive subconcussive head impacts in awake adolescent rodents. Awake male and female Sprague Dawley rats (postnatal Day 35) received 140 closed-head impacts over the course of a week. Impacted and sham-impacted animals were restrained in a plastic cone, and unrestrained control animals were included to account for effects of restraint and normal development. Animals (n = 43) underwent repeated diffusion tensor imaging prior to and over 1 month following the final impact. A separate cohort (n = 53) was assessed behaviourally for fine motor control, emotional-affective behaviour and memory at acute and chronic time points. Histological and immunohistochemical analyses, which were exploratory in nature due to smaller sample sizes, were completed at 1 month following the final impact. All animals tolerated the protocol with no overt changes in behaviour or stigmata of traumatic brain injury, such as alteration of consciousness, intracranial haemorrhage or skull fracture. We detected longitudinal, sex-dependent diffusion tensor imaging changes (fractional anisotropy and axial diffusivity decline) in corpus callosum and external capsule of repetitive subconcussive head impact animals, which diverged from both sham and control. Compared to sham animals, repetitive subconcussive head impact animals exhibited acute but transient mild motor deficits. Repetitive subconcussive head impact animals also exhibited chronic anxiety and spatial memory impairment that differed from the control animals, but these effects were not different from those seen in the sham condition. We observed trends in the data for thinning of the corpus callosum as well as regions with elevated Iba-1 in the corpus callosum and cerebral white matter among repetitive subconcussive head impact animals. While replication with larger study samples is needed, our findings suggest that subconcussive head impacts cause microstructural tissue changes in the developing rat brain, which are detectable with diffusion tensor imaging, with suggestion of correlates in tissue pathology and behaviour. The results point to potential mechanisms underpinning consequences of subconcussive head impacts that have been described in humans. The congruence of our imaging findings with human subconcussive head impacts suggests that neuroimaging could serve as a translational bridge to advance study of injury mechanisms and development of interventions.
Collapse
Affiliation(s)
- Wouter S Hoogenboom
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Clinical Investigation, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Todd G Rubin
- Department of Neurology, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Kamalakar Ambadipudi
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Min-Hui Cui
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Kenny Ye
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Henry Foster
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Esther Elkouby
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Jinyuan Liu
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Craig A Branch
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Michael L Lipton
- Department of Radiology, Columbia University Irving Medical Center, NewYork, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, NewYork, NY 10032, USA
| |
Collapse
|
7
|
Gamage R, Rossetti I, Niedermayer G, Münch G, Buskila Y, Gyengesi E. Chronic neuroinflammation during aging leads to cholinergic neurodegeneration in the mouse medial septum. J Neuroinflammation 2023; 20:235. [PMID: 37833764 PMCID: PMC10576363 DOI: 10.1186/s12974-023-02897-5] [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: 08/07/2023] [Accepted: 09/14/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Low-grade, chronic inflammation in the central nervous system characterized by glial reactivity is one of the major hallmarks for aging-related neurodegenerative diseases like Alzheimer's disease (AD). The basal forebrain cholinergic neurons (BFCN) provide the primary source of cholinergic innervation of the human cerebral cortex and may be differentially vulnerable in various neurodegenerative diseases. However, the impact of chronic neuroinflammation on the cholinergic function is still unclear. METHODS To gain further insight into age-related cholinergic decline, we investigated the cumulative effects of aging and chronic neuroinflammation on the structure and function of the septal cholinergic neurons in transgenic mice expressing interleukin-6 under the GFAP promoter (GFAP-IL6), which maintains a constant level of gliosis. Immunohistochemistry combined with unbiased stereology, single cell 3D morphology analysis and in vitro whole cell patch-clamp measurements were used to validate the structural and functional changes of BFCN and their microglial environment in the medial septum. RESULTS Stereological estimation of MS microglia number displayed significant increase across all three age groups, while a significant decrease in cholinergic cell number in the adult and aged groups in GFAP-IL6 mice compared to control. Moreover, we observed age-dependent alterations in the electrophysiological properties of cholinergic neurons and an increased excitability profile in the adult GFAP-IL6 group due to chronic neuroinflammation. These results complimented the significant decrease in hippocampal pyramidal spine density seen with aging and neuroinflammation. CONCLUSIONS We provide evidence of the significant impact of both aging and chronic glial activation on the cholinergic and microglial numbers and morphology in the MS, and alterations in the passive and active electrophysiological membrane properties of septal cholinergic neurons, resulting in cholinergic dysfunction, as seen in AD. Our results indicate that aging combined with gliosis is sufficient to cause cholinergic disruptions in the brain, as seen in dementias.
Collapse
Affiliation(s)
- Rashmi Gamage
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Ilaria Rossetti
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Garry Niedermayer
- School of Science, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Gerald Münch
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Erika Gyengesi
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia.
| |
Collapse
|
8
|
Barreto GE, Gonzalez J, Ramírez D. Network pharmacology and topological analysis on tibolone metabolites and their molecular mechanisms in traumatic brain injury. Biomed Pharmacother 2023; 165:115089. [PMID: 37418975 DOI: 10.1016/j.biopha.2023.115089] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/09/2023] Open
Abstract
Traumatic brain injury (TBI) is a pathology of great social impact, affecting millions of people worldwide. Despite the scientific advances to improve the management of TBI in recent years, we still do not have a specific treatment that controls the inflammatory process after mechanical trauma. The discovery and implementation of new treatments is a long and expensive process, making the repurpose of approved drugs for other pathologies a clinical interest. Tibolone is a drug in use for the treatment of symptoms associated with menopause and has been shown to have a broad spectrum of actions by regulating estrogen, androgen and progesterone receptors, whose activation exerts potent anti-inflammatory and antioxidant effects. In the present study, we aimed to investigate the therapeutic potential of the tibolone metabolites 3α-Hydroxytibolone, 3β-Hydroxytibolone, and Δ4-Tibolone as a possible therapy in TBI using network pharmacology and network topology analysis. Our results demonstrate that the estrogenic component mediated by the α and β metabolites can regulate synaptic transmission and cell metabolism, while the Δ metabolite may be involved in modulating the post-TBI inflammatory process. We identified several molecular targets, including KDR, ESR2, AR, NR3C1, PPARD, and PPARA, which are known to play critical roles in the pathogenesis of TBI. Tibolone metabolites were predicted to regulate the expression of key genes involved in oxidative stress, inflammation, and apoptosis. Overall, the repurposing of tibolone as a neuroprotective treatment for TBI holds promise for future clinical trials. However, further studies are needed to confirm its efficacy and safety in TBI patients.
Collapse
Affiliation(s)
- George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| | - Janneth Gonzalez
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - David Ramírez
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
9
|
Lendvai D, Whittemore R, Womack JA, Fortier CB, Milberg WP, Fonda JR. The Impact of Blast Exposure-With or Without Traumatic Brain Injury-on Metabolic Abnormalities in Post-9/11 Veterans. J Head Trauma Rehabil 2023; 38:380-390. [PMID: 36951458 PMCID: PMC10514232 DOI: 10.1097/htr.0000000000000874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
OBJECTIVE The primary aim included explorations of: (1) the associations between the history of blast exposure (BE), close blast exposure (CBE), and blast-related traumatic brain injury (bTBI) and metabolic abnormality; and (2) the potential mediating effect of comorbid psychological and somatic conditions on these associations. The secondary aim explored the association of dose-response impact of BE, CBE, and bTBI and metabolic abnormality. SETTING Data were collected by the Translational Research Center for TBI and Stress Disorders (TRACTS). PARTICIPANTS Post-9/11 veterans from the TRACTS baseline sample who had conflict-zone deployment experience ( N = 734). DESIGN Cross-sectional secondary data analysis. We computed relative risks (RRs) and 95% CI using modified Poisson regression. We quantified the impact of co-occurring psychological and somatic conditions on this association using mediation analyses. MAIN MEASURES Exposures included BE (<100 m), CBE (<10 m), and bTBI. Metabolic abnormality outcomes included (1) overweight/obesity (defined by abnormal waist-hip ratio [WHR] and abnormal waist circumference [WC]); (2) glucose dysregulation; and (3) meeting criteria for cardiometabolic syndrome (defined by guidelines). RESULTS The sample was majority male (91%) and White (68%), with a mean age of 34.6 years (SD = 8.99). Most participants had 1 or more BE (83%); 48% experienced 1 or more CBE. Overweight/obesity was highly prevalent in the sample (51% had abnormal WHR and 60% abnormal WC). There was no significant direct or indirect association between BE, CBE, and bTBI and metabolic abnormalities (RRs: 0.70-1.51; P 's > .05). CONCLUSION Future research is needed to investigate the association of BE with metabolic abnormalities with larger, more targeted sample selection, and longer follow-up. Effective and sustainable weight management and metabolic health prevention interventions for this veteran cohort are needed.
Collapse
Affiliation(s)
- Dora Lendvai
- VA Connecticut Healthcare System, West Haven, Connecticut
- Yale University, School of Nursing, Orange, Connecticut
| | | | - Julie A. Womack
- VA Connecticut Healthcare System, West Haven, Connecticut
- Yale University, School of Nursing, Orange, Connecticut
| | - Catherine B. Fortier
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research, Education and Clinical Center (GRECC), VA Boston Healthcare System, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - William P. Milberg
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research, Education and Clinical Center (GRECC), VA Boston Healthcare System, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Jennifer R. Fonda
- Translational Research Center for TBI and Stress Disorders (TRACTS) and Geriatric Research, Education and Clinical Center (GRECC), VA Boston Healthcare System, Boston, Massachusetts
- Department of Psychiatry, Boston University School of Medicine, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
10
|
Whitehead B, Velazquez-Cruz R, Albowaidey A, Zhang N, Karelina K, Weil ZM. Mild Traumatic Brain Injury Induces Time- and Sex-Dependent Cerebrovascular Dysfunction and Stroke Vulnerability. J Neurotrauma 2023; 40:578-591. [PMID: 36322789 PMCID: PMC9986031 DOI: 10.1089/neu.2022.0335] [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] [Indexed: 11/06/2022] Open
Abstract
Mild traumatic brain injury (mTBI) produces subtle cerebrovascular impairments that persist over time and promote increased ischemic stroke vulnerability. We recently established a role for vascular impairments in exacerbating stroke outcomes 1 week after TBI, but there is a lack of research regarding long-term impacts of mTBI-induced vascular dysfunction, as well as a significant need to understand how mTBI promotes stroke vulnerability in both males and females. Here, we present data using a mild closed head TBI model and an experimental stroke occurring either 7 or 28 days later in both male and female mice. We report that mTBI induces larger stroke volumes 7 days after injury, however, this increased vulnerability to stroke persists out to 28 days in female but not male mice. Importantly, mTBI-induced changes in blood-brain barrier permeability, intravascular coagulation, angiogenic factors, total vascular area, and glial expression were differentially altered across time and by sex. Taken together, these data suggest that mTBI can result in persistent cerebrovascular dysfunction and increased susceptibility to worsened ischemic outcomes, although these dysfunctions occur differently in male and female mice.
