1
|
Danielewicz J, Llamosas N, Durá I, de Souza DB, Rodrigues S, Encinas-Pérez JM, Mateos DM. Biphasic changes in hippocampal granule cells after traumatic brain injury. Exp Neurol 2025; 390:115281. [PMID: 40294739 DOI: 10.1016/j.expneurol.2025.115281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/02/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
Traumatic brain injury (TBI) leads to a wide range of long-lasting physical and cognitive impairments. Changes in neuronal excitability and synaptic functions in the hippocampus have been proposed to underlie cognitive alterations. The dentate gyrus (DG) acts as a "gatekeeper" of hippocampal information processing and as a filter of excessive or aberrant input activity. In this study, we investigated the effects of controlled cortical impact, a model of TBI, on the excitability of granule cells (GCs) and spontaneous excitatory postsynaptic currents (sEPSCs) in the DG at three time points, 3 days, 15 days and 4 months after the injury in male and female mice. Our results indicate that changes in the short term are related to intrinsic properties, while changes in the long term are more related to input and synaptic activity, in agreement with the notion that TBI-related pathology courses with an acute phase and a later long-term secondary phase. A biphasic response, a reduction in the shorter term and an increase in the long term, was found in TBI neurons in the frequency of sEPSCs. These changes correlated with a loss of complexity in the pattern of the synaptic input, an alteration that could therefore play a role in the chronic and recurrent TBI-associated hyperexcitation.
Collapse
Affiliation(s)
- Joanna Danielewicz
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, Spain; BCAM Basque Center for Applied Mathematics, Bilbao, Bizkaia, Spain
| | - Nerea Llamosas
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, Spain; IKERBASQUE, The Basque Science Foundation, Bilbao, Bizkaia, Spain.
| | - Irene Durá
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, Spain; University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | | | - Serafim Rodrigues
- BCAM Basque Center for Applied Mathematics, Bilbao, Bizkaia, Spain; University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; IKERBASQUE, The Basque Science Foundation, Bilbao, Bizkaia, Spain
| | - Juan Manuel Encinas-Pérez
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, Spain; University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; IKERBASQUE, The Basque Science Foundation, Bilbao, Bizkaia, Spain
| | - Diego Martin Mateos
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, Spain; Consejo Nacional de Ciencia y Técnica (CONICET), Santa Fe, Argentina.
| |
Collapse
|
2
|
Nasir F, Yadav P, Sivanandam TM. NaHS alters synaptic plasticity proteins and enhances dendritic arborization to improve cognitive and motor deficits after traumatic brain injury in mice. Br J Pharmacol 2025; 182:1183-1205. [PMID: 39562524 DOI: 10.1111/bph.17386] [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: 11/08/2023] [Revised: 09/12/2024] [Accepted: 10/01/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Traumatic brain injury (TBI) is a complex medical condition affecting people globally. Hydrogen sulfide (H2S) is a recently discovered gaseous mediator and is dysregulated in the brain after TBI. Sodium hydrogen sulfide (NaHS), a known donor of H2S, is beneficial in various biological processes involving aging and diseases, including injury. It is neuroprotective against oxidative stress, neuroinflammation, and other secondary injury processes. However, the NaHS-H2S system has not been investigated as a regulator of injury-mediated synaptic plasticity proteins and the underlying mechanisms after TBI. EXPERIMENTAL APPROACH We developed a model of TBI in Swiss albino mice to study the effects of exogenous H2S, administered as NaHS. We assessed cognitive function (Barnes maze and novel object recognition) and motor function (rotarod). Brain tissue was analysed with ELISA, qRT-PCR, immunoblotting, Golgi-cox staining, and immunofluorescence. KEY RESULTS NaHS administration restored the injury-caused decline in H2S levels. Injury-mediated oxidative stress parameters were improved following NaHS. It down-regulated TBI biomarkers, ameliorated the synaptic marker proteins, and improved cognitive and motor deficits. These changes were accompanied by enhanced dendritic arborization and spine number. Restoration of N-methyl D-aspartate receptor subunits and diminished glutamate and calcium levels, along with marked changes in microtubule-associated protein 2 A and calcium/calmodulin-dependent protein kinase II, formed the basis of the underlying mechanism(s). CONCLUSION AND IMPLICATIONS Our findings suggest that NaHS could have therapeutic activity against TBI, as it ameliorated cognitive and motor deficits caused by changes in synaptic plasticity proteins and dendritic arborisation, in our model.
Collapse
Affiliation(s)
- Farheen Nasir
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Priyanka Yadav
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Thamil Mani Sivanandam
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
3
|
Chahin M, Mutschler J, Dzhuleva SP, Dieterle C, Jimenez LR, Bhattarai SR, Van Steenbergen V, Bareyre FM. Repetitive concussions promote microglia-mediated engulfment of presynaptic excitatory input associated with cognitive dysfunction. Commun Biol 2025; 8:335. [PMID: 40021832 PMCID: PMC11871131 DOI: 10.1038/s42003-025-07729-1] [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: 07/09/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
Concussions are a current health concern and account for the vast majority of head trauma. While symptoms after a single impact are usually transient, repetitive concussions, as often occur in sports, are responsible for persistent acute and chronic deficits. Here, we used a model of bilateral midline-centered concussions in mice to show that repetitive concussions selectively induce impairments in learning ability compared to single-impact injuries. Since microglial cells and their activation are considered key factors in degenerative pathology after brain trauma, we examined their structure and function after single and repetitive concussions in the cortex underlying the concussions and in the hippocampus. We found that only repetitive concussions led to a significant long-lasting structural activation of microglia and an increase in microglia-mediated engulfment of presynaptic excitatory synapses, while the elimination of inhibitory synapses was not altered. Since the density of excitatory input did not change during the 6-week study period, we hypothesize that there is a turnover of excitatory synapses following repetitive concussion that can be compensated for, anatomically but not behaviorally.
Collapse
Affiliation(s)
- Maryam Chahin
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Planegg-Martinsried, Germany
| | - Julius Mutschler
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Stephanie P Dzhuleva
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Clara Dieterle
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Leidy Reyes Jimenez
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Srijan Raj Bhattarai
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Valerie Van Steenbergen
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany.
