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] [MESH Headings] [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
|
Spadacenta S, Dicke PW, Thier P. Minimally invasive electrocorticography (ECoG) recording in common marmosets. J Neurosci Methods 2025; 417:110409. [PMID: 40010648 DOI: 10.1016/j.jneumeth.2025.110409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/01/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Electrocorticography (ECoG) provides a valuable compromise between spatial and temporal resolution for recording brain activity with excellent signal quality, crucial for presurgical epilepsy mapping and advancing neuroscience, including brain-machine interface development. ECoG is particularly effective in the common marmoset (Callithrix jacchus), whose lissencephalic (unfolded) brain surface provides broad cortical access. One of the key advantages of ECoG recordings is the ability to study interactions between distant brain regions. Traditional methods rely on large electrode arrays, necessitating extensive trepanations and a trade-off between size and electrode spacing. NEW METHOD This study introduces a refined ECoG technique for examining interactions among multiple cortical areas in marmosets, combining circumscribed trepanations with high-density electrode arrays at specific sites of interest. COMPARISON WITH EXISTING METHODS Standard ECoG techniques typically require large electrode arrays and extensive trepanation, which heighten surgical risks and the likelihood of infection, while potentially compromising spatial resolution. In contrast, our method facilitates detailed and stable recordings across multiple cortical areas with minimized invasiveness and reduced complication risks, all while preserving high spatial resolution. RESULTS Two adult marmosets underwent ECoG implantation in frontal, temporal, and parietal regions. Postoperative monitoring confirmed rapid recovery, long-term health, and stable, high-quality neural recordings during various behavioral tasks. CONCLUSIONS This refined ECoG method enhances the study of cortical interactions in marmosets while minimizing surgical invasiveness and complication risks. It offers potential for broader application in other species and opens new avenues for long-term data collection, ultimately advancing both neuroscience and brain-machine interface research.
Collapse
Affiliation(s)
- Silvia Spadacenta
- Cognitive Neurology Laboratory, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Str. 27, Tübingen 72076, Germany.
| | - Peter W Dicke
- Cognitive Neurology Laboratory, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Str. 27, Tübingen 72076, Germany
| | - Peter Thier
- Cognitive Neurology Laboratory, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Str. 27, Tübingen 72076, Germany.
| |
Collapse
|
3
|
Comparan HDM, Khaliq A, Frota LM, Pomar-Forero D, Ahmad B, Marnet E, Teixeira FJP, Thomas A, Patel P, Brunkal H, Singireddy S, Lucke-Wold B, Maciel CB, Busl KM. Cyclooxygenase 2 Inhibitors for Headache After Elective Cranial Neurosurgery: Results from a Systematic Review of Efficacy of Cyclooxygenase 2 Inhibitors for Headache After Acute Brain Injury. Neurocrit Care 2025; 42:680-689. [PMID: 39266865 DOI: 10.1007/s12028-024-02114-y] [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: 04/07/2024] [Accepted: 08/22/2024] [Indexed: 09/14/2024]
Abstract
Headache management after acute brain injury (ABI) is challenging. Although opioids are commonly used, selective cyclooxygenase 2 inhibitors (COXIBs) may be promising alternatives. However, concerns about cardiovascular effects and bleeding risk have limited their use. We aimed at summarizing available data on efficacy of COXIBs for headache management following ABI. A systematic review was conducted through MEDLINE and Embase for articles published through September 2023 (PROSPERO identifier: CRD42022320453). No language filters were applied to the initial searches. Interventional or observational studies and systematic reviews assessing efficacy of COXIBs for headache in adults with ABI were eligible. Article selection was performed by two independent reviewers using DistillerSR. Descriptive statistics were used for data analysis, and meta-analysis was unfeasible because of study heterogeneity. Of 3190 articles identified, 6 studies met inclusion criteria: 4 randomized controlled trials and 2 retrospective cohort studies, all conducted in elective cranial neurosurgical patients (total N = 738) between 2006 and 2022. Five studies used COXIBs in the intervention group only. Of the six studies, four found a reduction in overall pain scores in the intervention group, whereas one showed improvement only at 6 h postoperatively, and one did not find significant differences. Pain scores decreased between 4 and 15%, the largest shift being from moderate to mild severity. Three studies found an overall opioid use reduction throughout hospitalization in the intervention group, whereas one reported a reduction at 12 h postoperatively only. Opioid consumption decreased between 9 and 90%. Two studies found a decrease in hospital length of stay by ~ 1 day in the intervention group. The one study reporting postoperative hemorrhage found a statistically nonsignificant 3% reduction in the intervention group. COXIBs may serve as opioid-sparing adjunctive analgesics for headache control after elective cranial surgery. Limited or no literature exists for other forms of ABI, and additional safety data remain to be elucidated.