Collapse
Affiliation(s)
- Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Ruth Velazquez-Cruz
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Ali Albowaidey
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Ning Zhang
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Zachary M. Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
11
|
O'Brien WT, Wright DK, van Emmerik ALJJ, Bain J, Brkljaca R, Christensen J, Yamakawa GR, Chen Z, Giesler LP, Sun M, O'Brien TJ, Monif M, Shultz SR, McDonald SJ. Serum neurofilament light as a biomarker of vulnerability to a second mild traumatic brain injury. Transl Res 2022; 255:77-84. [PMID: 36402367 DOI: 10.1016/j.trsl.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022]
Abstract
A second mild traumatic brain injury (mTBI) sustained prior to neuropathological recovery can lead to exacerbated effects. Without objective indicators of this neuropathology, individuals may return to activities at risk of mTBI when their brain is still vulnerable. With axonal injury recognized as a neuropathological hallmark of mTBI, we hypothesized that serum levels of neurofilament light (NfL), a highly sensitive biomarker of axonal injury, may be predictive of vulnerability to worse outcomes in the event of a second mTBI. Given this hypothesis is difficult to test clinically, we used a two-hit model of mTBI in rats and staggered inter-injury intervals by 1-, 3-, 7-, or 14-days. Repeat-mTBI rats were dichotomized into NfLhigh (NfL>median at the time of re-injury) and NfLlow (NfL<median) groups, with behavior and NfL levels analyzed throughout the 28-days, followed by ex vivo diffusion tensor imaging. NfL levels at the time of the second mTBI were found to be predictive of vulnerability to re-injury, with NfLhigh rats displaying more neurological signs and a greater potentiation of NfL levels after the second mTBI. Importantly, this potentiation phenomenon remained even when limiting analyses to rats with longer inter-injury intervals, providing evidence that vulnerability to re-injury may not be exclusively dependent on inter-injury interval. Finally, NfL levels correlated with, and were predictive of, the severity of neurological signs following the second mTBI. These findings provide evidence that measurement of NfL during mTBI recovery may be reflective of the vulnerability to a second mTBI, and as such may have utility to assist return to sport, duty and work decisions.
Collapse
Affiliation(s)
- William T O'Brien
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia
| | | | - Jesse Bain
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia
| | | | | | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia
| | - Zhibin Chen
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia
| | - Lauren P Giesler
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, 3004, Australia
| | - Mastura Monif
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, 3004, Australia; Health and Human Services, Vancouver Island University, Nanaimo, V9R 5S5, Canada
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, 3004, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, 3004, Australia.
| |
Collapse
|
12
|
Weil ZM, White B, Whitehead B, Karelina K. The role of the stress system in recovery after traumatic brain injury: A tribute to Bruce S. McEwen. Neurobiol Stress 2022; 19:100467. [PMID: 35720260 PMCID: PMC9201063 DOI: 10.1016/j.ynstr.2022.100467] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 11/21/2022] Open
Abstract
Traumatic brain injury (TBI) represents a major public health concern. Although the majority of individuals that suffer mild-moderate TBI recover relatively quickly, a substantial subset of individuals experiences prolonged and debilitating symptoms. An exacerbated response to physiological and psychological stressors after TBI may mediate poor functional recovery. Individuals with TBI can suffer from poor stress tolerance, impairments in the ability to evaluate stressors, and poor initiation (and cessation) of neuroendocrine stress responses, all of which can exacerbate TBI-mediated dysfunction. Here, we pay tribute to the pioneering neuroendocrinologist Dr. Bruce McEwen by discussing the ways in which his work on stress physiology and allostatic loading impacts the TBI patient population both before and after their injuries. Specifically, we will discuss the modulatory role of hypothalamic-pituitary-adrenal axis responses immediately after TBI and later in recovery. We will also consider the impact of stressors and stress responses in promoting post-concussive syndrome and post-traumatic stress disorders, two common sequelae of TBI. Finally, we will explore the role of early life stressors, prior to brain injuries, as modulators of injury outcomes.
Collapse
Affiliation(s)
- Zachary M. Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Brishti White
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Rd, Morgantown, WV, 26506, USA
| |
Collapse
|
13
|
Dunn CS, Ferreira LA, Venier SM, Ali SF, Wolchok JC, Balachandran K. Functional Analysis of the Cortical Transcriptome and Proteome Reveal Neurogenesis, Inflammation, and Cell Death after Repeated Traumatic Brain Injury In vivo. Neurotrauma Rep 2022; 3:224-239. [PMID: 35919509 PMCID: PMC9279125 DOI: 10.1089/neur.2021.0059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The pathological effects of repeated traumatic brain injuries (TBIs) are largely unknown. To gain a detailed understanding of the cortical tissue acute biological response after one or two TBIs, we utilized RNA-sequencing and protein mass spectrometry techniques. Using our previously validated C57Bl/6 weight-drop model, we administered one or two TBIs of a mild or moderate severity. Double injury conditions were spaced 7 days apart, and cortical tissue was isolated 24 h after final injury. Analysis was carried out through functional gene annotation, utilizing Gene Ontology, for both the proteome and transcriptome. Major themes across the four different conditions include: neurogenesis; inflammation and immune response; cell death; angiogenesis; protein modification; and cell communication. Proteins associated with neurogenesis were found to be upregulated after single injuries. Transcripts associated with angiogenesis were upregulated in the moderate single, mild double, and moderate double TBI conditions. Genes associated with inflammation and immune response were upregulated in every condition, with the moderate single condition reporting the most functional groups. Proteins or genes involved in cell death, or apoptosis, were upregulated in every condition. Our results emphasize the significant differences found in proteomic and transcriptomic changes in single versus double injuries. Further, cortical omics analysis offers important insights for future studies aiming to deepen current knowledge on the development of secondary injuries and neurobehavioral impairments after brain trauma.
Collapse
Affiliation(s)
- Celeste S. Dunn
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
| | - Laís A. Ferreira
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
| | - Sara M. Venier
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, USA
| | - Syed F. Ali
- Neurochemistry Laboratory, Division of Neurotoxicology, National Center for Toxicological Research, Jefferson, Arkansas, USA
- Center for Integrativve Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, USA
| | - Jeffrey C. Wolchok
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Kartik Balachandran
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
14
|
Knight JM, Szabo A, Arapi I, Wu R, Emmrich A, Hackett E, Sauber G, Yim S, Johnson B, Hari P, Schneider D, Dropulic B, Cusatis RN, Cole SW, Hillard CJ, Shah NN. Patient-reported outcomes and neurotoxicity markers in patients treated with bispecific LV20.19 CAR T cell therapy. COMMUNICATIONS MEDICINE 2022; 2:49. [PMID: 35603278 PMCID: PMC9098435 DOI: 10.1038/s43856-022-00116-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 04/25/2022] [Indexed: 01/24/2023] Open
Abstract
Background With the rising number of chimeric antigen receptor (CAR) T cell treated patients, it is increasingly important to understand the treatment's impact on patient-reported outcomes (PROs) and, ideally, identify biomarkers of central nervous system (CNS) adverse effects. Methods The purpose of this exploratory study was to assess short-term PROs and serum kynurenine metabolites for associated neurotoxicity among patients treated in an anti-CD20, anti-CD19 (LV20.19) CAR T cell phase I clinical trial (NCT03019055). Fifteen CAR T treated patients from the parent trial provided serum samples and self-report surveys 15 days before and 14, 28, and 90 days after treatment. Results Blood kynurenine concentrations increased over time in patients with evidence of neurotoxicity (p = 0.004) and were increased in self-reported depression (r = 0.52, p = 0.002). Depression improved after CAR T infusion (p = 0.035). Elevated 3-hydroxyanthranilic acid (3HAA) concentrations prior to cell infusion were also predictive of neurotoxicity onset (p = 0.031), suggesting it is a biomarker of neurotoxicity following CAR T cell therapy. Conclusions Elevated levels of kynurenine pathway metabolites among CAR T cell recipients are associated with depressed mood and neurotoxicity. Findings from this exploratory study are preliminary and warrant validation in a larger cohort.
Collapse
Affiliation(s)
- Jennifer M. Knight
- grid.30760.320000 0001 2111 8460Department of Psychiatry, Medical College of Wisconsin, Milwaukee, WI USA ,grid.30760.320000 0001 2111 8460Departments of Medicine and Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI USA
| | - Aniko Szabo
- grid.30760.320000 0001 2111 8460Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI USA
| | - Igli Arapi
- grid.30760.320000 0001 2111 8460Department of Psychiatry, Medical College of Wisconsin, Milwaukee, WI USA
| | - Ruizhe Wu
- grid.30760.320000 0001 2111 8460Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI USA
| | - Amanda Emmrich
- grid.30760.320000 0001 2111 8460Department of Psychiatry, Medical College of Wisconsin, Milwaukee, WI USA
| | - Edward Hackett
- grid.30760.320000 0001 2111 8460Medical College of Wisconsin, Milwaukee, WI USA
| | - Garrett Sauber
- grid.30760.320000 0001 2111 8460Department of Pharmacology and Toxicology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI USA
| | - Sharon Yim
- grid.30760.320000 0001 2111 8460BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI USA
| | - Bryon Johnson
- grid.30760.320000 0001 2111 8460BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI USA
| | - Parameswaran Hari
- grid.30760.320000 0001 2111 8460BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI USA
| | - Dina Schneider
- Lentigen, a Miltenyi Biotec company, Gaithersburg, MD USA
| | - Boro Dropulic
- Lentigen, a Miltenyi Biotec company, Gaithersburg, MD USA
| | - Rachel N. Cusatis
- grid.30760.320000 0001 2111 8460Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI USA
| | - Steve W. Cole
- grid.19006.3e0000 0000 9632 6718Departments of Psychiatry and Biobehavioral Sciences and Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Cecilia J. Hillard
- grid.30760.320000 0001 2111 8460Department of Pharmacology and Toxicology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI USA
| | - Nirav N. Shah
- grid.30760.320000 0001 2111 8460BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI USA
| |
Collapse
|
15
|
Saffron extract and crocin exert anti-inflammatory and anti-oxidative effects in a repetitive mild traumatic brain injury mouse model. Sci Rep 2022; 12:5004. [PMID: 35322143 PMCID: PMC8943204 DOI: 10.1038/s41598-022-09109-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/10/2022] [Indexed: 12/25/2022] Open
Abstract
Saffron Crocus sativus L. (C. sativus) is a flower from the iridaceous family. Crocin, saffron’s major constituent, and saffron have anti-oxidative and anti-inflammatory activities. In this work, the neuroprotective effects of saffron and crocin are being investigated in a repetitive mild traumatic brain injury (rmTBI) mouse model. A weight drop model setup was employed to induce mild brain injury in male albino BABL/c mice weighing 30–40 g. Saffron (50 mg/kg) and crocin (30 mg/kg) were administrated intraperitoneally 30 min before mTBI induction. Behavioral tests were conducted to assess behavioral deficits including the modified neurological severity score (NSS), Morris water maze (MWM), pole climb test, rotarod test, and adhesive test. The levels of TNF alpha (TNF-α), interferon-gamma (IFN-γ), myeloperoxidase activity (MPO), malonaldehyde (MDA), and reduced glutathione (GSH) were measured. Histological analysis of different brain parts was performed. Both saffron and crocin demonstrated marked improved neurological, cognitive, motor, and sensorimotor functions. Besides, both compounds significantly reduced the oxidative stress and inflammatory processes. No abnormal histological features were observed in any of the injured groups. Saffron extract and crocin provide a neuroprotective effect in a mouse model of rmTBI by decreasing oxidative stress, inflammatory responses, and behavioral deficits.