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
4
|
Bayat Tork MA, Saberifar M, Joneidi Yekta H, Hajinejad M, Hosseini Ravandi H, Gorji A, Sahab Negah S. Nano-scaffold containing functional motif of stromal cell-derived factor 1 enhances neural stem cell behavior and synaptogenesis in traumatic brain injury. Sci Rep 2025; 15:5811. [PMID: 39962142 PMCID: PMC11832925 DOI: 10.1038/s41598-025-85698-5] [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: 04/23/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and morbidity worldwide, presenting a significant challenge due to the lack of effective therapies. Neural stem cells (NSCs) have shown promising potential in preclinical studies as a therapy for TBI. However, their application is limited by challenges related to poor survival and integration within the injured brain. This study investigated the effect of a novel nano-scaffold containing stromal cell-derived factor 1 (SDF-1) on NSC behavior and synaptogenesis after TBI. Using an innovative design, we successfully fabricated a nano-scaffold with Young's modulus of approximately 3.21 kPa, which aligns closely with the mechanical properties exhibited by neural tissue. This achievement marks the first time such a scaffold has been created and has promising implications for its potential use in neural tissue engineering applications. Our findings demonstrate that the nano-scaffold enhances NSC proliferation, migration, and differentiation capacity in vitro. Moreover, when transplanted into the injured brain, the nano-scaffold promotes the survival and integration of NSCs, leading to increased synaptogenesis and functional recovery. These findings suggest that using the novel nano-scaffold containing SDF-1 could provide a promising approach to treating TBI by improving NSC behavior and promoting synaptogenesis.
Collapse
Affiliation(s)
- Mohammad Amin Bayat Tork
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Saberifar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Joneidi Yekta
- New Technologies Research Center, Amirkabir University of Technology, Tehran, Iran
| | - Mehrdad Hajinejad
- Qaen Faculty of Medical Science, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery, Westfälische Wilhelms-Universität, 48149, Munster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, 48149, Munster, Germany.
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Campbell KR, Antonellis P, Peterka RJ, Wilhelm JL, Scanlan KT, Pettigrew NC, Chen S, Parrington L, Fino PC, Chesnutt JC, Horak FB, Hullar TE, King LA. In People With Subacute Mild Traumatic Brain Injury, Earlier Physical Therapy Improved Symptoms at a Faster Rate Than Later Physical Therapy: Randomized Controlled Trial. Phys Ther 2025; 105:pzae180. [PMID: 39693261 PMCID: PMC11878761 DOI: 10.1093/ptj/pzae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/28/2024] [Accepted: 09/14/2024] [Indexed: 12/20/2024]
Abstract
IMPORTANCE There is unclear evidence on when to initiate physical therapy after mild traumatic brain injury (mTBI) in a non-athlete, adult population. OBJECTIVE The objective of this study was to investigate physical therapy timing after mTBI through changes in patient-reported and clinically-assessed tools and objective and mechanism measurements of sensorimotor balance control. DESIGN This study was an investigator-blinded randomized control trial (NCT03479541). SETTING The study took place at an academic research center. PARTICIPANTS Two hundred and three participants were randomized to earlier physical therapy (n = 82) or to later physical therapy (n = 121). INTERVENTION After enrollment, the earlier physical therapy group started rehabilitation within 1 week and the later group started rehabilitation after a 6-week wait period. All participants received similar rehabilitation; 6-week program administered and progressed by licensed physical therapists. MAIN OUTCOMES AND MEASURES The primary outcome was the Dizziness Handicap Inventory (DHI). Secondary outcomes included common patient-reported/clinical assessments of mTBI and objective/mechanism measurements of balance, including novel measures of central sensorimotor integration. Differences between and within the groups on outcomes were examined with linear mixed-effect models, t tests, and effect sizes. RESULTS While both groups significantly improved and reached similar levels on patient-reported outcomes (DHI and secondary outcomes), the earlier physical therapy group had significantly larger and faster rates of improvement compared to later physical therapy. There were differential effects of physical therapy timing on the objective/mechanism-measured outcomes. Specifically, there were significant improvements in sensorimotor time delay for the earlier physical therapy group and no change in the later group. Further, the later group worsened in the motor activation components for balance control while there was no change in the early group. CONCLUSION AND RELEVANCE Earlier physical therapy after mTBI can improve symptoms at a faster rate relative to later physical therapy. Earlier physical therapy also showed improvements in sensorimotor aspects of balance control, not seen in the later group. There may be an important window to address central sensorimotor deficits after mTBI.
Collapse
Affiliation(s)
- Kody R Campbell
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Injury Surveillance Program, Datalys Center for Sports Injury Research and Prevention, Indianapolis, IN, United States
| | - Prokopios Antonellis
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Robert J Peterka
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- National Center for Rehabilitative Auditory Research (NCRAR), Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Jennifer L Wilhelm
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- National Center for Rehabilitative Auditory Research (NCRAR), Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Kathleen T Scanlan
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Natalie C Pettigrew
- National Center for Rehabilitative Auditory Research (NCRAR), Veterans Affairs Portland Health Care System, Portland, OR, United States
- Center for Regenerative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Siting Chen
- School of Public Health, Oregon Health & Science University, Portland, OR, United States
| | - Lucy Parrington
- Department of Sport, Exercise and Nutrition Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Peter C Fino
- Department of Health and Kinesiology, University of Utah, Salt Lake City, UT, United States
| | - James C Chesnutt
- Department of Family Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Fay B Horak
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Timothy E Hullar
- National Center for Rehabilitative Auditory Research (NCRAR), Veterans Affairs Portland Health Care System, Portland, OR, United States
- Department of Otolaryngology-Head and Neck Surgery, Oregon Health & Science University, Portland, OR, United States
| | - Laurie A King
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- National Center for Rehabilitative Auditory Research (NCRAR), Veterans Affairs Portland Health Care System, Portland, OR, United States
| |
Collapse
|
6
|
Shukla S, Comerci CJ, Süel GM, Jahed Z. Bioelectronic tools for understanding the universal language of electrical signaling across species and kingdoms. Biosens Bioelectron 2025; 267:116843. [PMID: 39426280 DOI: 10.1016/j.bios.2024.116843] [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: 07/24/2023] [Revised: 09/10/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
Modern bioelectronic tools are rapidly advancing to detect electric potentials within networks of electrogenic cells, such as cardiomyocytes, neurons, and pancreatic beta cells. However, it is becoming evident that electrical signaling is not limited to the animal kingdom but may be a universal form of cell-cell communication. In this review, we discuss the existing evidence of, and tools used to collect, subcellular, single-cell and network-level electrical signals across kingdoms, including bacteria, plants, fungi, and even viruses. We discuss how cellular networks employ altered electrical "circuitry" and intercellular mechanisms across kingdoms, and we assess the functionality and scalability of cutting-edge nanobioelectronics to collect electrical signatures regardless of cell size, shape, or function. Researchers today aim to design micro- and nano-topographic structures which harness mechanosensitive membrane and cytoskeletal pathways that enable tight electrical coupling to subcellular compartments within high-throughput recording systems. Finally, we identify gaps in current knowledge of inter-species and inter-kingdom electrical signaling and propose critical milestones needed to create a central theory of electrical signaling across kingdoms. Our discussion demonstrates the need for high resolution, high throughput tools which can probe multiple, diverse cell types at once in their native or experimentally-modeled environments. These advancements will not only reveal the underlying biophysical laws governing the universal language of electrical communication, but can enable bidirectional electrical communication and manipulation of biological systems.