Collapse
Affiliation(s)
- Hector David Meza Comparan
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Anum Khaliq
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Luciola Martins Frota
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Daniela Pomar-Forero
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Bakhtawar Ahmad
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Erica Marnet
- Department of Medicine, Bridgeport Hospital, Bridgeport, CT, 06610, USA
| | - Fernanda J P Teixeira
- Department of Neurology, University of Miami and Jackson Memorial Hospital, Miami, FL, 33130, USA
| | - Anita Thomas
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Priyank Patel
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Haley Brunkal
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Saanvi Singireddy
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Carolina B Maciel
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Neurology, University of Utah, Salt Lake City, UT, 84132, USA
| | - Katharina M Busl
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
4
|
Gureev AP, Nesterova VV, Babenkova PI, Ivanov ME, Plotnikov EY, Silachev DN. L-Carnitine and Mildronate Demonstrate Divergent Protective Effects on Mitochondrial DNA Quality Control and Inflammation Following Traumatic Brain Injury. Int J Mol Sci 2025; 26:2902. [PMID: 40243464 PMCID: PMC11988827 DOI: 10.3390/ijms26072902] [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: 02/17/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Traumatic brain injuries (TBIs) are a serious problem affecting individuals of all ages. Mitochondrial dysfunctions represent a significant form of secondary injury and may serve as a promising target for therapeutic intervention. Our research demonstrated that craniotomy, which precedes the experimental induction of trauma in mice, can cause considerable damage to mitochondrial DNA (mtDNA), disrupt the regulatory expression of angiogenesis, and increase inflammation. However, the reduction in the mtDNA copy number and glial activation occur only after a direct impact to the brain. We explored two potential therapeutic agents: the dietary supplement L-carnitine-a potential reserve source of ATP for the brain-and the cardiac drug mildronate, which inhibits L-carnitine but activates alternative compensatory pathways for the brain to adapt to metabolic disturbances. We found that L-carnitine injections could protect against mtDNA depletion by promoting mitochondrial biogenesis. However, they also appeared to aggravate inflammatory responses, likely due to changes in the composition of the gut microbiome. On the other hand, mildronate enhanced the expression of genes related to angiogenesis while also reducing local and systemic inflammation. Therefore, both compounds, despite their opposing metabolic effects, have the potential to be used in the treatment of secondary injuries caused by TBI.
Collapse
Affiliation(s)
- Artem P. Gureev
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (A.P.G.); (V.V.N.); (P.I.B.)
| | - Veronika V. Nesterova
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (A.P.G.); (V.V.N.); (P.I.B.)
| | - Polina I. Babenkova
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (A.P.G.); (V.V.N.); (P.I.B.)
| | - Mikhail E. Ivanov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (M.E.I.); (E.Y.P.)
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (M.E.I.); (E.Y.P.)
| | - Denis N. Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (M.E.I.); (E.Y.P.)
| |
Collapse
|
5
|
Redell JB, Maynard ME, Hylin MJ, Hood KN, Sedlock A, Maric D, Zhao J, Moore AN, Roysam B, Pati S, Dash PK. A Combination of Low Doses of Lithium and Valproate Improves Cognitive Outcomes after Mild Traumatic Brain Injury. J Neurotrauma 2025; 42:437-453. [PMID: 39463282 PMCID: PMC11971536 DOI: 10.1089/neu.2024.0311] [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: 10/29/2024] Open
Abstract
The prevalence of mild traumatic brain injury (mTBI) is high compared with moderate and severe TBI, comprising almost 80% of all brain injuries. mTBI activates a complex cascade of biochemical, molecular, structural, and pathological changes that can result in neurological and cognitive impairments. These impairments can manifest even in the absence of overt brain damage. Given the complexity of changes triggered by mTBI, a combination of drugs that target multiple TBI-activated cascades may be required to improve mTBI outcomes. It has been previously demonstrated that cotreatment with the U.S. Food and Drug Administration (FDA)-approved drugs lithium plus valproate (Li + VPA) for 3 weeks after a moderate-to-severe controlled cortical impact injury reduced cortical tissue loss and improved motor function. Since both lithium and valproate can exhibit toxicity at high doses, it would be beneficial to determine if this combination treatment is effective when administered at low doses and for a shorter duration, and if it can improve cognitive function, after a mild diffuse TBI. In the present study, we tested if the combination of low doses of lithium (1 mEq/kg or 0.5 mEq/kg) plus valproate (20 mg/kg) administered for 3 days after a mild fluid percussion injury can improve hippocampal-dependent learning and memory. Our data show that the combination of low-dose Li + VPA improved spatial learning and memory, effects not seen when either drug was administered alone. In addition, postinjury Li + VPA treatment improved recognition memory and sociability and reduced fear generalization. Postinjury Li + VPA also reduced the number of anti-ionized calcium binding adaptor molecule 1 (Iba1)-positive microglia counted using a convolutional neural network, indicating a reduction in neuroinflammation. These findings indicate that low-dose Li + VPA administered acutely after mTBI may have translational utility to reduce pathology and improve cognitive function.