Collapse
|
16
|
Mitoquinone supplementation alleviates oxidative stress and pathologic outcomes following repetitive mild traumatic brain injury at a chronic time point. Exp Neurol 2022; 351:113987. [DOI: 10.1016/j.expneurol.2022.113987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/23/2021] [Accepted: 01/13/2022] [Indexed: 11/17/2022]
|
17
|
Cai X, Harding IC, Sadaka AH, Colarusso B, Kulkarni P, Ebong E, Qiao J, O'Hare NR, Ferris CF. Mild repetitive head impacts alter perivascular flow in the midbrain dopaminergic system in awake rats. Brain Commun 2021; 3:fcab265. [PMID: 34806002 PMCID: PMC8600963 DOI: 10.1093/braincomms/fcab265] [Citation(s) in RCA: 7] [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: 05/06/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 11/24/2022] Open
Abstract
Head injury is a known risk factor for Parkinson's disease. Disruption in the perivascular clearance of metabolic waste and unwanted proteins is thought to be a contributing factor to disease progression. We hypothesized that repetitive mild head impacts, without evidence of structural brain damage, would increase microgliosis and AQP4 expression and depolarization and alter perivascular flow in the midbrain dopaminergic system. Adult male rats were subjected to sham, or two mild head impacts separated by 48 h. Three weeks later, fully awake rats were imaged using dynamic, contrast-enhanced MRI to follow the distribution of intraventricular gadobenate dimeglumine contrast agent. Images were registered to and analysed using a 3D MRI rat atlas providing site-specific data on 171 different brain areas. Following imaging, rats were tested for cognitive function using the Barnes maze assay. Histological analyses of tyrosine hydroxylase, microglia activation and AQP4 expression and polarization were performed on a parallel cohort of head impacted rats at 20 days post insult to coordinate with the time of imaging. There was no change in the global flux of contrast agent between sham and head impacted rats. The midbrain dopaminergic system showed a significant decrease in the influx of contrast agent as compared to sham controls together with a significant increase in microgliosis, AQP4 expression and depolarization. There were no deficits in cognitive function. The histology showed a significant level of neuroinflammation in the midbrain dopaminergic system 3 weeks post mild repetitive head impact but no loss in tyrosine hydroxylase. MRI revealed no structural brain damage emphasizing the potential serious consequences of mild head impacts on sustained brain neuroinflammation in this area critical to the pathophysiology of Parkinson's.
Collapse
Affiliation(s)
- Xuezhu Cai
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Ian C Harding
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Aymen H Sadaka
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Bradley Colarusso
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Praveen Kulkarni
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Eno Ebong
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ju Qiao
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
| | - Nick R O'Hare
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Craig F Ferris
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA 02115, USA
- Department of Psychology, Northeastern University, Boston, MA 02115, USA
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
18
|
Leaston J, Qiao J, Harding IC, Kulkarni P, Gharagouzloo C, Ebong E, Ferris CF. Quantitative Imaging of Blood-Brain Barrier Permeability Following Repetitive Mild Head Impacts. Front Neurol 2021; 12:729464. [PMID: 34659094 PMCID: PMC8515019 DOI: 10.3389/fneur.2021.729464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/24/2021] [Indexed: 12/28/2022] Open
Abstract
This was an exploratory study designed to evaluate the feasibility of a recently established imaging modality, quantitative ultrashort time-to-echo contrast enhanced (QUTE-CE), to follow the early pathology and vulnerability of the blood brain barrier in response to single and repetitive mild head impacts. A closed-head, momentum exchange model was used to produce three consecutive mild head impacts aimed at the forebrain separated by 24 h each. Animals were measured at baseline and within 1 h of impact. Anatomical images were collected to assess the extent of structural damage. QUTE-CE biomarkers for BBB permeability were calculated on 420,000 voxels in the brain and were registered to a bilateral 3D brain atlas providing site-specific information on 118 anatomical regions. Blood brain barrier permeability was confirmed by extravasation of labeled dextran. All head impacts occurred in the absence of any structural brain damage. A single mild head impact had measurable effects on blood brain barrier permeability and was more significant after the second and third impacts. Affected regions included the prefrontal ctx, basal ganglia, hippocampus, amygdala, and brainstem. Our findings support the concerns raised by the healthcare community regarding mild head injuries in participants in organized contact sports and military personnel in basic training and combat.
Collapse
Affiliation(s)
| | - Ju Qiao
- Center for Translational Neuroimaging, Northeastern University, Boston, MA, United States
| | - Ian C. Harding
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | | | - Codi Gharagouzloo
- Imaginostics, Inc., Cambridge, MA, United States
- Center for Translational Neuroimaging, Northeastern University, Boston, MA, United States
| | - Eno Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Craig F. Ferris
- Center for Translational Neuroimaging, Northeastern University, Boston, MA, United States
- Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| |
Collapse
|
19
|
Haque A, Drasites KP, Cox A, Capone M, Myatich AI, Shams R, Matzelle D, Garner DP, Bredikhin M, Shields DC, Vertegel A, Banik NL. Protective Effects of Estrogen via Nanoparticle Delivery to Attenuate Myelin Loss and Neuronal Death after Spinal Cord Injury. Neurochem Res 2021; 46:2979-2990. [PMID: 34269965 DOI: 10.1007/s11064-021-03401-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 11/24/2022]
Abstract
Spinal cord injury (SCI) is associated with devastating neurological deficits affecting more than 11,000 Americans each year. Although several therapeutic agents have been proposed and tested, no FDA-approved pharmacotherapy is available for SCI treatment. We have recently demonstrated that estrogen (E2) acts as an antioxidant and anti-inflammatory agent, attenuating gliosis in SCI. We have also demonstrated that nanoparticle-mediated focal delivery of E2 to the injured spinal cord decreases lesion size, reactive gliosis, and glial scar formation. The current study tested in vitro effects of E2 on reactive oxygen species (ROS) and calpain activity in microglia, astroglia, macrophages, and fibroblasts, which are believed to participate in the inflammatory events and glial scar formation after SCI. E2 treatment decreased ROS production and calpain activity in these glial cells, macrophages, and fibroblast cells in vitro. This study also tested the efficacy of fast- and slow-release nanoparticle-E2 constructs in a rat model of SCI. Focal delivery of E2 via nanoparticles increased tissue distribution of E2 over time, attenuated cell death, and improved myelin preservation in injured spinal cord. Specifically, the fast-release nanoparticle-E2 construct reduced the Bax/Bcl-2 ratio in injured spinal cord tissues, and the slow-release nanoparticle-E2 construct prevented gliosis and penumbral demyelination distal to the lesion site. These data suggest this novel E2 delivery strategy to the lesion site may decrease inflammation and improve functional outcomes following SCI.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| | - Kelsey P Drasites
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.,Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA.,Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
| | - April Cox
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
| | - Mollie Capone
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.,Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
| | - Ali I Myatich
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.,Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA.,Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
| | - Ramsha Shams
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.,Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA.,Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA.,Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
| | - Dena P Garner
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
| | | | - Donald C Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
| | - Alexey Vertegel
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA. .,Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA. .,Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA. .,Department of Neurosurgery and Neurology, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA.
| |
Collapse
|
20
|
Karelina K, Schneiderman K, Shah S, Fitzgerald J, Cruz RV, Oliverio R, Whitehead B, Yang J, Weil ZM. Moderate Intensity Treadmill Exercise Increases Survival of Newborn Hippocampal Neurons and Improves Neurobehavioral Outcomes after Traumatic Brain Injury. J Neurotrauma 2021; 38:1858-1869. [PMID: 33470170 PMCID: PMC8219196 DOI: 10.1089/neu.2020.7389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Physician-prescribed rest after traumatic brain injury (TBI) is both commonplace and an increasingly scrutinized approach to TBI treatment. Although this practice remains a standard of patient care for TBI, research of patient outcomes reveals little to no benefit of prescribed rest after TBI, and in some cases prolonged rest has been shown to interfere with patient well-being. In direct contrast to the clinical advice regarding physical activity after TBI, animal models of brain injury consistently indicate that exercise is neuroprotective and promotes recovery. Here, we assessed the effect of low and moderate intensity treadmill exercise on functional outcome and hippocampal neural proliferation after brain injury. Using the controlled cortical impact (CCI) mouse model of TBI, we show that 10 days of moderate intensity treadmill exercise initiated after CCI reduces anxiety-like behavior, improves hippocampus-dependent spatial memory, and promotes hippocampal proliferation and newborn neuronal survival. Pathophysiological measures including lesion volume and axon degeneration were not altered by exercise. Taken together, these data reveal that carefully titrated physical activity may be a safe and effective approach to promoting recovery after brain injury.
Collapse
Affiliation(s)
- Kate Karelina
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Katarina Schneiderman
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Sarthak Shah
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Julie Fitzgerald
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ruth Velazquez Cruz
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Robin Oliverio
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Bailey Whitehead
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Jingzhen Yang
- Nationwide Children's Hospital, Center for Injury Research and Policy, Columbus, Ohio, USA
| | - Zachary M. Weil
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
21
|
Postolache TT, Wadhawan A, Can A, Lowry CA, Woodbury M, Makkar H, Hoisington AJ, Scott AJ, Potocki E, Benros ME, Stiller JW. Inflammation in Traumatic Brain Injury. J Alzheimers Dis 2021; 74:1-28. [PMID: 32176646 DOI: 10.3233/jad-191150] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is an increasing evidence that inflammation contributes to clinical and functional outcomes in traumatic brain injury (TBI). Many successful target-engaging, lesion-reducing, symptom-alleviating, and function-improving interventions in animal models of TBI have failed to show efficacy in clinical trials. Timing and immunological context are paramount for the direction, quality, and intensity of immune responses to TBI and the resulting neuroanatomical, clinical, and functional course. We present components of the immune system implicated in TBI, potential immune targets, and target-engaging interventions. The main objective of our article is to point toward modifiable molecular and cellular mechanisms that may modify the outcomes in TBI, and contribute to increasing the translational value of interventions that have been identified in animal models of TBI.
Collapse
Affiliation(s)
- Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD, USA
| | - Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Saint Elizabeths Hospital, Department of Psychiatry, Washington, DC, USA
| | - Adem Can
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Christopher A Lowry
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Margaret Woodbury
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew J Hoisington
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, CO, USA.,Systems Engineering and Management, Air Force Institute of Technology, Wright-Patterson AFB, OH, USA
| | - Alison J Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Eileen Potocki
- VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Michael E Benros
- Copenhagen Research Center for Mental Health-CORE, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - John W Stiller
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Maryland State Athletic Commission, Baltimore, MD, USA.,Saint Elizabeths Hospital, Neurology Consultation Services, Washington, DC, USA
| |
Collapse
|
22
|
Weil ZM, Karelina K, Whitehead B, Velazquez-Cruz R, Oliverio R, Pinti M, Nwafor DC, Nicholson S, Fitzgerald JA, Hollander J, Brown CM, Zhang N, DeVries AC. Mild traumatic brain injury increases vulnerability to cerebral ischemia in mice. Exp Neurol 2021; 342:113765. [PMID: 33992581 DOI: 10.1016/j.expneurol.2021.113765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/15/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
Recent studies have reported that TBI is an independent risk factor for subsequent stroke. Here, we tested the hypothesis that TBI would exacerbate experimental stroke outcomes via alternations in neuroimmune and neurometabolic function. We performed a mild closed-head TBI and then one week later induced an experimental stroke in adult male mice. Mice that had previously experienced TBI exhibited larger infarcts, greater functional deficits, and more pronounced neuroinflammatory responses to stroke. We hypothesized that impairments in central metabolic physiology mediated poorer outcomes after TBI. To test this, we treated mice with the insulin sensitizing drug pioglitazone (Pio) after TBI. Pio prevented the exacerbation of ischemic outcomes induced by TBI and also blocked the induction of insulin insensitivity by TBI. However, tissue respiratory function was not improved by Pio. Finally, TBI altered microvascular responses including promoting vascular accumulation of serum proteins and significantly impairing blood flow during the reperfusion period after stroke, both of which were reversed by treatment with Pio. Thus, TBI appears to exacerbate ischemic outcomes by impairing metabolic and microvascular physiology. These data have important implications because TBI patients experience strokes at greater rates than individuals without a history of head injury, but these data suggest that those strokes may also cause greater tissue damage and functional impairments in that population.