Collapse
Affiliation(s)
- Shivani Shukla
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, United States; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, United States
| | - Colin J Comerci
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | - Gürol M Süel
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | - Zeinab Jahed
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, United States; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
7
|
May HG, Tsikonofilos K, Donat CK, Sastre M, Kozlov AS, Sharp DJ, Bruyns-Haylett M. EEG hyperexcitability and hyperconnectivity linked to GABAergic inhibitory interneuron loss following traumatic brain injury. Brain Commun 2024; 6:fcae385. [PMID: 39605970 PMCID: PMC11600960 DOI: 10.1093/braincomms/fcae385] [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: 03/13/2024] [Revised: 09/04/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
Traumatic brain injury represents a significant global health burden and has the highest prevalence among neurological disorders. Even mild traumatic brain injury can induce subtle, long-lasting changes that increase the risk of future neurodegeneration. Importantly, this can be challenging to detect through conventional neurological assessment. This underscores the need for more sensitive diagnostic tools, such as electroencephalography, to uncover opportunities for therapeutic intervention. Progress in the field has been hindered by a lack of studies linking mechanistic insights at the microscopic level from animal models to the macroscale phenotypes observed in clinical imaging. Our study addresses this gap by investigating a rat model of mild blast traumatic brain injury using both immunohistochemical staining of inhibitory interneurons and translationally relevant electroencephalography recordings. Although we observed no pronounced effects immediately post-injury, chronic time points revealed broadband hyperexcitability and increased connectivity, accompanied by decreased density of inhibitory interneurons. This pattern suggests a disruption in the balance between excitation and inhibition, providing a crucial link between cellular mechanisms and clinical hallmarks of injury. Our findings have significant implications for the diagnosis, monitoring, and treatment of traumatic brain injury. The emergence of electroencephalography abnormalities at chronic time points, despite the absence of immediate effects, highlights the importance of long-term monitoring in traumatic brain injury patients. The observed decrease in inhibitory interneuron density offers a potential cellular mechanism underlying the electroencephalography changes and may represent a target for therapeutic intervention. This study demonstrates the value of combining cellular-level analysis with macroscale neurophysiological recordings in animal models to elucidate the pathophysiology of traumatic brain injury. Future research should focus on translating these findings to human studies and exploring potential therapeutic strategies targeting the excitation-inhibition imbalance in traumatic brain injury.
Collapse
Affiliation(s)
- Hazel G May
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Konstantinos Tsikonofilos
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 65, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 65, Sweden
| | - Cornelius K Donat
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
- Department of Medicinal Radiochemistry, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Andriy S Kozlov
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - David J Sharp
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Michael Bruyns-Haylett
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
- Department of Bioengineering, Institut Quimic de Sarria, Universitat Ramon Llul, Barcelona 08017, Spain
- Department of Quantitative Methods, Institut Quimic de Sarria, Universitat Ramon Llul, Barcelona 08017, Spain
| |
Collapse
|
8
|
Chan A, Ouyang J, Nguyen K, Jones A, Basso S, Karasik R. Traumatic brain injuries: a neuropsychological review. Front Behav Neurosci 2024; 18:1326115. [PMID: 39444788 PMCID: PMC11497466 DOI: 10.3389/fnbeh.2024.1326115] [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: 10/22/2023] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
The best predictor of functional outcome in victims of traumatic brain injury (TBI) is a neuropsychological evaluation. An exponential growth of research into TBI has focused on diagnosis and treatment. Extant literature lacks a comprehensive neuropsychological review that is simultaneously scholarly and practical. In response, our group included, and went beyond a general overview of TBI's, which commonly include definition, types, severity, and pathophysiology. We incorporate reasons behind the use of particular neuroimaging techniques, as well as the most recent findings on common neuropsychological assessments conducted in TBI cases, and their relationship to outcome. In addition, we include tables outlining estimated recovery trajectories of different age groups, their risk factors and we encompass phenomenological studies, further covering the range of existing-promising tools for cognitive rehabilitation/remediation purposes. Finally, we highlight gaps in current research and directions that would be beneficial to pursue.
Collapse
Affiliation(s)
- Aldrich Chan
- Graduate School of Education and Psychology, Pepperdine University, Los Angeles, CA, United States
- Center for Neuropsychology and Consciousness, Miami, FL, United States
| | - Jason Ouyang
- Graduate School of Education and Psychology, Pepperdine University, Los Angeles, CA, United States
- Center for Neuropsychology and Consciousness, Miami, FL, United States
| | - Kristina Nguyen
- Graduate School of Education and Psychology, Pepperdine University, Los Angeles, CA, United States
- Center for Neuropsychology and Consciousness, Miami, FL, United States
| | - Aaliyah Jones
- Graduate School of Education and Psychology, Pepperdine University, Los Angeles, CA, United States
- Center for Neuropsychology and Consciousness, Miami, FL, United States
| | - Sophia Basso
- Graduate School of Education and Psychology, Pepperdine University, Los Angeles, CA, United States
- Center for Neuropsychology and Consciousness, Miami, FL, United States
| | - Ryan Karasik
- Graduate School of Education and Psychology, Pepperdine University, Los Angeles, CA, United States
- Center for Neuropsychology and Consciousness, Miami, FL, United States
| |
Collapse
|
9
|
Parthimos TP, Schulpis KH, Karousi AD, Loukas YL, Dotsikas Y. The relationship between neurotransmission-related amino acid blood concentrations and neuropsychological performance following acute exercise. APPLIED NEUROPSYCHOLOGY. ADULT 2024; 31:560-574. [PMID: 35227132 DOI: 10.1080/23279095.2022.2043327] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Amino acid neurotransmitters, including glutamate, phenylalanine, tyrosine, alanine, and glycine, underlie the majority of the excitatory and inhibitory neurotransmission in the nervous system, and acute exercise has been shown to modulate their concentrations. We aimed to determine whether any correlation exists between the above-mentioned amino acid blood concentrations and the neuropsychological performance after an acute exercise intervention. Sixty basketball players were randomly assigned to one of two experimental conditions: exercise or inactive resting. All participants underwent a comprehensive neuropsychological assessment and blood samples were taken on a Guthrie card before and after the end of the experimental conditions. Amino acid blood concentrations were significantly elevated and cognitive performance significantly improved post-exercise on specific neuropsychological assessments. Significant intervention × group interaction effects were apparent for Trail Making Test part-B [F(1,58) = 20.46, p < .0001, η2 = .26] and Digit Span Backwards [F(1,58) = 15.47, p < .0001, η2 = .21] neuropsychological assessments. Additionally, regression analysis indicated that tyrosine accounted for 38.0% of the variance in the Trail Making Test part-A test. These results suggest that elevated blood concentrations of neurotransmission-related amino acids are associated with improved neuropsychological performance after a single bout of high-intensity exercise.