Collapse
Affiliation(s)
- John B. Redell
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Mark E. Maynard
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, USA
| | - Michael J. Hylin
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Kimberly N. Hood
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Andrea Sedlock
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Anthony N. Moore
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Badrinath Roysam
- Department of Electrical and Computer Engineering, University of Houston, Houston, Texas, USA
| | - Shibani Pati
- Departments of Pathology and Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Pramod K. Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas, USA
| |
Collapse
|
6
|
Astakhova O, Ivanova A, Komoltsev I, Gulyaeva N, Enikolopov G, Lazutkin A. Traumatic Brain Injury Promotes Neurogenesis and Oligodendrogenesis in Subcortical Brain Regions of Mice. Cells 2025; 14:92. [PMID: 39851520 PMCID: PMC11764027 DOI: 10.3390/cells14020092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Traumatic brain injury (TBI) is one of the major causes of severe neurological disorders and long-term dysfunction in the nervous system. Besides inducing neurodegeneration, TBI alters stem cell activity and neurogenesis within primary neurogenic niches. However, the fate of dividing cells in other brain regions remains unclear despite offering potential targets for therapeutic intervention. Here, we investigated cell division and differentiation in non-neurogenic brain regions during the acute and delayed phases of TBI-induced neurodegeneration. We subjected mice to lateral fluid percussion injury (LFPI) to model TBI and analyzed them 1 or 7 weeks later. To assess cellular proliferation and differentiation, we administered 5-ethinyl-2'-deoxyuridine (EdU) and determined the number and identity of dividing cells 2 h later using markers of neuronal precursors and astro-, micro-, and oligodendroglia. Our results demonstrated a significant proliferative response in several brain regions at one week post-injury that notably diminished by seven weeks, except in the optic tract. In addition to active astro- and microgliosis, we detected oligodendrogenesis in the striatum and optic tract. Furthermore, we observed trauma-induced neurogenesis in the striatum. These findings suggest that subcortical structures, particularly the striatum and optic tract, may possess a potential for self-repair through neuronal regeneration and axon remyelination.
Collapse
Affiliation(s)
- Olga Astakhova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (O.A.)
- Department of Human and Animal Physiology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - Anna Ivanova
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow 119991, Russia;
| | - Ilia Komoltsev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (O.A.)
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow 115419, Russia
| | - Natalia Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (O.A.)
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow 115419, Russia
| | - Grigori Enikolopov
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Alexander Lazutkin
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (O.A.)
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
7
|
Markicevic M, Mandino F, Toyonaga T, Cai Z, Fesharaki-Zadeh A, Shen X, Strittmatter SM, Lake EM. Repetitive Mild Closed-Head Injury Induced Synapse Loss and Increased Local BOLD-fMRI Signal Homogeneity. J Neurotrauma 2024; 41:2528-2544. [PMID: 39096127 PMCID: PMC11698675 DOI: 10.1089/neu.2024.0095] [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: 08/04/2024] Open
Abstract
Repeated mild head injuries due to sports, or domestic violence and military service are increasingly linked to debilitating symptoms in the long term. Although symptoms may take decades to manifest, potentially treatable neurobiological alterations must begin shortly after injury. Better means to diagnose and treat traumatic brain injuries requires an improved understanding of the mechanisms underlying progression and means through which they can be measured. Here, we employ a repetitive mild traumatic brain injury (rmTBI) and chronic variable stress mouse model to investigate emergent structural and functional brain abnormalities. Brain imaging is achieved with [18F]SynVesT-1 positron emission tomography, with the synaptic vesicle glycoprotein 2A ligand marking synapse density and BOLD (blood-oxygen-level-dependent) functional magnetic resonance imaging (fMRI). Animals were scanned six weeks after concluding rmTBI/Stress procedures. Injured mice showed widespread decreases in synaptic density coupled with an increase in local BOLD-fMRI synchrony detected as regional homogeneity. Injury-affected regions with higher synapse density showed a greater increase in fMRI regional homogeneity. Taken together, these observations may reflect compensatory mechanisms following injury. Multimodal studies are needed to provide deeper insights into these observations.