Collapse
Affiliation(s)
- Zachary M Weil
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA.
| | - Kate Karelina
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA
| | - Bailey Whitehead
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA
| | - Ruth Velazquez-Cruz
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA
| | - Robin Oliverio
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA
| | - Mark Pinti
- Department of Exercise Physiology, West Virginia University School of Medicine, 1 Medical Center Dr., Morgantown, WV 26506, USA; Mitochondria, Metabolism, & Bioenergetics Working Group, West Virginia University School of Medicine, 1 Medical Center Dr., Morgantown, WV 26506, USA
| | - Divine C Nwafor
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA
| | - Samuel Nicholson
- Department of Neuroscience, Ohio State University, 460 West 12th Ave., Columbus, OH 43210, USA
| | - Julie A Fitzgerald
- Department of Neuroscience, Ohio State University, 460 West 12th Ave., Columbus, OH 43210, USA
| | - John Hollander
- Department of Exercise Physiology, West Virginia University School of Medicine, 1 Medical Center Dr., Morgantown, WV 26506, USA; Mitochondria, Metabolism, & Bioenergetics Working Group, West Virginia University School of Medicine, 1 Medical Center Dr., Morgantown, WV 26506, USA
| | - Candice M Brown
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA
| | - Ning Zhang
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA
| | - A Courtney DeVries
- Department of Medicine, WVU Cancer Institute, WVU Rockefeller Neuroscience Institute, West Virginia University, BMRC, 1 Medical Center Dr., PO Box 9303, Morgantown, WV 26506, USA
| |
Collapse
|
23
|
McDaid J, Briggs CA, Barrington NM, Peterson DA, Kozlowski DA, Stutzmann GE. Sustained Hippocampal Synaptic Pathophysiology Following Single and Repeated Closed-Head Concussive Impacts. Front Cell Neurosci 2021; 15:652721. [PMID: 33867941 PMCID: PMC8044326 DOI: 10.3389/fncel.2021.652721] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/11/2021] [Indexed: 11/24/2022] Open
Abstract
Traumatic brain injury (TBI), and related diseases such as chronic traumatic encephalopathy (CTE) and Alzheimer's (AD), are of increasing concern in part due to enhanced awareness of their long-term neurological effects on memory and behavior. Repeated concussions, vs. single concussions, have been shown to result in worsened and sustained symptoms including impaired cognition and histopathology. To assess and compare the persistent effects of single or repeated concussive impacts on mediators of memory encoding such as synaptic transmission, plasticity, and cellular Ca2+ signaling, a closed-head controlled cortical impact (CCI) approach was used which closely replicates the mode of injury in clinical cases. Adult male rats received a sham procedure, a single impact, or three successive impacts at 48-hour intervals. After 30 days, hippocampal slices were prepared for electrophysiological recordings and 2-photon Ca2+ imaging, or fixed and immunostained for pathogenic phospho-tau species. In both concussion groups, hippocampal circuits showed hyper-excitable synaptic responsivity upon Schaffer collateral stimulation compared to sham animals, indicating sustained defects in hippocampal circuitry. This was not accompanied by sustained LTP deficits, but resting Ca2+ levels and voltage-gated Ca2+ signals were elevated in both concussion groups, while ryanodine receptor-evoked Ca2+ responses decreased with repeat concussions. Furthermore, pathogenic phospho-tau staining was progressively elevated in both concussion groups, with spreading beyond the hemisphere of injury, consistent with CTE. Thus, single and repeated concussions lead to a persistent upregulation of excitatory hippocampal synapses, possibly through changes in postsynaptic Ca2+ signaling/regulation, which may contribute to histopathology and detrimental long-term cognitive symptoms.
Collapse
Affiliation(s)
- John McDaid
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Clark A. Briggs
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Nikki M. Barrington
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Daniel A. Peterson
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Center for Stem Cell and Regenerative Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Dorothy A. Kozlowski
- Department of Biological Sciences and Neuroscience Program, DePaul University, Chicago, IL, United States
| | - Grace E. Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Center for Stem Cell and Regenerative Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
24
|
O'Brien WT, Symons GF, Bain J, Major BP, Costello DM, Sun M, Kimpton JS, Chen Z, Brady RD, Mychasiuk R, O'Brien TJ, Monif M, Shultz SR, McDonald SJ. Elevated Serum Interleukin-1β Levels in Male, but not Female, Collision Sport Athletes with a Concussion History. J Neurotrauma 2021; 38:1350-1357. [PMID: 33308001 DOI: 10.1089/neu.2020.7479] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It is increasingly reported that a history of concussion may be associated with chronic deleterious consequences. While the pathophysiology that contributes to these consequences is not well understood, neuroinflammation is postulated to be critical. Activation of multi-protein complexes termed inflammasomes, a key component of this inflammatory response, has been reported in more severe TBIs; however, it has not been investigated in milder TBIs, such as concussion. This study investigated serum levels of interleukin (IL)-1β and IL-18 (key proteins activated downstream of these inflammasomes) at acute, sub-acute, and chronic time-points post-concussion. We recruited 105 Australian footballers (65 male, 40 female) during the pre-season, then prospectively followed these players for the occurrence of concussion during the season. At baseline, 58 footballers reported a previous concussion history, and 47 reported no previous concussion history. Additionally, 25 players sustained a mid-season concussion and were sampled at 2, 6, and 13 days post-concussion. Serum levels of IL-1β and IL-18 were quantified using highly sensitive Simoa HD-X Analyzer assays. At baseline, IL-1β levels were higher in male, but not female, footballers with a previous concussion history compared with footballers with no concussion history. There was also a positive correlation between years of collision sport participation and IL-18 levels in males. No evidence was found in males or females to indicate that IL-1β or IL-18 levels differed at 2, 6, or 13 days post-concussion. These findings provide novel insights into potential sex-specific physiological consequences of concussion, and suggest that neuroinflammation may be persistent chronically following concussion in male athletes.
Collapse
Affiliation(s)
- William T O'Brien
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Georgia F Symons
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Jesse Bain
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Brendan P Major
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Daniel M Costello
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Joshua S Kimpton
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Zhibin Chen
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Epidemiology, Monash University, Melbourne, Victoria, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Melbourne Health, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Mastura Monif
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Melbourne Health, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.,Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Blackwell LS, Martinez M, Fournier-Goodnight A, Figueroa J, Appert A, Vats A, Wali B, Sayeed I, Reisner A. Patterns of Osteopontin Expression in Abusive Head Trauma Compared with Other Causes of Pediatric Traumatic Brain Injury. J Pediatr 2020; 227:170-175. [PMID: 32622673 PMCID: PMC7686267 DOI: 10.1016/j.jpeds.2020.06.080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To examine levels of plasma osteopontin (OPN), a recently described neuroinflammatory biomarker, in children with abusive head trauma (AHT) compared with children with other types of traumatic brain injury (TBI). STUDY DESIGN The study cohort comprised children aged <4 years diagnosed with TBI and seen in the intensive care unit in a tertiary children's hospital. Patients were classified as having confirmed or suspected AHT or TBI by other mechanisms (eg, motor vehicle accidents), as identified by a Child Protection Team clinician. Serial blood samples were collected at admission and at 24, 48, and 72 hours after admission. Levels of OPN were compared across groups. RESULTS Of 77 patients identified, 24 had confirmed AHT, 12 had suspected AHT, and 41 had TBI. There were no differences in the Glasgow Coma Scale score between the patients with confirmed AHT and those with suspected AHT and those with TBI (median score, 4.5 vs 4 and 7; P = .39). At admission to the emergency department, OPN levels were significantly higher in children with confirmed AHT compared with the other 2 groups (mean confirmed AHT, 471.5 ng/mL; median suspected AHT, 322.3 ng/mL; mean TBI, 278.0 ng/mL; P = .03). Furthermore, the adjusted mean trajectory levels of OPN were significantly higher in the confirmed AHT group compared with the other 2 groups across all subsequent time points (P = <.01). CONCLUSIONS OPN is significantly elevated in children with confirmed AHT compared with those with suspected AHT and those with other types of TBI. OPN expression may help identify children with suspected AHT to aid resource stratification and triage of appropriate interventions for children who are potential victims of abuse.
Collapse
Affiliation(s)
- Laura S. Blackwell
- Department of Neuropsychology, Children’s Healthcare of Atlanta, Atlanta, GA
| | | | | | | | - Andrew Appert
- Department of Neuropsychology, Children’s Healthcare of Atlanta, Atlanta, GA
| | - Atul Vats
- Department of Neuropsychology, Children’s Healthcare of Atlanta, Atlanta, GA,Emory University, Atlanta, GA
| | | | | | - Andrew Reisner
- Department of Neuropsychology, Children’s Healthcare of Atlanta, Atlanta, GA
| |
Collapse
|
26
|
Hoffman J, Yu J, Kirstein C, Kindy MS. Combined Effects of Repetitive Mild Traumatic Brain Injury and Alcohol Drinking on the Neuroinflammatory Cytokine Response and Cognitive Behavioral Outcomes. Brain Sci 2020; 10:brainsci10110876. [PMID: 33228251 PMCID: PMC7699568 DOI: 10.3390/brainsci10110876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 12/22/2022] Open
Abstract
The relationship between alcohol consumption and traumatic brain injury (TBI) often focuses on alcohol consumption increasing the likelihood of incurring a TBI, rather than alcohol use outcomes after TBI. However, patients without a history of an alcohol use disorder can also show increased problem drinking after single or multiple TBIs. Alcohol and mild TBI share diffuse deleterious neurological impacts and cognitive impairments; therefore, the purpose of these studies was to determine if an interaction on brain and behavior outcomes occurs when alcohol is consumed longitudinally after TBI. To examine the impact of mild repetitive TBI (rmTBI) on voluntary alcohol consumption, mice were subjected to four mild TBI or sham procedures over a 2 week period, then offered alcohol (20% v/v) for 2 weeks using the two-bottle choice, drinking in the dark protocol. Following the drinking period, mice were evaluated for neuroinflammatory cytokine response or tested for cognitive and behavioral deficits. Results indicate no difference in alcohol consumption or preference following rmTBI as compared to sham; however, increases in the neuroinflammatory cytokine response due to alcohol consumption and some mild cognitive behavioral deficits after rmTBI and alcohol consumption were observed. These data suggest that the cytokine response to alcohol drinking and rmTBI + alcohol drinking is not necessarily aggregate, but the combination does result in an exacerbation of cognitive behavioral outcomes.