Collapse
Affiliation(s)
- Theodore P Parthimos
- Division of Psychology, Faculty of Life and Health Sciences, De Montfort University, Leicester, UK
| | - Kleopatra H Schulpis
- Institute of Child Health, Research Center, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Alexandra D Karousi
- Department of Psychology, Human Sciences Research Centre, College of Life and Natural Sciences, University of Derby, Derby, UK
| | - Yannis L Loukas
- Laboratory of Pharm. Analysis, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Yannis Dotsikas
- Laboratory of Pharm. Analysis, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Borucki DM, Rohrer B, Tomlinson S. Complement propagates visual system pathology following traumatic brain injury. J Neuroinflammation 2024; 21:98. [PMID: 38632569 PMCID: PMC11022420 DOI: 10.1186/s12974-024-03098-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is associated with the development of visual system disorders. Visual deficits can present with delay and worsen over time, and may be associated with an ongoing neuroinflammatory response that is known to occur after TBI. Complement system activation is strongly associated with the neuroinflammatory response after TBI, but whether it contributes to vision loss after TBI is unexplored. METHODS Acute and chronic neuroinflammatory changes within the dorsal lateral geniculate nucleus (dLGN) and retina were investigated subsequent to a moderate to severe murine unilateral controlled cortical impact. Neuroinflammatory and histopathological outcomes were interpreted in the context of behavioral and visual function data. To investigate the role of complement, cohorts were treated after TBI with the complement inhibitor, CR2-Crry. RESULTS At 3 days after TBI, complement component C3 was deposited on retinogeniculate synapses in the dLGN both ipsilateral and contralateral to the lesion, which was reduced in CR2-Crry treated animals. This was associated with microglia morphological changes in both the ipsilateral and contralateral dLGN, with a less ramified phenotype in vehicle compared to CR2-Crry treated animals. Microglia in vehicle treated animals also had a greater internalized VGlut2 + synaptic volume after TBI compared to CR2-Crry treated animals. Microglia morphological changes seen acutely persisted for at least 49 days after injury. Complement inhibition also reduced microglial synaptic internalization in the contralateral dLGN and increased the association between VGLUT2 and PSD95 puncta, indicating preservation of intact synapses. Unexpectedly, there were no changes in the thickness of the inner retina, retinal nerve fiber layer or retinal ganglion layer. Neuropathological changes in the dLGN were accompanied by reduced visual acuity at subacute and chronic time points after TBI, with improvement seen in CR2-Crry treated animals. CONCLUSION TBI induces complement activation within the dLGN and promotes microglial activation and synaptic internalization. Complement inhibition after TBI in a clinically relevant paradigm reduces complement activation, maintains a more surveillance-like microglia phenotype, and preserves synaptic density within the dLGN. Together, the data indicate that complement plays a key role in the development of visual deficits after TBI via complement-dependent microglial phagocytosis of synapses within the dLGN.
Collapse
Affiliation(s)
- Davis M Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Baerbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA.
- Ralph Johnson VA Medical Center, Charleston, SC, USA.
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Ralph Johnson VA Medical Center, Charleston, SC, USA.
| |
Collapse
|
11
|
Jiang H, Giarratana AO, Theis T, Nagaraj V, Zhou X, Thakker-Varia S, Schachner M, Alder J. Single Nucleotide Polymorphism in Cell Adhesion Molecule L1 Affects Learning and Memory in a Mouse Model of Traumatic Brain Injury. Int J Mol Sci 2024; 25:3043. [PMID: 38474289 DOI: 10.3390/ijms25053043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
The L1 cell adhesion molecule (L1) has demonstrated a range of beneficial effects in animal models of spinal cord injury, neurodegenerative disease, and ischemia; however, the role of L1 in TBI has not been fully examined. Mutations in the L1 gene affecting the extracellular domain of this type 1 transmembrane glycoprotein have been identified in patients with L1 syndrome. These patients suffer from hydrocephalus, MASA (mental retardation, adducted thumbs, shuffling gait, aphasia) symptoms, and corpus callosum agenesis. Clinicians have observed that recovery post-traumatic brain injury (TBI) varies among the population. This variability may be explained by the genetic differences present in the general population. In this study, we utilized a novel mouse model of L1 syndrome with a mutation at aspartic acid position 201 in the extracellular domain of L1 (L1-201). We assessed the impact of this specific single nucleotide polymorphism (SNP) localized to the X-chromosome L1 gene on recovery outcomes following TBI by comparing the L1-201 mouse mutants with their wild-type littermates. We demonstrate that male L1-201 mice exhibit significantly worse learning and memory outcomes in the Morris water maze after lateral fluid percussion (LFP) injury compared to male wild-type mice and a trend to worse motor function on the rotarod. However, no significant changes were observed in markers for inflammatory responses or apoptosis after TBI.