Collapse
Affiliation(s)
- Marija Markicevic
- Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Francesca Mandino
- Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Arman Fesharaki-Zadeh
- Department of Neurology, School of Medicine, Yale University, New Haven, Connecticut, USA
- Department of Psychiatry, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Xilin Shen
- Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Stephen M. Strittmatter
- Department of Neurology, School of Medicine, Yale University, New Haven, Connecticut, USA
- Department of Neuroscience, School of Medicine, Yale University, New Haven, Connecticut, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, Connecticut, USA
| | - Evelyn M.R. Lake
- Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, Connecticut, USA
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
8
|
Schmidt R, Welzel B, Merten A, Naundorf H, Löscher W. Temporal development of seizure threshold and spontaneous seizures after neonatal asphyxia and the effect of prophylactic treatment with midazolam in rats. Exp Neurol 2024; 383:115042. [PMID: 39505250 DOI: 10.1016/j.expneurol.2024.115042] [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: 08/19/2024] [Revised: 10/15/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Birth asphyxia (BA) and subsequent hypoxic-ischemic encephalopathy (HIE) is one of the most serious birth complications affecting full-term infants and can result in severe disabilities including mental retardation, cerebral palsy, and epilepsy. Animal models of BA and HIE are important to characterize the functional and behavioral correlates of injury, explore cellular and molecular mechanisms, and assess the potential of novel therapeutic strategies. Here we used a non-invasive, physiologically validated rat model of BA and acute neonatal seizures that mimics many features of BA and HIE in human infants to study (i) the temporal development of epilepsy with spontaneous recurrent seizures (SRS) in the weeks and months after the initial brain injury, (ii) alterations in seizure threshold and hippocampal EEG that may precede the onset of SRS, and (iii) the effect of prophylactic treatment with midazolam. For this purpose, a total of 89 rat pups underwent asphyxia or sham asphyxia at postnatal day 11 and were examined over 8-10.5 months. In vehicle-treated animals, the incidence of electroclinical SRS progressively increased from 0 % at 2.5 months to 50 % at 6.5 months, 75 % at 8.5 months, and > 80 % at 10.5 months after asphyxia. Unexpectedly, post-asphyxial rats did not differ from sham-exposed rats in seizure threshold or interictal epileptiform discharges in the EEG. Treatment with midazolam (1 mg/kg i.p.) after asphyxia, which suppressed acute symptomatic neonatal seizures in about 60 % of the rat pups, significantly reduced the incidence of SRS regardless of its effect on neonatal seizures. This antiepileptogenic effect of midazolam adds to the recently reported prophylactic effects of this drug on BA-induced neuroinflammation, brain damage, behavioral alterations, and cognitive impairment in the rat asphyxia model of HIE.
Collapse
Affiliation(s)
- Ricardo Schmidt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience Hannover, Germany; Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany
| | - Björn Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Annika Merten
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Hannah Naundorf
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience Hannover, Germany; Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
9
|
Santana-Gomez C, Smith G, Mousavi A, Shamas M, Harris NG, Staba R. The Surgical Method of Craniectomy Differentially Affects Acute Seizures, Brain Deformation, and Behavior in a Traumatic Brain Injury Animal Model. Neurotrauma Rep 2024; 5:969-981. [PMID: 39440152 PMCID: PMC11491586 DOI: 10.1089/neur.2024.0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of morbidity and mortality worldwide. Multiple injury models have been developed to study this neurological disorder. One such model is the lateral fluid percussion injury (LFPI) rodent model. The LFPI model can be generated with different surgical procedures that could affect the injury and be reflected in neurobehavioral dysfunction and acute electroencephalograph (EEG) changes. A craniectomy was performed either with a trephine hand drill or with a trephine electric drill that was centered over the left hemisphere of adult, male Sprague Dawley rats. Sham craniectomy groups were assessed by hand-drilled (ShamHMRI) and electric-drilled (ShamEMRI) to evaluate by magnetic resonance imaging (MRI). Then, TBI was induced in separate groups, (TBIH) and (TBIE), using a fluid-percussion device. Sham-injured rats (ShamH/ShamE) underwent the same surgical procedures as the TBI rats. During the same surgery session, rats were implanted with screw and microwire electrodes positioned in the neocortex and hippocampus and the EEG activity was recorded 24 h for the first 7 days after TBI for assessing the acute EEG seizure and gamma event coupling. The electric drilling craniectomy induced greater tissue damage and sensorimotor deficits compared with the hand drill. Analysis of the EEG revealed acute seizures in at least one animal from each group after the procedure. Both TBI and Sham rats from the electric drill groups had a significant greater total number of seizures than the animals that were craniectomized manually (p < 0.05). Similarly, EEG functional connectivity was lower in ShamE compared with ShamH rats. These results suggest that electrical versus hand-drilling craniectomies produce cortical injury in addition to the LFPI which increases the likelihood for acute post-traumatic seizures. Differences in the surgical approach could be one reason for the variability in the injury that makes it difficult to replicate results between preclinical TBI studies.
Collapse
Affiliation(s)
- Cesar Santana-Gomez
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Gregory Smith
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ava Mousavi
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mohamad Shamas
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Neil G. Harris
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Richard Staba
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
10
|
Jha RM, Rajasundaram D, Sneiderman C, Schlegel BT, O'Brien C, Xiong Z, Janesko-Feldman K, Trivedi R, Vagni V, Zusman BE, Catapano JS, Eberle A, Desai SM, Jadhav AP, Mihaljevic S, Miller M, Raikwar S, Rani A, Rulney J, Shahjouie S, Raphael I, Kumar A, Phuah CL, Winkler EA, Simon DW, Kochanek PM, Kohanbash G. A single-cell atlas deconstructs heterogeneity across multiple models in murine traumatic brain injury and identifies novel cell-specific targets. Neuron 2024; 112:3069-3088.e4. [PMID: 39019041 DOI: 10.1016/j.neuron.2024.06.021] [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: 09/28/2023] [Revised: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 07/19/2024]
Abstract
Traumatic brain injury (TBI) heterogeneity remains a critical barrier to translating therapies. Identifying final common pathways/molecular signatures that integrate this heterogeneity informs biomarker and therapeutic-target development. We present the first large-scale murine single-cell atlas of the transcriptomic response to TBI (334,376 cells) across clinically relevant models, sex, brain region, and time as a foundational step in molecularly deconstructing TBI heterogeneity. Results were unique to cell populations, injury models, sex, brain regions, and time, highlighting the importance of cell-level resolution. We identify cell-specific targets and previously unrecognized roles for microglial and ependymal subtypes. Ependymal-4 was a hub of neuroinflammatory signaling. A distinct microglial lineage shared features with disease-associated microglia at 24 h, with persistent gene-expression changes in microglia-4 even 6 months after contusional TBI, contrasting all other cell types that mostly returned to naive levels. Regional and sexual dimorphism were noted. CEREBRI, our searchable atlas (https://shiny.crc.pitt.edu/cerebri/), identifies previously unrecognized cell subtypes/molecular targets and is a leverageable platform for future efforts in TBI and other diseases with overlapping pathophysiology.