Collapse
Affiliation(s)
- Jessica Hoffman
- Department of Psychiatry, Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: (J.H.); (M.S.K.); Tel.: +1-919-843-4389 (J.H.)
| | - Jin Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA;
| | - Cheryl Kirstein
- Department of Psychology, College of Arts and Sciences, University of South Florida, Tampa, FL 33612, USA;
| | - Mark S. Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA;
- James A. Haley VA Medical Center, Tampa, FL 33612, USA
- Shriners Hospital for Children, Tampa, FL 33612, USA
- Correspondence: (J.H.); (M.S.K.); Tel.: +1-919-843-4389 (J.H.)
| |
Collapse
|
27
|
Chiasseu M, Fesharaki-Zadeh A, Saito T, Saido TC, Strittmatter SM. Gene-environment interaction promotes Alzheimer's risk as revealed by synergy of repeated mild traumatic brain injury and mouse App knock-in. Neurobiol Dis 2020; 145:105059. [PMID: 32858147 PMCID: PMC7572902 DOI: 10.1016/j.nbd.2020.105059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/17/2020] [Accepted: 08/20/2020] [Indexed: 11/20/2022] Open
Abstract
There is a strong unmet need for translational progress towards Alzheimer's disease (AD) modifying therapy. Unfortunately, preclinical modeling of the disease has been disappointing, relying primarily on transgenic mouse overexpression of rare dominant mutations. Clinical manifestation of AD symptoms is known to reflect interaction between environmental and genetic risks. Mild traumatic brain injury (mTBI) is an environmental risk for dementia, including Alzheimer's, but there has been limited mechanistic analysis of mTBI contribution to AD. Here, we investigate the interplay between mTBI and Aβ precursor protein gene mutation in AD pathogenesis. We employed a knock-in (KI) model of AD that expresses the Aß-containing exons from human APP bearing the Swedish and Iberian mutations, namely AppNL-F/NL-F mice. Without environmental risk, this genetic variation yields minimal mouse symptomatology. Anesthetized 4-month-old KI mice and their age-matched wild type (WT) controls were subjected to repeated mild closed head injury (rmCHI), once daily for 14 days. Anesthetized, uninjured genotype- and age-matched mice were used as sham controls. At 3- and 8-months post-injury, amyloid-β, phospho-tau and Iba1 expression in the injured KI cortices were assessed. Our data reveal that rmCHI enhances accumulation of amyloid-β and hyperphosphorylated tau inclusions, as well as neuroinflammation in AppNL-F/NL-F mice. Furthermore, novel object recognition and Morris water maze tests demonstrated that rmCHI greatly exacerbates persistent cognitive deficits in APPNL-F/NL-F mice. Therefore, study of gene-environment interaction demonstrates that combining risk factors provides a more robust model for AD, and that repeated mTBI substantially accelerates AD pathology in a genetically susceptible situation.
Collapse
Affiliation(s)
- Marius Chiasseu
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA; Departments of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Arman Fesharaki-Zadeh
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA; Departments of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-ku, Mizuho-cho, Nagoya, Aichi 467-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA; Departments of Neurology, Yale University School of Medicine, New Haven, CT, USA; Departments of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
28
|
Kim HN, Langley MR, Simon WL, Yoon H, Kleppe L, Lanza IR, LeBrasseur NK, Matveyenko A, Scarisbrick IA. A Western diet impairs CNS energy homeostasis and recovery after spinal cord injury: Link to astrocyte metabolism. Neurobiol Dis 2020; 141:104934. [PMID: 32376475 PMCID: PMC7982964 DOI: 10.1016/j.nbd.2020.104934] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/28/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
A diet high in fat and sucrose (HFHS), the so-called Western diet promotes metabolic syndrome, a significant co-morbidity for individuals with spinal cord injury (SCI). Here we demonstrate that the spinal cord of mice consuming HFHS expresses reduced insulin-like growth factor 1 (IGF-1) and its receptor and shows impaired tricarboxylic acid cycle function, reductions in PLP and increases in astrogliosis, all prior to SCI. After SCI, Western diet impaired sensorimotor and bladder recovery, increased microgliosis, exacerbated oligodendrocyte loss and reduced axon sprouting. Direct and indirect neural injury mechanisms are suggested since HFHS culture conditions drove parallel injury responses directly and indirectly after culture with conditioned media from HFHS-treated astrocytes. In each case, injury mechanisms included reductions in IGF-1R, SIRT1 and PGC-1α and were prevented by metformin. Results highlight the potential for a Western diet to evoke signs of neural insulin resistance and injury and metformin as a strategy to improve mechanisms of neural neuroprotection and repair.
Collapse
Affiliation(s)
- Ha Neui Kim
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Monica R Langley
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Whitney L Simon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Laurel Kleppe
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Ian R Lanza
- Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Nathan K LeBrasseur
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Aleksey Matveyenko
- Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America
| | - Isobel A Scarisbrick
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Rehabilitation Medicine Research Center, Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America; Neurosciuence Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, United States of America.
| |
Collapse
|
29
|
Balasubramanian N, Srivastava A, Pawar N, Sagarkar S, Sakharkar AJ. Repeated mild traumatic brain injury induces persistent variations in mitochondrial DNA copy number in mesocorticolimbic neurocircuitry of the rat. Neurosci Res 2020; 155:34-42. [DOI: 10.1016/j.neures.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022]
|
30
|
O'Brien WT, Pham L, Symons GF, Monif M, Shultz SR, McDonald SJ. The NLRP3 inflammasome in traumatic brain injury: potential as a biomarker and therapeutic target. J Neuroinflammation 2020; 17:104. [PMID: 32252777 PMCID: PMC7137518 DOI: 10.1186/s12974-020-01778-5] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/17/2020] [Indexed: 01/14/2023] Open
Abstract
There is a great clinical need to identify the underlying mechanisms, as well as related biomarkers, and treatment targets, for traumatic brain injury (TBI). Neuroinflammation is a central pathophysiological feature of TBI. NLRP3 inflammasome activity is a necessary component of the innate immune response to tissue damage, and dysregulated inflammasome activity has been implicated in a number of neurological conditions. This paper introduces the NLRP3 inflammasome and its implication in the pathogenesis of neuroinflammatory-related conditions, with a particular focus on TBI. Although its role in TBI has only recently been identified, findings suggest that priming and activation of the NLRP3 inflammasome are upregulated following TBI. Moreover, recent studies utilizing specific NLRP3 inhibitors have provided further evidence that this inflammasome is a major driver of neuroinflammation and neurobehavioral disturbances following TBI. In addition, there is emerging evidence that circulating inflammasome-associated proteins may have utility as diagnostic biomarkers of neuroinflammatory conditions, including TBI. Finally, novel and promising areas of research will be highlighted, including the potential involvement of the NLRP3 inflammasome in mild TBI, how factors such as biological sex may affect NLRP3 activity in TBI, and the use of emerging biomarker platforms. Taken together, this review highlights the exciting potential of the NLRP3 inflammasome as a target for treatments and biomarkers that may ultimately be used to improve TBI management.
Collapse
Affiliation(s)
- William T O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Louise Pham
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Georgia F Symons
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Mastura Monif
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC, 3004, Australia.,Department of Neurology, Melbourne Health, Melbourne, VIC, 3004, Australia.,Department of Physiology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia. .,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
31
|
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
|
32
|
Mild Hyperthermia Aggravates Glucose Metabolic Consequences in Repetitive Concussion. Int J Mol Sci 2020; 21:ijms21020609. [PMID: 31963504 PMCID: PMC7013838 DOI: 10.3390/ijms21020609] [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: 12/09/2019] [Revised: 01/06/2020] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of mortality and disability around the world. Mild TBI (mTBI) makes up approximately 80% of reported cases and often results in transient psychological abnormalities and cognitive disruption. At-risk populations for mTBI include athletes and other active individuals who may sustain repetitive concussive injury during periods of exercise and exertion when core temperatures are elevated. Previous studies have emphasized the impact that increased brain temperature has on adverse neurological outcomes. A lack of diagnostic tools to assess concussive mTBI limits the ability to effectively identify the post-concussive period during which the brain is uniquely susceptible to damage upon sustaining additional injury. Studies have suggested that a temporal window of increased vulnerability that exists corresponds to a period of injury-induced depression of cerebral glucose metabolism. In the current study, we sought to evaluate the relationship between repetitive concussion, local cerebral glucose metabolism, and brain temperature using the Marmarou weight drop model to generate mTBI. Animals were injured three consecutive times over a period of 7 days while exposed to either normothermic or hyperthermic temperatures for 15 min prior to and 1 h post each injury. A 14C-2-deoxy-d-glucose (2DG) autoradiography was used to measure local cerebral metabolic rate of glucose (lCMRGlc) in 10 diverse brain regions across nine bregma levels 8 days after the initial insult. We found that repetitive mTBI significantly decreased glucose utilization bilaterally in several cortical areas, such as the cingulate, visual, motor, and retrosplenial cortices, as well as in subcortical areas, including the caudate putamen and striatum, compared to sham control animals. lCMRGlc was significant in both normothermic and hyperthermic repetitive mTBI animals relative to the sham group, but to a greater degree when exposed to hyperthermic conditions. Taken together, we report significant injury-induced glucose hypometabolism after repetitive concussion in the brain, and additionally highlight the importance of temperature management in the acute period after brain injury.
Collapse
|
33
|
Harper MM, Woll AW, Evans LP, Delcau M, Akurathi A, Hedberg-Buenz A, Soukup DA, Boehme N, Hefti MM, Dutca LM, Anderson MG, Bassuk AG. Blast Preconditioning Protects Retinal Ganglion Cells and Reveals Targets for Prevention of Neurodegeneration Following Blast-Mediated Traumatic Brian Injury. Invest Ophthalmol Vis Sci 2019; 60:4159-4170. [PMID: 31598627 PMCID: PMC6785841 DOI: 10.1167/iovs.19-27565] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose The purpose of this study was to examine the effect of multiple blast exposures and blast preconditioning on the structure and function of retinal ganglion cells (RGCs), to identify molecular pathways that contribute to RGC loss, and to evaluate the role of kynurenine-3-monooxygenase (KMO) inhibition on RGC structure and function. Methods Mice were subjected to sham blast injury, one single blast injury, or three blast injuries separated by either 1 hour or 1 week, using a blast intensity of 20 PSI. To examine the effect of blast preconditioning, mice were subjected to sham blast injury, one single 20-PSI injury, or three blast injuries separated by 1 week (5 PSI, 5 PSI, 20 PSI and 5 PSI, 5 PSI, 5 PSI). RGC structure was analyzed by optical coherence tomography (OCT) and function was analyzed by the pattern electroretinogram (PERG). BRN3A-positive cells were quantified to determine RGC density. RNA-seq analysis was used to identify transcriptional changes between groups. Results Analysis of mice with multiple blast exposures of 20 PSI revealed no significant differences compared to one 20-pounds per square inch (PSI) exposure using OCT, PERG, or BRN3A cell counts. Analysis of mice exposed to two preconditioning 5-PSI blasts prior to one 20-PSI blast showed preservation of RGC structure and function. RNA-seq analysis of the retina identified multiple transcriptomic changes between conditions. Pharmacologic inhibition of KMO preserved RGC responses compared to vehicle-treated mice. Conclusions Preconditioning protects RGC from blast injury. Protective effects appear to involve changes in KMO activity, whose inhibition is also protective.