Collapse
Affiliation(s)
- Haoyu Jiang
- Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Anna O Giarratana
- Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Thomas Theis
- Cell Biology and Neuroscience, Rutgers School of Arts and Sciences, Piscataway, NJ 08854, USA
| | - Vini Nagaraj
- Cell Biology and Neuroscience, Rutgers School of Arts and Sciences, Piscataway, NJ 08854, USA
| | - Xiaofeng Zhou
- Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Smita Thakker-Varia
- Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Melitta Schachner
- Cell Biology and Neuroscience, Rutgers School of Arts and Sciences, Piscataway, NJ 08854, USA
| | - Janet Alder
- Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| |
Collapse
|
12
|
Borucki D, Rohrer B, Tomlinson S. Complement propagates visual system pathology following traumatic brain injury. RESEARCH SQUARE 2024:rs.3.rs-3970621. [PMID: 38464312 PMCID: PMC10925413 DOI: 10.21203/rs.3.rs-3970621/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Traumatic brain injury (TBI) is associated with the development of visual system disorders. Visual deficits can present with delay and worsen over time, and may be associated with an ongoing neuroinflammatory response that is known to occur after TBI. Complement activation is strongly associated with the neuroinflammatory response after TBI, but whether it contributes to vision loss after TBI is unexplored. Methods Acute and chronic neuroinflammatory changes within the dorsal lateral geniculate nucleus (dLGN) and retina were investigated subsequent to murine controlled unilateral cortical impact. Neuroinflammatory and histopathological data were interpreted in the context of behavioral and visual function data. To investigate the role of complement, cohorts were treated after TBI with the complement inhibitor, CR2-Crry. Results At 3 days after TBI, complement C3 was deposited on retinogeniculate synapses in the dLGN both ipsilateral and contralateral to the lesion, which was reduced in CR2-Crry treated animals. This was associated with microglia morphological changes in both the ipsilateral and contralateral dLGN, with a more amoeboid phenotype in vehicle compared to CR2-Crry treated animals. Microglia in vehicle treated animals also had a greater internalized VGlut2+ synaptic volume after TBI compared to CR2-Crry treated animals. Microglia morphological changes seen acutely persisted for at least 49 days after injury. Complement inhibition also reduced microglial synaptic internalization in the contralateral dLGN and increased the association between VGLUT2 and PSD95 puncta, indicating preservation of intact synapses. Unexpectedly, there were no changes in the thickness of the inner retina, retinal nerve fiber layer or retinal ganglion layer. Pathologies were accompanied by reduced visual acuity at subacute and chronic time points after TBI, with improvement seen in CR2-Crry treated animals. Conclusion TBI induces complement activation within the dLGN and promotes microglial activation and synaptic internalization. Complement inhibition after TBI in a clinically relevant paradigm reduces complement activation, maintains a more surveillance-like microglia phenotype, and preserves synaptic density within the dLGN. Together, the data indicate that complement plays a key role in the development of visual deficits after TBI via complement-dependent microglial phagocytosis of synapses within the dLGN.
Collapse
|
13
|
Varghese N, Morrison B. Partial Depletion of Microglia Attenuates Long-Term Potentiation Deficits following Repeated Blast Traumatic Brain Injury in Organotypic Hippocampal Slice Cultures. J Neurotrauma 2023; 40:547-560. [PMID: 36508265 PMCID: PMC10081725 DOI: 10.1089/neu.2022.0284] [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: 12/14/2022] Open
Abstract
Blast-induced traumatic brain injury (bTBI) has been a health concern in both military and civilian populations due to recent military and geopolitical conflicts. Military service members are frequently exposed to repeated bTBI throughout their training and deployment. Our group has previously reported compounding functional deficits as a result of increased number of blast exposures. In this study, we further characterized the decrease in long-term potentiation (LTP) by varying the blast injury severity and the inter-blast interval between two blast exposures. LTP deficits were attenuated with increasing inter-blast intervals. We also investigated changes in microglial activation; expression of CD68 was increased and expression of CD206 was decreased after multiple blast exposures. Expression of macrophage inflammatory protein (MIP)-1α, interleukin (IL)-1β, monocyte chemoattractant protein (MCP)-1, interferon gamma-inducible protein (IP)-10, and regulated on activation, normal T cell expressed and secreted (RANTES) increased, while expression of IL-10 decreased in the acute period after both single and repeated bTBI. By partially depleting microglia prior to injury, LTP deficits after injury were significantly reduced. Treatment with the novel drug, MW-189, prevented LTP deficits when administered immediately following a repeated bTBI and even when administered only for an acute period (24 h) between two blast injuries. These findings could inform the development of therapeutic strategies to treat the neurological deficits of repeated bTBI suggesting that microglia play a major role in functional neuronal deficits and may be a viable therapeutic target to lessen the neurophysiological deficits after bTBI.
Collapse
Affiliation(s)
- Nevin Varghese
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| |
Collapse
|
14
|
Song JL, Westover MB, Zhang R. A mechanistic model of calcium homeostasis leading to occurrence and propagation of secondary brain injury. J Neurophysiol 2022; 128:1168-1180. [PMID: 36197012 PMCID: PMC9621713 DOI: 10.1152/jn.00045.2022] [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: 02/08/2022] [Revised: 09/13/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022] Open
Abstract
Secondary brain injury (SBI) refers to new or worsening brain insult after primary brain injury (PBI). Neurophysiological experiments show that calcium (Ca2+) is one of the major culprits that contribute to neuronal damage and death following PBI. However, mechanistic details about how alterations of Ca2+ levels contribute to SBI are not well characterized. In this paper, we first build a biophysical model for SBI related to calcium homeostasis (SBI-CH) to study the mechanistic details of PBI-induced disruption of CH, and how these disruptions affect the occurrence of SBI. Then, we construct a coupled SBI-CH model by formulating synaptic interactions to investigate how disruption of CH affects synaptic function and further promotes the propagation of SBI between neurons. Our model shows how the opening of voltage-gated calcium channels (VGCCs), decreasing of plasma membrane calcium pump (PMCA), and reversal of the Na+/Ca2+ exchanger (NCX) during and following PBI, could induce disruption of CH and further promote SBI. We also show that disruption of CH causes synaptic dysfunction, which further induces loss of excitatory-inhibitory balance in the system, and this might promote the propagation of SBI and cause neighboring tissue to be injured. Our findings offer a more comprehensive understanding of the complex interrelationship between CH and SBI.NEW & NOTEWORTHY We build a mechanistic model SBI-CH for calcium homeostasis (CH) to study how alterations of Ca2+ levels following PBI affect the occurrence and propagation of SBI. Specifically, we investigate how the opening of VGCCs, decreasing of PMCA, and reversal of NCX disrupt CH, and further induce the occurrence of SBI. We also present a coupled SBI-CH model to show how disrupted CH causes synaptic dysfunction, and further promotes the propagation of SBI between neurons.