Collapse
Affiliation(s)
- Ruchira M Jha
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Safar Center for Resuscitation-Research, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Division of Health Informatics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Chaim Sneiderman
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Brent T Schlegel
- Department of Pediatrics, Division of Health Informatics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Casey O'Brien
- Department of Pediatrics, Division of Health Informatics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Zujian Xiong
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation-Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Ria Trivedi
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Vincent Vagni
- Safar Center for Resuscitation-Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Benjamin E Zusman
- Department of Neurology, Massachusetts General Hospital, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Joshua S Catapano
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Adam Eberle
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | | | - Ashutosh P Jadhav
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Sandra Mihaljevic
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Margaux Miller
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Sudhanshu Raikwar
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Anupama Rani
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jarrod Rulney
- University of Arizona School of Medicine, Tucson, AZ 85724, USA
| | - Shima Shahjouie
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Department of Neurology, Pennsylvania State University, Hershey, PA 17033, USA
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Aditya Kumar
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Chia-Ling Phuah
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Ethan A Winkler
- Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dennis W Simon
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation-Research, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
11
|
Vita SM, Cruise SC, Gilpin NW, Molina PE. HISTOLOGICAL COMPARISON OF REPEATED MILD WEIGHT DROP AND LATERAL FLUID PERCUSSION INJURY MODELS OF TRAUMATIC BRAIN INJURY IN FEMALE AND MALE RATS. Shock 2024; 62:398-409. [PMID: 38813916 DOI: 10.1097/shk.0000000000002395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
ABSTRACT In preclinical traumatic brain injury (TBI) research, the animal model should be selected based on the research question and outcome measures of interest. Direct side-by-side comparisons of different injury models are essential for informing such decisions. Here, we used immunohistochemistry to compare the outcomes from two common models of TBI, lateral fluid percussion (LFP) and repeated mild weight drop (rmWD) in adult female and male Wistar rats. Specifically, we measured the effects of LFP and rmWD on markers of cerebrovascular and tight junction disruption, neuroinflammation, mature neurons, and perineuronal nets in the cortical site of injury, cortex adjacent to injury, dentate gyrus, and the CA 2/3 area of the hippocampus. Animals were randomized into the LFP or rmWD group. On day 1, the LFP group received a craniotomy, and on day 4, injury (or sham procedure; randomly assigned). The rmWD animals underwent either injury or isoflurane only (randomly assigned) on each of those 4 days. Seven days after injury, brains were harvested for analysis. Overall, our observations revealed that the most significant disruptions were evident in response to LFP, followed by craniotomy only, whereas rmWD animals showed the least residual changes compared with isoflurane-only controls, supporting consideration of rmWD as a mild injury. LFP led to longer-lasting disruptions, perhaps more representative of moderate TBI. We also report that craniotomy and LFP produced greater disruptions in females relative to males. These findings will assist the field in the selection of animal models based on target severity of postinjury outcomes and support the inclusion of both sexes and appropriate control groups.
Collapse
Affiliation(s)
| | - Shealan C Cruise
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | | | | |
Collapse
|
12
|
Candamo-Lourido M, Dopico-López A, López-Arias E, López-Amoedo S, Correa-Paz C, Chantada-Vázquez MP, Bugallo-Casal A, del Pozo-Filíu L, Pérez-Gayol L, Palomar-Alonso N, Bravo SB, Campos F, Pérez-Mato M. Comparative Brain Proteomic Analysis between Sham and Cerebral Ischemia Experimental Groups. Int J Mol Sci 2024; 25:7538. [PMID: 39062782 PMCID: PMC11277324 DOI: 10.3390/ijms25147538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Sham control groups are essential in experimental animal studies to reduce the influence of surgical intervention. The intraluminal filament procedure is one of the most common models of middle cerebral artery occlusion (MCAO) used in the study of brain ischemia. However, a sham group is usually not included in the experimental design of these studies. In this study, we aimed to evaluate the relevance of the sham group by analyzing and comparing the brain protein profiles of the sham and MCAO groups. In the sham group, 98 dysregulated proteins were detected, compared to 171 in the ischemic group. Moreover, a comparative study of protein profiles revealed the existence of a pool of 57 proteins that appeared to be dysregulated in both sham and ischemic animals. These results indicated that the surgical procedure required for the intraluminal occlusion of the middle cerebral artery (MCA) induces changes in brain protein expression that are not associated with ischemic lesions. This study highlights the importance of including sham control groups in the experimental design, to ensure that surgical intervention does not affect the therapeutic target under study.