Collapse
Affiliation(s)
- Matthew M. Harper
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Addison W. Woll
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Lucy P. Evans
- Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, United States
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States
| | - Michael Delcau
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Abhigna Akurathi
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa
| | - Adam Hedberg-Buenz
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States
| | - Dana A. Soukup
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States
| | - Nickolas Boehme
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Marco M. Hefti
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States
| | - Laura M. Dutca
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - Michael G. Anderson
- The Iowa City Department of Veterans Affairs Medical Center, Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States
| | - Alexander G. Bassuk
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States
- Department of Neurology, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
34
|
Putnam LJ, Willes AM, Kalata BE, Disher ND, Brusich DJ. Expansion of a fly TBI model to four levels of injury severity reveals synergistic effects of repetitive injury for moderate injury conditions. Fly (Austin) 2019; 13:1-11. [PMID: 31524048 DOI: 10.1080/19336934.2019.1664363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Several million traumatic brain injury (TBI) events are reported in the United States annually. However, mild TBI events often go unreported, and mild and repetitive mild TBI conditions are challenging to model. Fruit flies (Drosophila melanogaster) have gained traction for the study of TBI. The best-characterized fly TBI model is the high-impact trauma (HIT) method. We replicated the HIT method and confirmed several previous findings at the standard level of injury severity. We then expanded upon the HIT model by characterizing mortality across three reduced levels of injury severity. Importantly, we found reduced mortality with reduced injury severity and synergistic effects on mortality in response to repetitive TBI by our moderate injury conditions. Last, we compared moderate, repetitive TBI to a single severe TBI via assessment of the pattern of mortality and geotaxis performance in the 24 h following TBI. We found the number and severity of injuries could result in different patterns of death, while all TBI conditions led to impaired geotaxis compared to uninjured flies at 0.5 h and 6 h post-TBI. Thus, we have extended a well-characterized model of TBI in flies, and shown the utility of this model for making unique insights into TBI across various severities, injury numbers, and time-points post-injury.
Collapse
Affiliation(s)
- Lauren J Putnam
- Human Biology, College of Science, Engineering, and Technology, University of Wisconsin - Green Bay, Green Bay, WI, USA
| | - Ashley M Willes
- Human Biology, College of Science, Engineering, and Technology, University of Wisconsin - Green Bay, Green Bay, WI, USA
| | - Brooke E Kalata
- Human Biology, College of Science, Engineering, and Technology, University of Wisconsin - Green Bay, Green Bay, WI, USA
| | - Nathaniel D Disher
- Human Biology, College of Science, Engineering, and Technology, University of Wisconsin - Green Bay, Green Bay, WI, USA
| | - Douglas J Brusich
- Human Biology, College of Science, Engineering, and Technology, University of Wisconsin - Green Bay, Green Bay, WI, USA
| |
Collapse
|
35
|
Kulkarni P, Morrison TR, Cai X, Iriah S, Simon N, Sabrick J, Neuroth L, Ferris CF. Neuroradiological Changes Following Single or Repetitive Mild TBI. Front Syst Neurosci 2019; 13:34. [PMID: 31427931 PMCID: PMC6688741 DOI: 10.3389/fnsys.2019.00034] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 07/10/2019] [Indexed: 11/13/2022] Open
Abstract
Objectives To test the hypothesis that there are differences in neuroradiological measures between single and repeated mild traumatic brain injury using multimodal MRI. Methods A closed-head momentum exchange model was used to produce one or three mild head injuries in young adult male rats compared to non-injured, age and weight-matched controls. Six-seven weeks post-injury, rats were studied for deficits in cognitive and motor function. Seven-eight weeks post-injury changes in brain anatomy and function were evaluated through analysis of high resolution T2 weighted images, resting-state BOLD functional connectivity, and diffusion weighted imaging with quantitative anisotropy. Results Head injuries occurred without skull fracture or signs of intracranial bleeding or contusion. There were no significant differences in cognitive or motors behaviors between experimental groups. With a single mild hit, the affected areas were limited to the caudate/putamen and central amygdala. Rats hit three times showed altered diffusivity in white matter tracts, basal ganglia, central amygdala, brainstem, and cerebellum. Comparing three hits to one hit showed a similar pattern of change underscoring a dose effect of repeated head injury on the brainstem and cerebellum. Disruption of functional connectivity was pronounced with three mild hits. The midbrain dopamine system, hippocampus, and brainstem/cerebellum showed hypoconnectivity. Interestingly, rats exposed to one hit showed enhanced functional connectivity (or hyperconnectivity) across brain sites, particularly between the olfactory system and the cerebellum. Interpretation Neuroradiological evidence of altered brain structure and function, particularly in striatal and midbrain dopaminergic areas, persists long after mild repetitive head injury. These changes may serve as biomarkers of neurodegeneration and risk for dementia later in life.
Collapse
Affiliation(s)
- Praveen Kulkarni
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Thomas R Morrison
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Xuezhu Cai
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Sade Iriah
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Neal Simon
- Azevan Pharmaceuticals, Bethlehem, PA, United States.,Department of Biological Sciences, College of Arts and Sciences, Lehigh University, Bethlehem, PA, United States
| | - Julia Sabrick
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Lucas Neuroth
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Craig F Ferris
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| |
Collapse
|
36
|
Rege SD, Royes L, Tsai B, Zhang G, Yang X, Gomez-Pinilla F. Brain Trauma Disrupts Hepatic Lipid Metabolism: Blame It on Fructose? Mol Nutr Food Res 2019; 63:e1801054. [PMID: 31087499 DOI: 10.1002/mnfr.201801054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/21/2019] [Indexed: 02/06/2023]
Abstract
SCOPE The action of brain disorders on peripheral metabolism is poorly understood. The impact of traumatic brain injury (TBI) on peripheral organ function and how TBI effects can be influenced by the metabolic perturbation elicited by fructose ingestion are studied. METHODS AND RESULTS It is found that TBI affects glucose metabolism and signaling proteins for insulin and growth hormone in the liver; these effects are exacerbated by fructose ingestion. Fructose, principally metabolized in the liver, potentiates the action of TBI on hepatic lipid droplet accumulation. Studies in isolated cultured hepatocytes identify GH and fructose as factors for the synthesis of lipids. The liver has a major role in the synthesis of lipids used for brain function and repair. TBI results in differentially expressed genes in the hypothalamus, primarily associated with lipid metabolism, providing cues to understand central control of peripheral alterations. Fructose-fed TBI animals have elevated levels of markers of inflammation, lipid peroxidation, and cell energy metabolism, suggesting the pro-inflammatory impact of TBI and fructose in the liver. CONCLUSION Results reveal the impact of TBI on systemic metabolism and the aggravating action of fructose. The hypothalamic-pituitary-growth axis seems to play a major role in the regulation of the peripheral TBI pathology.
Collapse
Affiliation(s)
- Shraddha D Rege
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Luiz Royes
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Centro De Educacao Fisica e Desportos, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, 97105, Brazil
| | - Brandon Tsai
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Guanglin Zhang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xia Yang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Department of Neurosurgery, UCLA Brain Injury Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
37
|
Abstract
The underlying mechanisms that result in neurophysiological changes and cognitive sequelae in the context of repetitive mild traumatic brain injury (rmTBI) remain poorly understood. Animal models provide a unique opportunity to examine cellular and molecular responses using histological assessment, which can give important insights on the neurophysiological changes associated with the evolution of brain injury. To better understand the potential cumulative effects of multiple concussions, the focus of animal models is shifting from single to repetitive head impacts. With a growing body of literature on this subject, a review and discussion of current findings is valuable to better understand the neuropathology associated with rmTBI, to evaluate the current state of the field, and to guide future research efforts. Despite variability in experimental settings, existing animal models of rmTBI have contributed to our understanding of the underlying mechanisms following repeat concussion. However, how to reconcile the various impact methods remains one of the major challenges in the field today.
Collapse
Affiliation(s)
- Wouter S Hoogenboom
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Clinical Investigation, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA.
| | - Craig A Branch
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Physiology and Biophysics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| | - Michael L Lipton
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Departments of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| |
Collapse
|
38
|
Marshall CM, Chan N, Tran P, DeMatteo C. The use of an intensive physical exertion test as a final return to play measure in concussed athletes: a prospective cohort. PHYSICIAN SPORTSMED 2019; 47:158-166. [PMID: 30372657 DOI: 10.1080/00913847.2018.1542258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To examine the utility of a novel physical exertion test developed by the Chicago Blackhawks medical staff as a final return to play (RTP) clearance test in youth and young adult athletes, and to determine the relationship between participant and test variables on RTP within asymptomatic athletes diagnosed with concussion. METHODS Once asymptomatic and following completion of all RTP steps, concussed athletes underwent the Gapski-Goodman Test (GGT) or modified GGT (mGGT) at partnered Complete Concussion Management Inc. (CCMI) clinics as part of RTP decision-making. Prospective data was collected electronically by trained CCMI clinicians utilizing the CCMI Concussion Database System. A review was conducted to examine data collected between January 2016 and February 2017. Participant and test variables were analyzed to determine relationships with pass/fail rate of the GGT/mGGT. RESULTS A total of 759 athletes performed the GGT/mGGT in the study period. Although all asymptomatic, 14.6% of concussed athletes failed the GGT/mGGT while attempting to achieve RTP clearance. Statistically significant relationships were found between failure of the test and symptom severity score on initial presentation and self-reported history of pre-morbid anxiety. When taken together, sex, age, and pre-morbid anxiety significantly predicted the length of time between injury and RTP clearance. CONCLUSION The GGT may identify individuals who are not ready to RTP despite a self-reported asymptomatic status and completion of all RTP steps. These results illustrate that RTP clearance decisions based on self-reported asymptomatic status at rest may be inadequate. Instead, monitored, intensive, sport-specific, physical exertion testing should be utilized to inform clinical RTP decisions.
Collapse
Affiliation(s)
- Cameron M Marshall
- a Department of Research , Complete Concussion Management Inc ., Oakville , ON , Canada
| | - Nicole Chan
- b School of Rehabilitation Sciences, Faculty of Health Sciences , McMaster University, Institute of Applied Health Sciences , Hamilton , ON , Canada
| | - Pauline Tran
- b School of Rehabilitation Sciences, Faculty of Health Sciences , McMaster University, Institute of Applied Health Sciences , Hamilton , ON , Canada
| | - Carol DeMatteo
- b School of Rehabilitation Sciences, Faculty of Health Sciences , McMaster University, Institute of Applied Health Sciences , Hamilton , ON , Canada
| |
Collapse
|
39
|
Bolton-Hall AN, Hubbard WB, Saatman KE. Experimental Designs for Repeated Mild Traumatic Brain Injury: Challenges and Considerations. J Neurotrauma 2019; 36:1203-1221. [PMID: 30351225 PMCID: PMC6479246 DOI: 10.1089/neu.2018.6096] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mild traumatic brain injury (mild TBI) is a growing public concern, as evidence mounts that even brain injuries classified as "mild" can result in persistent neurological dysfunction. Multiple brain injuries heighten the likelihood of worsened or more prolonged symptomatology and may trigger long-term neurodegeneration. Animal models provide a logical platform to identify key parameters, such as loading forces, duration between injuries, and number of injuries, which contribute to additive or synergistic damage after repeated mild TBI. Despite the tremendous increase in research productivity in the field of repeated mild TBI, relatively few studies have been designed in such a way as to provide experimental-based insights into the dependence of cellular and functional outcomes on the prescribed parameters of mild TBI. In this review, we summarize how standard models of TBI have been adapted to produce mild TBI and highlight commonly observed aspects of neuropathology replicated in rodent models of mild TBI. The complexity of designing studies of repeated TBI is discussed, including challenges of incorporating appropriate control groups, informative experimental design, and relevant outcome measures. We then feature studies that provide a well-controlled, within-study design varying either the number of injuries or the interinjury interval. Harnessing the power of experimental models of TBI to elucidate which injury parameters are critical contributors to acute and chronic damage after repeated injury can further efforts at prevention and provide improved models for testing mechanisms and therapeutic interventions.