Collapse
Affiliation(s)
- Jiang-Ling Song
- The Medical Big Data Research Center, Northwest University, Xi'an, People's Republic of China
| | - M Brandon Westover
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rui Zhang
- The Medical Big Data Research Center, Northwest University, Xi'an, People's Republic of China
| |
Collapse
|
15
|
Fronczak KM, Roberts A, Svirsky S, Parry M, Holets E, Henchir J, Dixon CE, Carlson SW. Assessment of behavioral, neuroinflammatory, and histological responses in a model of rat repetitive mild fluid percussion injury at 2 weeks post-injury. Front Neurol 2022; 13:945735. [PMID: 36341117 PMCID: PMC9630846 DOI: 10.3389/fneur.2022.945735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Repetitive mild traumatic brain injury (rmTBI) is a prominent public health concern, with linkage to debilitating chronic sequelae. Developing reliable and well-characterized preclinical models of rmTBI is imperative in the investigation of the underlying pathophysiological mechanisms, as models can have varying parameters, affecting the overall pathology of the resulting injury. The lateral fluid percussion injury (FPI) model is a reliable and frequently used method of TBI replication in rodent subjects, though it is currently relatively underutilized in rmTBI research. In this study, we have performed a novel description of a variation of the lateral repetitive mild FPI (rmFPI) model, showing the graded acute behavioral impairment and histopathology occurring in response to one, two or four mild FPI (1.25 atm) or sham surgeries, implemented 24h apart. Beam walking performance revealed significant motor impairment in injured animals, with dysfunction increasing with additional injury. Based upon behavioral responses and histological observations, we further investigated the subacute pathophysiological outcomes of the dual FPI (dFPI). Immunoreactivity assessments showed that dFPI led to regionally-specific reductions in the post-synaptic protein neurogranin and increased subcortical white matter staining of the presynaptic protein synaptophysin at 2 weeks following dFPI. Immunohistochemical assessments of the microglial marker Iba-1 showed a striking increase in in several brain regions, and assessment of the astrocytic marker GFAP showed significantly increased immunoreactivity in the subcortical white matter and thalamus. With this study, we have provided a novel account of the subacute post injury outcomes occurring in response to a rmFPI utilizing these injury and frequency parameters, and thereby also demonstrating the reliability of the lateral FPI model in rmTBI replication.
Collapse
Affiliation(s)
| | - Andrea Roberts
- Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sarah Svirsky
- Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Madison Parry
- Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Erik Holets
- Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jeremy Henchir
- Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - C. Edward Dixon
- Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
- VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - Shaun W. Carlson
- Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
16
|
Langlois LD, Selvaraj P, Simmons SC, Gouty S, Zhang Y, Nugent FS. Repetitive mild traumatic brain injury induces persistent alterations in spontaneous synaptic activity of hippocampal CA1 pyramidal neurons. IBRO Neurosci Rep 2022; 12:157-162. [PMID: 35746968 PMCID: PMC9210462 DOI: 10.1016/j.ibneur.2022.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/20/2021] [Accepted: 02/07/2022] [Indexed: 11/17/2022] Open
Abstract
Mild traumatic brain injury (mTBI) or concussion is the most common form of TBI which frequently results in persistent cognitive impairments and memory deficits in affected individuals [1]. Although most studies have investigated the role of hippocampal synaptic dysfunction in earlier time points following a single injury, the long-lasting effects of mTBI on hippocampal synaptic transmission following multiple brain concussions have not been well-elucidated. Using a repetitive closed head injury (3XCHI) mouse model of mTBI, we examined the alteration of spontaneous synaptic transmission onto hippocampal CA1 pyramidal neurons by recording spontaneous excitatory AMPA receptor (AMPAR)- and inhibitory GABAAR-mediated postsynaptic currents (sEPSCs and sIPSCs, respectively) in adult male mice 2-weeks following the injury. We found that mTBI potentiated postsynaptic excitatory AMPAR synaptic function while depressed postsynaptic inhibitory GABAAR synaptic function in CA1 pyramidal neurons. Additionally, mTBI slowed the decay time of AMPAR currents while shortened the decay time of GABAAR currents suggesting changes in AMPAR and GABAAR subunit composition by mTBI. On the other hand, mTBI reduced the frequency of sEPSCs while enhanced the frequency of sIPSCs resulting in a lower ratio of sEPSC/sIPSC frequency in CA1 pyramidal neurons of mTBI animals compared to sham animals. Altogether, our results suggest that mTBI induces persistent postsynaptic modifications in AMPAR and GABAAR function and their synaptic composition in CA1 neurons while triggering a compensatory shift in excitation/inhibition (E/I) balance of presynaptic drives towards more inhibitory synaptic drive to hippocampal CA1 cells. The persistent mTBI-induced CA1 synaptic dysfunction and E/I imbalance could contribute to deficits in hippocampal plasticity that underlies long-term hippocampal-dependent learning and memory deficits in mTBI patients long after the initial injury.
Collapse
Affiliation(s)
- Ludovic D. Langlois
- Uniformed Services University of the Health Sciences, Edward Hebert School of Medicine, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD 20814, USA
| | - Prabhuanand Selvaraj
- Uniformed Services University of the Health Sciences, Edward Hebert School of Medicine, Department of Anatomy, Physiology and Genetics, Bethesda, MD 20814, USA
| | - Sarah C. Simmons
- Uniformed Services University of the Health Sciences, Edward Hebert School of Medicine, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD 20814, USA
| | - Shawn Gouty
- Uniformed Services University of the Health Sciences, Edward Hebert School of Medicine, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD 20814, USA
| | - Yumin Zhang
- Uniformed Services University of the Health Sciences, Edward Hebert School of Medicine, Department of Anatomy, Physiology and Genetics, Bethesda, MD 20814, USA
- Corresponding authors.
| | - Fereshteh S. Nugent
- Uniformed Services University of the Health Sciences, Edward Hebert School of Medicine, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD 20814, USA
- Corresponding authors.