Collapse
Affiliation(s)
- María Candamo-Lourido
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - Antonio Dopico-López
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - Esteban López-Arias
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - Sonia López-Amoedo
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - Clara Correa-Paz
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - María Pilar Chantada-Vázquez
- Proteomic Unit, Research Institute of Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain; (M.P.C.-V.); (S.B.B.)
| | - Ana Bugallo-Casal
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - Lucía del Pozo-Filíu
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - Lara Pérez-Gayol
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - Nuria Palomar-Alonso
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - Susana B. Bravo
- Proteomic Unit, Research Institute of Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain; (M.P.C.-V.); (S.B.B.)
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| | - María Pérez-Mato
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.-L.); (A.D.-L.); (E.L.-A.); (S.L.-A.); (C.C.-P.); (A.B.-C.); (L.P.-G.); (N.P.-A.)
| |
Collapse
|
13
|
Santana-Gomez C, Smith G, Mousavi A, Shamas M, Harris NG, Staba R. The Surgical Method of Craniectomy Differentially Affects Acute Seizures, Brain Deformation and Behavior in a TBI Animal Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601257. [PMID: 39005303 PMCID: PMC11244902 DOI: 10.1101/2024.06.28.601257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Traumatic brain injury (TBI) is the leading cause of morbidity and mortality worldwide. Multiple injury models have been developed to study this neurological disorder. One such model is the lateral fluid-percussion injury (LFPI) rodent model. The LFPI model can be generated with different surgical procedures that could affect the injury and be reflected in neurobehavioral dysfunction and acute EEG changes. A craniectomy was performed either with a trephine hand drill or with a trephine electric drill that was centered over the left hemisphere of adult, male Sprague Dawley rats. Sham craniectomy groups were assessed by hand-drilled (ShamHMRI) and electric-drilled (ShamEMRI) to evaluate by MRI. Then, TBI was induced in separate groups (TBIH) and (TBIE) using a fluid-percussion device. Sham-injured rats (ShamH/ShamE) underwent the same surgical procedures as the TBI rats. During the same surgery session, rats were implanted with screw and microwire electrodes positioned in the neocortex and hippocampus and the EEG activity was recorded 24 hours for the first 7 days after TBI for assessing the acute EEG seizure and Gamma Event Coupling (GEC). The electric drilling craniectomy induced greater tissue damage and sensorimotor deficits compared to the hand drill. Analysis of the EEG revealed acute seizures in at least one animal from each group after the procedure. Both TBI and Sham rats from the electric drill groups had a significant greater total number of seizures than the animals that were craniectomized manually (p<0.05). Similarly, EEG functional connectivity was lower in ShamE compared to ShamH rats. These results suggest that electrical versus hand drilling craniectomies produce cortical injury in addition to the LFPI which increases the likelihood for acute post-traumatic seizures. Differences in the surgical approach could be one reason for the variability in the injury that makes it difficult to replicate results between preclinical TBI studies.
Collapse
|
14
|
Koloski MF, O'Hearn CM, Frankot M, Giesler LP, Ramanathan DS, Vonder Haar C. Behavioral Interventions Can Improve Brain Injury-Induced Deficits in Behavioral Flexibility and Impulsivity Linked to Impaired Reward-Feedback Beta Oscillations. J Neurotrauma 2024; 41:e1721-e1737. [PMID: 38450560 PMCID: PMC11339556 DOI: 10.1089/neu.2023.0448] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024] Open
Abstract
Traumatic brain injury (TBI) affects a large population, resulting in severe cognitive impairments. Although cognitive rehabilitation is an accepted treatment for some deficits, studies in patients are limited in ability to probe physiological and behavioral mechanisms. Therefore, animal models are needed to optimize strategies. Frontal TBI in a rat model results in robust and replicable cognitive deficits, making this an ideal candidate for investigating various behavioral interventions. In this study, we report three distinct frontal TBI experiments assessing behavior well into the chronic post-injury period using male Long-Evans rats. First, we evaluated the impact of frontal injury on local field potentials recorded simultaneously from 12 brain regions during a probabilistic reversal learning (PbR) task. Next, a set of rats were tested on a similar PbR task or an impulsivity task (differential reinforcement of low-rate behavior [DRL]) and half received salient cues associated with reinforcement contingencies to encourage engagement in the target behavior. After intervention on the PbR task, brains were stained for markers of activity. On the DRL task, cue relevance was decoupled from outcomes to determine if beneficial effects persisted on impulsive behavior. TBI decreased the ability to detect reinforced outcomes; this was evident in task performance and reward-feedback signals occurring at beta frequencies in lateral orbitofrontal cortex (OFC) and associated frontostriatal regions. The behavioral intervention improved flexibility and increased OFC activity. Intervention also reduced impulsivity, even after cues were decoupled, which was partially mediated by improvements in timing behavior. The current study established a platform to begin investigating cognitive rehabilitation in rats and identified a strong role for dysfunctional OFC signaling in probabilistic learning after frontal TBI.