Collapse
Affiliation(s)
- Amanda N. Bolton-Hall
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan
| | - W. Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
40
|
Holschneider DP, Guo Y, Wang Z, Vidal M, Scremin OU. Positive Allosteric Modulation of Cholinergic Receptors Improves Spatial Learning after Cortical Contusion Injury in Mice. J Neurotrauma 2019; 36:2233-2245. [PMID: 30688147 DOI: 10.1089/neu.2018.6036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We examined benzyl quinolone carboxylic acid (BQCA), a novel M1 muscarinic-positive allosteric modulator, for improving memory and motor dysfunction after cerebral cortical contusion injury (CCI). Adult mice received unilateral motorsensory cortical CCI or sham injury. Benzyl quinolone carboxylic acid (BQCA; 5, 10, and 20 mg/kg, intraperitoneally [i.p.] × 2/day × 3-4 weeks) or vehicle (Veh) were administered, and weekly evaluations were undertaken using a battery of motor tests, as well as the Morris water maze. Thereafter, cerebral metabolic activation was investigated in awake animals during walking with [14C]-2-deoxygIucose autoradiography, comparing CCI mice previously treated with BQCA (20 mg/kg) or vehicle. Relative changes in local cerebral glucose uptake (rCGU) were evaluated in three-dimensional-reconstructed brains using statistical parametric mapping. CCI resulted in mild hyperactivity in the open field, and modest significant motor deficits, as well as significantly decreased spatial learning at 3 weeks. BQCA in CCI mice resulted in significantly improved spatial recall during the third week, with minimal effects on motor outcomes. CCI significantly decreased rCGU in the ipsilesional basal ganglia-thalamocortical circuit and in somatosensory regions, with relative increases noted contralaterally, as well as in the cerebellum. Significant decreases in rCGU were noted in subregions of the ipsilesional hippocampal formation, with significant increases noted contralesionally. BQCA compared to vehicle-treated mice showed modest, though significantly increased, rCGU in motor regions, as well as a partial reversal of lesion-related rCGU findings in subregions of the hippocampal formation. rCGU in ipsilesional posterior CA1 demonstrated a significant inverse correlation with latency to find the submerged platform. BQCA at 20 mg/kg had no significant effect on general motor activity, body weight, or acute motor, secretory, or respiratory symptoms. Results suggest that BQCA is a candidate compound to improve learning and memory function after brain trauma and may not suffer the associated central nervous system side effects typically associated with even modest doses of other cholinergic enhancers.
Collapse
Affiliation(s)
- Daniel P Holschneider
- 1 Department of Psychiatry and the Behavioral Sciences and Biomedical Engineering, University of Southern California, Los Angeles, California.,2 Department of Neurology, Biomedical Engineering, University of Southern California, Los Angeles, California.,3 Greater Los Angeles VA Healthcare System, Los Angeles, California
| | - Yumei Guo
- 1 Department of Psychiatry and the Behavioral Sciences and Biomedical Engineering, University of Southern California, Los Angeles, California
| | - Zhuo Wang
- 1 Department of Psychiatry and the Behavioral Sciences and Biomedical Engineering, University of Southern California, Los Angeles, California
| | - Milagros Vidal
- 1 Department of Psychiatry and the Behavioral Sciences and Biomedical Engineering, University of Southern California, Los Angeles, California
| | - Oscar U Scremin
- 3 Greater Los Angeles VA Healthcare System, Los Angeles, California
| |
Collapse
|
41
|
Fehily B, Bartlett CA, Lydiard S, Archer M, Milbourn H, Majimbi M, Hemmi JM, Dunlop SA, Yates NJ, Fitzgerald M. Differential responses to increasing numbers of mild traumatic brain injury in a rodent closed-head injury model. J Neurochem 2019; 149:660-678. [PMID: 30702755 DOI: 10.1111/jnc.14673] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/20/2018] [Accepted: 01/14/2019] [Indexed: 01/13/2023]
Abstract
Following mild traumatic brain injury (mTBI), further mild impacts can exacerbate negative outcomes. To compare chronic damage and deficits following increasing numbers of repeated mTBIs, a closed-head weight-drop model of repeated mTBI was used to deliver 1, 2 or 3 mTBIs to adult female rats at 24 h intervals. Outcomes were assessed at 3 months following the first mTBI. No gross motor, sensory or reflex deficits were identified (p > 0.05), consistent with current literature. Cognitive function assessed using a Morris water maze revealed chronic memory deficits following 1 and 2, but not 3 mTBI compared to shams (p ≤ 0.05). Oxidative damage to DNA was assessed immunohistochemically in the dentate hilus of the hippocampus and splenium of the corpus callosum; no changes were observed. IBA1-positive microglia were increased in size in the cortex following 1 mTBI and in the corpus callosum following 2 mTBI compared to shams (p ≤ 0.05); no changes were observed in the dentate hilus. Glial fibrillary acidic protein (GFAP)-positive astrocyte immunoreactivity was assessed in all three brain regions and no chronic changes were observed. Integrity of myelin ultrastructure in the corpus callosum was assessed using transmission electron microscopy. G ratio was decreased following 2 mTBIs compared to shams (p ≤ 0.05) at post hoc level only. The changing patterns of damage and deficits following increasing numbers of mTBI may reflect dynamic responses to small numbers of mTBIs or a conditioning effect such that increasing numbers of mTBIs do not necessarily result in worsening pathology. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14508.
Collapse
Affiliation(s)
- Brooke Fehily
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Carole A Bartlett
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Stephen Lydiard
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Michael Archer
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Hannah Milbourn
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Maimuna Majimbi
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Bentley, WA, Australia
| | - Jan M Hemmi
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Sarah A Dunlop
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
| | - Nathanael J Yates
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, School of Biological Sciences, Crawley, WA, Australia
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Bentley, WA, Australia
- The Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute Building, Nedlands, WA, Australia
| |
Collapse
|
42
|
Bisicchia E, Sasso V, Molinari M, Viscomi MT. Plasticity of microglia in remote regions after focal brain injury. Semin Cell Dev Biol 2019; 94:104-111. [PMID: 30703556 DOI: 10.1016/j.semcdb.2019.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 02/06/2023]
Abstract
The CNS is endowed with an intrinsic ability to recover from and adapt secondary compensatory mechanisms to injury. The basis of recovery stems from brain plasticity, defined as the brain's ability to make adaptive changes on structural and functional levels, ranging from molecular, synaptic, and cellular changes in response to alterations in their environment. In this multitude of responses, microglia have an active role and contribute to brain plasticity through their dynamic responses. This review will provide an overview of microglial responses in the context of acute CNS injury and their function in post-traumatic repair and assess the changes that are induced by damage in remote areas from, but functionally connected to, the primary site of injury. In the second section, we highlight the effects of several therapeutic approaches, with particular interest paid to specialized pro-resolving lipid mediators, in modulating microglial responses in remote regions and enhancing long-term functional recovery via suppression of neurodegenerative cascades that are induced by damage, which may contribute to a translational bridge from bench to bedside.
Collapse
Affiliation(s)
- Elisa Bisicchia
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Valeria Sasso
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Marco Molinari
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Teresa Viscomi
- Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del S. Cuore, Rome, Italy.
| |
Collapse
|
43
|
Hubbard WB, Joseph B, Spry M, Vekaria HJ, Saatman KE, Sullivan PG. Acute Mitochondrial Impairment Underlies Prolonged Cellular Dysfunction after Repeated Mild Traumatic Brain Injuries. J Neurotrauma 2018; 36:1252-1263. [PMID: 30417732 DOI: 10.1089/neu.2018.5990] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mild traumatic brain injuries (mTBI), accounting for more than 80% of TBIs, can cause cognitive and behavioral impairments, the severity and duration of which increase after additional mTBIs. While mTBI does not cause widespread neuronal death, the mechanisms underlying increased cellular susceptibility to subsequent head impacts remain unknown. To investigate the hypothesis that altered mitochondrial bioenergetics underlie cellular vulnerability to repeated insults, we employed a mouse model of mild closed head injury (CHI) to examine mitochondrial function and oxidative stress, because these mechanisms are often intertwined. Mitochondrial respiration was assayed (Seahorse XFe24 Flux Analyzer) from cortex and hippocampus collected at 6 h, 24 h, 48 h, and 96 h post-injury. State III (adenosine diphosphate [ADP]-mediated) respiration was significantly decreased in the hippocampal mitochondria of the CHI group compared with sham at 48 h post-injury. Further, cortex-derived mitochondria exhibited a decrease in State III respiration at 24 h and 48 h post-injury. No significant differences were observed at 6 h or 96 h post-injury in either region of interest. A second CHI repeated either 48 h or 96 h after the first did not worsen State III respiration at 48 h after the final injury compared with a single CHI, but CHI repeated at a 48 h interval prolonged cortical mitochondrial dysfunction to 96 h after the final injury. Markers of oxidative stress were significantly elevated after two CHIs delivered 48 h apart, but not after single CHI or two CHI delivered 96 h apart. This study establishes that mTBI results in early mitochondrial dysfunction, which may be a determinant for cellular vulnerability to repeated head impacts. Thus, therapies targeting mitochondrial impairment could improve outcomes after repeated mTBI.
Collapse
Affiliation(s)
- W Brad Hubbard
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Binoy Joseph
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Malinda Spry
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Hemendra J Vekaria
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kathryn E Saatman
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky.,2 Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Patrick G Sullivan
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky.,3 Department of Neuroscience, University of Kentucky, Lexington, Kentucky.,4 Lexington VAMC, Lexington, Kentucky
| |
Collapse
|
44
|
McColl TJ, Brady RD, Shultz SR, Lovick L, Webster KM, Sun M, McDonald SJ, O'Brien TJ, Semple BD. Mild Traumatic Brain Injury in Adolescent Mice Alters Skull Bone Properties to Influence a Subsequent Brain Impact at Adulthood: A Pilot Study. Front Neurol 2018; 9:372. [PMID: 29887828 PMCID: PMC5980957 DOI: 10.3389/fneur.2018.00372] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/07/2018] [Indexed: 12/24/2022] Open
Abstract
Mild traumatic brain injuries (mTBI) are common during adolescence, and limited clinical evidence suggests that a younger age at first exposure to a mTBI may lead to worse long-term outcomes. In this study, we hypothesized that a mTBI during adolescence would predispose toward poorer neurobehavioral and neuropathological outcomes after a subsequent injury at adulthood. Mice received a mild weight drop injury (mTBI) at adolescence (postnatal day 35; P35) and/or at adulthood (P70). Mice were randomized to 6 groups: 'sham' (sham-surgery at P35 only); 'P35' (mTBI at P35 only); 'P35 + sham' (mTBI at P35 + sham at P70); 'sham + P70' (sham at P35 + mTBI at P70); 'sham + sham' (sham at both P35 and P70); or 'P35 + P70' (mTBI at both P35 and P70). Acute apnea and an extended righting reflex time confirmed a mTBI injury at P35 and/or P70. Cognitive, psychosocial, and sensorimotor function was assessed over 1-week post-injury. Injured groups performed similarly to sham controls across all tasks. Immunofluorescence staining at 1 week detected an increase in glial activation markers in Sham + P70 brains only. Strikingly, 63% of Sham + P70 mice exhibited a skull fracture at impact, compared to 13% of P35 + P70 mice. Micro computed tomography of parietal skull bones found that a mTBI at P35 resulted in increased bone volume and strength, which may account for the difference in fracture incidence. In summary, a single mTBI to the adolescent mouse brain did not exacerbate the cerebral effects of a subsequent mTBI in adulthood. However, the head impact at P35 induced significant changes in skull bone structure and integrity. These novel findings support future investigation into the consequences of mTBI on skull bone.