| |
Collapse
|
17
|
Chen S, Siedhoff HR, Zhang H, Liu P, Balderrama A, Li R, Johnson C, Greenlief CM, Koopmans B, Hoffman T, DePalma RG, Li DP, Cui J, Gu Z. Low-intensity blast induces acute glutamatergic hyperexcitability in mouse hippocampus leading to long-term learning deficits and altered expression of proteins involved in synaptic plasticity and serine protease inhibitors. Neurobiol Dis 2022; 165:105634. [PMID: 35077822 DOI: 10.1016/j.nbd.2022.105634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 11/26/2022] Open
Abstract
Neurocognitive consequences of blast-induced traumatic brain injury (bTBI) pose significant concerns for military service members and veterans with the majority of "invisible injury." However, the underlying mechanism of such mild bTBI by low-intensity blast (LIB) exposure for long-term cognitive and mental deficits remains elusive. Our previous studies have shown that mice exposed to LIB result in nanoscale ultrastructural abnormalities in the absence of gross or apparent cellular damage in the brain. Here we tested the hypothesis that glutamatergic hyperexcitability may contribute to long-term learning deficits. Using brain slice electrophysiological recordings, we found an increase in averaged frequencies with a burst pattern of miniature excitatory postsynaptic currents (mEPSCs) in hippocampal CA3 neurons in LIB-exposed mice at 1- and 7-days post injury, which was blocked by a specific NMDA receptor antagonist AP5. In addition, cognitive function assessed at 3-months post LIB exposure by automated home-cage monitoring showed deficits in dynamic patterns of discrimination learning and cognitive flexibility in LIB-exposed mice. Collected hippocampal tissue was further processed for quantitative global-proteomic analysis. Advanced data-independent acquisition for quantitative tandem mass spectrometry analysis identified altered expression of proteins involved in synaptic plasticity and serine protease inhibitors in LIB-exposed mice. Some were correlated with the ability of discrimination learning and cognitive flexibility. These findings show that acute glutamatergic hyperexcitability in the hippocampus induced by LIB may contribute to long-term cognitive dysfunction and protein alterations. Studies using this military-relevant mouse model of mild bTBI provide valuable insights into developing a potential therapeutic strategy to ameliorate hyperexcitability-modulated LIB injuries.
Collapse
Affiliation(s)
- Shanyan Chen
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Heather R Siedhoff
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Hua Zhang
- Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Pei Liu
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, MO 65211, USA
| | - Ashley Balderrama
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Runting Li
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Catherine Johnson
- Department of Mining and Nuclear Engineering, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - C Michael Greenlief
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, MO 65211, USA
| | | | - Timothy Hoffman
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Ralph G DePalma
- Office of Research and Development, Department of Veterans Affairs, Washington DC 20420, USA; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - De-Pei Li
- Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Jiankun Cui
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| | - Zezong Gu
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| |
Collapse
|
18
|
Chapman DP, Sloley SS, Caccavano AP, Vicini S, Burns MP. High-Frequency Head Impact Disrupts Hippocampal Neural Ensemble Dynamics. Front Cell Neurosci 2022; 15:763423. [PMID: 35115908 PMCID: PMC8806157 DOI: 10.3389/fncel.2021.763423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/21/2021] [Indexed: 12/03/2022] Open
Abstract
We have recently shown that the cognitive impairments in a mouse model of high-frequency head impact (HFHI) are caused by chronic changes to synaptic physiology. To better understand these synaptic changes occurring after repeat head impact, we used Thy1-GcCAMP6f mice to study intracellular and intercellular calcium dynamics and neuronal ensembles in HFHI mice. We performed simultaneous calcium imaging and local field potential (LFP) recordings of the CA1 field during an early-LTP paradigm in acute hippocampal slice preparations 24 h post-impact. As previously reported, HFHI causes a decrease in early-LTP in the absence of any shift in the input-output curve. Calcium analytics revealed that HFHI hippocampal slices have similar numbers of active ROIs, however, the number of calcium transients per ROI was significantly increased in HFHI slices. Ensembles consist of coordinated activity between groups of active ROIs. We exposed the CA1 ensemble to Schaffer-collateral stimulation in an abbreviated LTP paradigm and observed novel coordinated patterns of post stimulus calcium ensemble activity. HFHI ensembles displayed qualitatively similar patterns of post-stimulus ensemble activity to shams but showed significant changes in quantitative ensemble inactivation and reactivation. Previous in vivo and in vitro reports have shown that ensemble activity frequently occurs through a similar set of ROIs firing in a repeating fashion. HFHI slices showed a decrease in such coordinated firing patterns during post stimulus ensemble activity. The present study shows that HFHI alters synaptic activity and disrupts neuronal organization of the ensemble, providing further evidence of physiological synaptic adaptation occurring in the brain after a high frequency of non-pathological head impacts.
Collapse
Affiliation(s)
- Daniel P. Chapman
- Georgetown Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Stephanie S. Sloley
- Georgetown Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Adam P. Caccavano
- Georgetown Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Stefano Vicini
- Georgetown Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Mark P. Burns
- Georgetown Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
- *Correspondence: Mark P. Burns,
| |
Collapse
|
19
|
Zhang R, Wang J, Huang L, Wang TJ, Huang Y, Li Z, He J, Sun C, Wang J, Chen X, Wang J. The pros and cons of motor, memory, and emotion-related behavioral tests in the mouse traumatic brain injury model. Neurol Res 2021; 44:65-89. [PMID: 34308784 DOI: 10.1080/01616412.2021.1956290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a medical emergency with high morbidity and mortality. Motor, memory, and emotion-related deficits are common symptoms following TBI, yet treatment is very limited. To develop new drugs and find new therapeutic avenues, a wide variety of TBI models have been established to mimic the heterogeneity of TBI. In this regard, along with histologic measures, behavioral functional outcomes provide valuable insight into the underlying neuropathology and guide neurorehabilitation efforts for neuropsychiatric impairment after TBI. Development, characterization, and application of behavioral tests that can assess functional neurologic deficits are essential to the development of translational therapies. This comprehensive review aims to summarize 19 common behavioral tests from three aspects (motor, memory, and emotion-related) that are associated with TBI pathology. Discussion covers the apparatus, the test steps, the evaluation indexes, data collection and analysis, animal performance and applications, advantages and disadvantages as well as precautions to eliminate bias wherever possible. We discussed recent studies on TBI-related preconditioning, biomarkers, and optimized behavioral protocols. The neuropsychologic tests employed in clinics were correlated with those used in mouse TBI models. In summary, this review provides a comprehensive, up-to-date reference for TBI researchers to choose the right neurobehavioral protocol according to the research objectives of their translational investigation.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junming Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Yinrou Huang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zefu Li
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxin He
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chen Sun
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuemei Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Intranasal Administration of Oxytocin Attenuates Social Recognition Deficits and Increases Prefrontal Cortex Inhibitory Postsynaptic Currents following Traumatic Brain Injury. eNeuro 2021; 8:ENEURO.0061-21.2021. [PMID: 34035071 PMCID: PMC8205495 DOI: 10.1523/eneuro.0061-21.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric traumatic brain injury (TBI) results in heightened risk for social deficits that can emerge during adolescence and adulthood. A moderate TBI in male and female rats on postnatal day 11 (equivalent to children below the age of 4) resulted in impairments in social novelty recognition, defined as the preference for interacting with a novel rat compared with a familiar rat, but not sociability, defined as the preference for interacting with a rat compared with an object in the three-chamber test when tested at four weeks (adolescence) and eight weeks (adulthood) postinjury. The deficits in social recognition were not accompanied by deficits in novel object recognition memory and were associated with a decrease in the frequency of spontaneous inhibitory postsynaptic currents (IPSCs) recorded from pyramidal neurons within Layer II/III of the medial prefrontal cortex (mPFC). Whereas TBI did not affect the expression of oxytocin (OXT) or the OXT receptor (OXTR) mRNAs in the hypothalamus and mPFC, respectively, intranasal administration of OXT before behavioral testing was found to reduce impairments in social novelty recognition and increase IPSC frequency in the mPFC in brain-injured animals. These results suggest that TBI-induced deficits in social behavior may be linked to increased excitability of neurons in the mPFC and suggests that the regulation of GABAergic neurotransmission in this region as a potential mechanism underlying these deficits.