Collapse
Affiliation(s)
- Miranda F. Koloski
- Mental Health, VA San Diego Medical Center, San Diego, California, USA
- Center of Excellence for Stress and Mental Health, San Diego, California, USA
- Department of Psychiatry, University of California-San Diego, San Diego, California, USA
| | | | - Michelle Frankot
- Department of Psychology, West Virginia University, Morgantown, West Virginia, USA
- Injury and Recovery Laboratory, Department of Neuroscience, Ohio State University, Columbus, Ohio, USA
| | - Lauren P. Giesler
- Department of Psychology, West Virginia University, Morgantown, West Virginia, USA
| | - Dhakshin S. Ramanathan
- Mental Health, VA San Diego Medical Center, San Diego, California, USA
- Center of Excellence for Stress and Mental Health, San Diego, California, USA
- Department of Psychiatry, University of California-San Diego, San Diego, California, USA
| | - Cole Vonder Haar
- Department of Psychology, West Virginia University, Morgantown, West Virginia, USA
- Injury and Recovery Laboratory, Department of Neuroscience, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
15
|
Gober IG, Russell AL, Shick TJ, Vagni VA, Carlson JC, Kochanek PM, Wagner AK. Exploratory assessment of the effect of systemic administration of soluble glycoprotein 130 on cognitive performance and chemokine levels in a mouse model of experimental traumatic brain injury. J Neuroinflammation 2024; 21:149. [PMID: 38840141 PMCID: PMC11155101 DOI: 10.1186/s12974-024-03129-0] [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: 02/28/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024] Open
Abstract
Uncontrolled neuroinflammation mediates traumatic brain injury (TBI) pathology and impairs recovery. Interleukin-6 (IL-6), a pleiotropic inflammatory regulator, is associated with poor clinical TBI outcomes. IL-6 operates via classical-signaling through membrane-bound IL-6 receptor (IL-6R) and trans-signaling through soluble IL-6 receptor (s)IL-6R. IL-6 trans-signaling specifically contributes to neuropathology, making it a potential precision therapeutic TBI target. Soluble glycoprotein 130 (sgp130) prevents IL-6 trans-signaling, sparing classical signaling, thus is a possible treatment. Mice received either controlled cortical impact (CCI) (6.0 ± 0.2 m/s; 2 mm; 50-60ms) or sham procedures. Vehicle (VEH) or sgp130-Fc was subcutaneously administered to sham (VEH or 1 µg) and CCI (VEH, 0.25 µg or 1 µg) mice on days 1, 4, 7, 10 and 13 post-surgery to assess effects on cognition [Morris Water Maze (MWM)] and ipsilateral hemisphere IL-6 related biomarkers (day 21 post-surgery). CCI + sgp130-Fc groups (0.25 µg and 1 µg) were combined for analysis given similar behavior/biomarker outcomes. CCI + VEH mice had longer latencies and path lengths to the platform and increased peripheral zone time versus Sham + VEH and Sham + sgp130-Fc mice, suggesting injury-induced impairments in learning and anxiety. CCI + sgp130-Fc mice had shorter platform latencies and path lengths and had decreased peripheral zone time, indicating a therapeutic benefit of sgp130-Fc after injury on learning and anxiety. Interestingly, Sham + sgp130-Fc mice had shorter platform latencies, path lengths and peripheral zone times than Sham + VEH mice, suggesting a beneficial effect of sgp130-Fc, independent of injury. CCI + VEH mice had increased brain IL-6 and decreased sgp130 levels versus Sham + VEH and Sham + sgp130-Fc mice. There was no treatment effect on IL-6, sIL6-R or sgp130 in Sham + VEH versus Sham + sgp130-Fc mice. There was also no treatment effect on IL-6 in CCI + VEH versus CCI + sgp130-Fc mice. However, CCI + sgp130-Fc mice had increased sIL-6R and sgp130 versus CCI + VEH mice, demonstrating sgp130-Fc treatment effects on brain biomarkers. Inflammatory chemokines (MIP-1β, IP-10, MIG) were increased in CCI + VEH mice versus Sham + VEH and Sham + sgp130-Fc mice. However, CCI + sgp130-Fc mice had decreased chemokine levels versus CCI + VEH mice. IL-6 positively correlated, while sgp130 negatively correlated, with chemokine levels. Overall, we found that systemic sgp130-Fc treatment after CCI improved learning, decreased anxiety and reduced CCI-induced brain chemokines. Future studies will explore sex-specific dosing and treatment mechanisms for sgp130-Fc therapy.