Collapse
Affiliation(s)
- Thomas J McColl
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Rhys D Brady
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Lauren Lovick
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Kyria M Webster
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Mujun Sun
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Terence J O'Brien
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Bridgette D Semple
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Abstract
Mild traumatic brain injury (mTBI) represents a significant public healthcare concern, accounting for the majority of all head injuries. While symptoms are generally transient, some patients go on to experience long-term cognitive impairments and additional mild impacts can result in exacerbated and persisting negative outcomes. To date, studies using a range of experimental models have reported chronic behavioral deficits in the presence of axonal injury and inflammation following repeated mTBI; assessments of oxidative stress and myelin pathology have thus far been limited. However, some models employed induced acute focal damage more suggestive of moderate–severe brain injury and are therefore not relevant to repeated mTBI. Given that the nature of mechanical loading in TBI is implicated in downstream pathophysiological changes, the mechanisms of damage and chronic consequences of single and repeated closed-head mTBI remain to be fully elucidated. This review covers literature on potential mechanisms of damage following repeated mTBI, integrating known mechanisms of pathology underlying moderate–severe TBIs, with recent studies on adult rodent models relevant to direct impact injuries rather than blast-induced damage. Pathology associated with excitotoxicity and cerebral blood flow-metabolism uncoupling, oxidative stress, cell death, blood-brain barrier dysfunction, astrocyte reactivity, microglial activation, diffuse axonal injury, and dysmyelination is discussed, followed by a summary of functional deficits and preclinical assessments of therapeutic strategies. Comprehensive characterization of the pathology underlying delayed and persisting deficits following repeated mTBI is likely to facilitate further development of therapeutic strategies to limit long-term sequelae.
Collapse
Affiliation(s)
- Brooke Fehily
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Melinda Fitzgerald
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia.,2 Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,3 Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
46
|
Abstract
Chronic traumatic encephalopathy (CTE) is a progressive neurodegenerative disease that presents as a late sequela from traumatic brain injury (TBI). TBI is a growing and under-recognized public health concern with a high degree of morbidity and large associated global costs. While the immune response to TBI is complex, its contribution to the development of CTE remains largely unknown. In this review, we summarize the current understanding of the link between CTE and the resident innate immune system of the brain-microglia. We discuss the neuropathology underlying CTE including the creation and aggregation of phosphorylated tau protein into neurofibrillary tangles and the formation of amyloid beta deposits. We also present how microglia, the resident innate immune cells of the brain, drive the continuous low-level inflammation associated with the insidious onset of CTE. In this review, we conclude that the latency period between the index brain injury and the long-term development of CTE presents an opportunity for therapeutic intervention. Encouraging advances with microtubule stabilizers, cis p-tau antibodies, and the ability to therapeutically alter the inflammatory state of microglia have shown positive results in both animal and human trials. Looking forward, recent advancements in next-generation sequencing technology for the study of genomic, transcriptomic, and epigenetic information will provide an opportunity for significant advancement in our understanding of prorepair and pro-injury gene signatures allowing for targeted intervention in this highly morbid injury process.
Collapse
|
47
|
Karelina K, Nicholson S, Weil ZM. Minocycline blocks traumatic brain injury-induced alcohol consumption and nucleus accumbens inflammation in adolescent male mice. Brain Behav Immun 2018; 69:532-539. [PMID: 29395778 PMCID: PMC6698899 DOI: 10.1016/j.bbi.2018.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 12/26/2022] Open
Abstract
Alcohol use is a well characterized risk factor for traumatic brain injury (TBI); however, emerging clinical and experimental research suggests that TBI may also be an independent risk factor for the development of alcohol use disorders. In particular, TBIs incurred early in life predict the development of problem alcohol use and increase vulnerability to neuroinflammation as a consequence of alcohol use. Critically, the neuroinflammatory response to alcohol, mediated in large part by microglia, may also function as a driver of further alcohol use. Here, we tested the hypothesis that TBI increases alcohol consumption through microglia-mediated neuroinflammation. Mice were injured as juveniles and alcohol consumption and preference were assessed in a free-choice voluntary drinking paradigm in adolescence. TBI increased alcohol consumption; however, treatment with minocycline, an inhibitor of microglial activation, reduced alcohol intake in TBI mice to sham levels. Moreover, a single injection of ethanol (2 g/kg) significantly increased microglial activation in the nucleus accumbens and microglial expression of the proinflammatory cytokine IL-1β in TBI, but not sham or minocycline-treated, mice. Our data implicate TBI-induced microglial activation as a possible mechanism for the development of alcohol use disorders.
Collapse
Affiliation(s)
- Kate Karelina
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Samuel Nicholson
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | | |
Collapse
|
48
|
Borniger JC, Ungerleider K, Zhang N, Karelina K, Magalang UJ, Weil ZM. Repetitive Brain Injury of Juvenile Mice Impairs Environmental Enrichment-Induced Modulation of REM Sleep in Adulthood. Neuroscience 2018; 375:74-83. [PMID: 29432885 DOI: 10.1016/j.neuroscience.2018.01.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 10/18/2022]
Abstract
Traumatic brain injuries (TBIs) are a common and costly ongoing public health concern. Injuries that occur during childhood development can have particularly profound and long-lasting effects. One common consequence and potential mediator of negative outcomes of TBI is sleep disruption which occurs in a substantial proportion of TBI patients. These individuals report greater incidences of insomnia and sleep fragmentation combined with a greater overall sleep requirement meaning that many patients are chronically sleep-deprived. We sought to develop an animal model of developmental TBI-induced sleep dysfunction. Specifically, we tested the hypothesis that early (postnatal day 21), repeated closed head injuries in Swiss-Webster mice, would impair basal and homeostatic sleep responses in adulthood. Further, we asked whether environmental enrichment (EE), a manipulation that improves functional recovery following TBI and has been shown to alter sleep physiology, would prevent TBI-induced sleep dysfunction and alter sleep-modulatory peptide expression. In contrast to our hypothesis, the mild, repeated head injury that we used did not significantly alter basal or homeostatic sleep responses in mice housed in standard laboratory conditions. Sham-injured mice housed in enriched environments exhibited enhanced rapid eye movement (REM) sleep and expression of the REM-promoting peptide pro-melanin-concentrating hormone, an effect that was not apparent in TBI mice housed in enriched environments. Thus, TBI blocked the REM-enhancing effects of EE. This work has important implications for the management and rehabilitation of the TBI patient population.
Collapse
Affiliation(s)
- Jeremy C Borniger
- Department of Neuroscience, Behavioral Neuroendocrinology Group, Neuroscience Research Institute, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kyra Ungerleider
- Department of Neuroscience, Behavioral Neuroendocrinology Group, Neuroscience Research Institute, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ning Zhang
- Department of Neuroscience, Behavioral Neuroendocrinology Group, Neuroscience Research Institute, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kate Karelina
- Department of Neuroscience, Behavioral Neuroendocrinology Group, Neuroscience Research Institute, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ulysses J Magalang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zachary M Weil
- Department of Neuroscience, Behavioral Neuroendocrinology Group, Neuroscience Research Institute, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
49
|
Wu LC, Kuo C, Loza J, Kurt M, Laksari K, Yanez LZ, Senif D, Anderson SC, Miller LE, Urban JE, Stitzel JD, Camarillo DB. Detection of American Football Head Impacts Using Biomechanical Features and Support Vector Machine Classification. Sci Rep 2017; 8:855. [PMID: 29321637 PMCID: PMC5762632 DOI: 10.1038/s41598-017-17864-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/01/2017] [Indexed: 12/27/2022] Open
Abstract
Accumulation of head impacts may contribute to acute and long-term brain trauma. Wearable sensors can measure impact exposure, yet current sensors do not have validated impact detection methods for accurate exposure monitoring. Here we demonstrate a head impact detection method that can be implemented on a wearable sensor for detecting field football head impacts. Our method incorporates a support vector machine classifier that uses biomechanical features from the time domain and frequency domain, as well as model predictions of head-neck motions. The classifier was trained and validated using instrumented mouthguard data from collegiate football games and practices, with ground truth data labels established from video review. We found that low frequency power spectral density and wavelet transform features (10~30 Hz) were the best performing features. From forward feature selection, fewer than ten features optimized classifier performance, achieving 87.2% sensitivity and 93.2% precision in cross-validation on the collegiate dataset (n = 387), and over 90% sensitivity and precision on an independent youth dataset (n = 32). Accurate head impact detection is essential for studying and monitoring head impact exposure on the field, and the approach in the current paper may help to improve impact detection performance on wearable sensors.
Collapse
Affiliation(s)
| | | | | | - Mehmet Kurt
- Stevens Institute of Technology, Hoboken, NJ, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tremoleda JL, Alvarez K, Aden A, Donnan R, Michael-Titus AT, Tomlins PH. Heart-rate sensitive optical coherence angiography for measuring vascular changes due to posttraumatic brain injury in mice. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:1-6. [PMID: 29210221 DOI: 10.1117/1.jbo.22.12.121710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/09/2017] [Indexed: 06/07/2023]
Abstract
Traumatic brain injury (TBI) results in direct vascular disruption, triggering edema, and reduction in cerebral blood flow. Therefore, understanding the pathophysiology of brain microcirculation following TBI is important for the development of effective therapies. Optical coherence angiography (OCA) is a promising tool for evaluating TBI in rodent models. We develop an approach to OCA that uses the heart-rate frequency to discriminate between static tissue and vasculature. This method operates on intensity data and is therefore not phase sensitive. Furthermore, it does not require spatial overlap of voxels and thus can be applied to pre-existing datasets for which oversampling may not have been explicitly considered. Heart-rate sensitive OCA was developed for dynamic assessment of mouse microvasculature post-TBI. Results show changes occurring at 5-min intervals within the first 50 min of injury.
Collapse
Affiliation(s)
- Jordi L Tremoleda
- Queen Mary University of London, Blizard Institute, Barts and the London School of Medicine and Dent, United Kingdom
| | - Karl Alvarez
- Queen Mary University of London, School of Electrical Engineering and Computer Science, London, United Kingdom
| | - Abdirahman Aden
- Queen Mary University of London, Institute of Dentistry, Barts and the London School of Medicine and, United Kingdom
| | - Robert Donnan
- Queen Mary University of London, School of Electrical Engineering and Computer Science, London, United Kingdom
| | - Adina T Michael-Titus
- Queen Mary University of London, Blizard Institute, Barts and the London School of Medicine and Dent, United Kingdom
| | - Peter H Tomlins
- Queen Mary University of London, Institute of Dentistry, Barts and the London School of Medicine and, United Kingdom
| |
Collapse
|