Collapse
|
21
|
Xu X, Cowan M, Beraldo F, Schranz A, McCunn P, Geremia N, Brown Z, Patel M, Nygard KL, Khazaee R, Lu L, Liu X, Strong MJ, Dekaban GA, Menon R, Bartha R, Daley M, Mao H, Prado V, Prado MAM, Saksida L, Bussey T, Brown A. Repetitive mild traumatic brain injury in mice triggers a slowly developing cascade of long-term and persistent behavioral deficits and pathological changes. Acta Neuropathol Commun 2021; 9:60. [PMID: 33823944 PMCID: PMC8025516 DOI: 10.1186/s40478-021-01161-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
We have previously reported long-term changes in the brains of non-concussed varsity rugby players using magnetic resonance spectroscopy (MRS), diffusion tensor imaging (DTI) and functional magnetic imaging (fMRI). Others have reported cognitive deficits in contact sport athletes that have not met the diagnostic criteria for concussion. These results suggest that repetitive mild traumatic brain injuries (rmTBIs) that are not severe enough to meet the diagnostic threshold for concussion, produce long-term consequences. We sought to characterize the neuroimaging, cognitive, pathological and metabolomic changes in a mouse model of rmTBI. Using a closed-skull model of mTBI that when scaled to human leads to rotational and linear accelerations far below what has been reported for sports concussion athletes, we found that 5 daily mTBIs triggered two temporally distinct types of pathological changes. First, during the first days and weeks after injury, the rmTBI produced diffuse axonal injury, a transient inflammatory response and changes in diffusion tensor imaging (DTI) that resolved with time. Second, the rmTBI led to pathological changes that were evident months after the injury including: changes in magnetic resonance spectroscopy (MRS), altered levels of synaptic proteins, behavioural deficits in attention and spatial memory, accumulations of pathologically phosphorylated tau, altered blood metabolomic profiles and white matter ultrastructural abnormalities. These results indicate that exceedingly mild rmTBI, in mice, triggers processes with pathological consequences observable months after the initial injury.
Collapse
|
22
|
Hamilton KA, Santhakumar V. Current ex Vivo and in Vitro Approaches to Uncovering Mechanisms of Neurological Dysfunction after Traumatic Brain Injury. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 14:18-24. [PMID: 32548365 PMCID: PMC7297186 DOI: 10.1016/j.cobme.2020.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Traumatic brain injury often leads to progressive alterations at the molecular to circuit levels resulting in epilepsy and memory impairments. Ex vivo and in vitro models have provided a powerful platform for investigating the multimodal alteration after trauma. Recent ex vivo analyses using voltage sensitive dye imaging, optogenetics, and glutamate uncaging have revealed circuit abnormalities following in vivo brain injury. In vitro injury models have enabled examination of early and progressive changes in activity while development of three-dimensional organoids derived from human induced pluripotent stem cells have opened novel avenues for injury research. Here, we highlight recent advances in ex vivo and in vitro systems, focusing on their potential for advancing mechanistic understandings, possible limitations, and implications for therapeutics.
Collapse
Affiliation(s)
- Kelly Andrew Hamilton
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
| | - Vijayalakshmi Santhakumar
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
23
|
Lengel D, Huh JW, Barson JR, Raghupathi R. Progesterone treatment following traumatic brain injury in the 11-day-old rat attenuates cognitive deficits and neuronal hyperexcitability in adolescence. Exp Neurol 2020; 330:113329. [PMID: 32335121 DOI: 10.1016/j.expneurol.2020.113329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/13/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) in children younger than 4 years old results in cognitive and psychosocial deficits in adolescence and adulthood. At 4 weeks following closed head injury on postnatal day 11, male and female rats exhibited impairment in novel object recognition memory (NOR) along with an increase in open arm time in the elevated plus maze (EPM), suggestive of risk-taking behaviors. This was accompanied by an increase in intrinsic excitability and frequency of spontaneous excitatory post-synaptic currents (EPSCs), and a decrease in the frequency of spontaneous inhibitory post-synaptic currents in layer 2/3 neurons within the medial prefrontal cortex (PFC), a region that is implicated in both object recognition and risk-taking behaviors. Treatment with progesterone for the first week after brain injury improved NOR memory at the 4-week time point in both sham and brain-injured rats and additionally attenuated the injury-induced increase in the excitability of neurons and the frequency of spontaneous EPSCs. The effect of progesterone on cellular excitability changes after injury may be related to its ability to decrease the mRNA expression of the β3 subunit of the voltage-gated sodium channel and increase the expression of the neuronal excitatory amino acid transporter 3 in the medial PFC in sham- and brain-injured animals and also increase glutamic acid decarboxylase mRNA expression in sham- but not brain-injured animals. Progesterone treatment did not affect injury-induced changes in the EPM test. These results demonstrate that administration of progesterone immediately after TBI in 11-day-old rats reduces cognitive deficits in adolescence, which may be mediated by progesterone-mediated regulation of excitatory signaling mechanisms within the medial PFC.
Collapse
Affiliation(s)
- Dana Lengel
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Jessica R Barson
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Ramesh Raghupathi
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States of America.
| |
Collapse
|