Collapse
Affiliation(s)
- Ian G Gober
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Ashley L Russell
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Tyler J Shick
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Vincent A Vagni
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenna C Carlson
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA.
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA.
- Center for Neuroscience, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Fox R, Santana-Gomez C, Shamas M, Pavade A, Staba R, Harris NG. Different Trajectories of Functional Connectivity Captured with Gamma-Event Coupling and Broadband Measures of EEG in the Rat Fluid Percussion Injury Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597056. [PMID: 38895342 PMCID: PMC11185526 DOI: 10.1101/2024.06.02.597056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Functional connectivity (FC) after TBI is affected by an altered excitatory-inhibitory balance due to neuronal dysfunction, and the mechanistic changes observed could be reflected differently by contrasting methods. Local gamma event coupling FC (GEC-FC) is believed to represent multiunit fluctuations due to inhibitory dysfunction, and we hypothesized that FC derived from widespread, broadband amplitude signal (BBA-FC) would be different, reflecting broader mechanisms of functional disconnection. We tested this during sleep and active periods defined by high delta and theta EEG activity, respectively, at 1,7 and 28d after rat fluid-percussion-injury (FPI) or sham injury (n=6/group) using 10 indwelling, bilateral cortical and hippocampal electrodes. We also measured seizure and high-frequency oscillatory activity (HFOs) as markers of electrophysiological burden. BBA-FC analysis showed early hyperconnectivity constrained to ipsilateral sensory-cortex-to-CA1-hippocampus that transformed to mainly ipsilateral FC deficits by 28d compared to shams. These changes were conserved over active epochs, except at 28d when there were no differences to shams. In comparison, GEC-FC analysis showed large regions of hyperconnectivity early after injury within similar ipsilateral and intrahemispheric networks. GEC-FC weakened with time, but hyperconnectivity persisted at 28d compared to sham. Edge- and global connectivity measures revealed injury-related differences across time in GEC-FC as compared to BBA-FC, demonstrating greater sensitivity to FC changes post-injury. There was no significant association between sleep fragmentation, HFOs, or seizures with FC changes. The within-animal, spatial-temporal differences in BBA-FC and GEC-FC after injury may represent different mechanisms driving FC changes as a result of primary disconnection and interneuron loss. Significance statement The present study adds to the understanding of functional connectivity changes in preclinical models of traumatic brain injury. In previously reported literature, there is heterogeneity in the directionality of connectivity changes after injury, resulting from factors such as severity of injury, frequency band studied, and methodology used to calculate FC. This study aims to further clarify differential mechanisms that result in altered network topography after injury, by using Broadband Amplitude-Derived FC and Gamma Event Coupling-Derived FC in EEG. We found post-injury changes that differ in complexity and directionality between measures at and across timepoints. In conjunction with known results and future studies identifying different neural drivers underlying these changes, measures derived from this study could provide useful means from which to minimally-invasively study temporally-evolving pathology after TBI.
Collapse
|
17
|
Markicevic M, Mandino F, Toyonaga T, Cai Z, Fesharaki-Zadeh A, Shen X, Strittmatter SM, Lake E. Repetitive mild closed-head injury induced synapse loss and increased local BOLD-fMRI signal homogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595651. [PMID: 38826468 PMCID: PMC11142233 DOI: 10.1101/2024.05.24.595651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Repeated mild head injuries due to sports, or domestic violence and military service are increasingly linked to debilitating symptoms in the long term. Although symptoms may take decades to manifest, potentially treatable neurobiological alterations must begin shortly after injury. Better means to diagnose and treat traumatic brain injuries, requires an improved understanding of the mechanisms underlying progression and means through which they can be measured. Here, we employ a repetitive mild closed-head injury (rmTBI) and chronic variable stress (CVS) mouse model to investigate emergent structural and functional brain abnormalities. Brain imaging is achieved with [ 18 F]SynVesT-1 positron emission tomography, with the synaptic vesicle glycoprotein 2A ligand marking synapse density and BOLD (blood-oxygen-level-dependent) functional magnetic resonance imaging (fMRI). Animals were scanned six weeks after concluding rmTBI/Stress procedures. Injured mice showed widespread decreases in synaptic density coupled with an i ncrease in local BOLD-fMRI synchrony detected as regional homogeneity. Injury-affected regions with higher synapse density showed a greater increase in fMRI regional homogeneity. Taken together, these observations may reflect compensatory mechanisms following injury. Multimodal studies are needed to provide deeper insights into these observations.
Collapse
|
18
|
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
|
19
|
Ritzel RM, Li Y, Jiao Y, Doran SJ, Khan N, Henry RJ, Brunner K, Loane DJ, Faden AI, Szeto GL, Wu J. Bi-directional neuro-immune dysfunction after chronic experimental brain injury. J Neuroinflammation 2024; 21:83. [PMID: 38581043 PMCID: PMC10996305 DOI: 10.1186/s12974-024-03082-y] [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: 01/15/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND It is well established that traumatic brain injury (TBI) causes acute and chronic alterations in systemic immune function and that systemic immune changes contribute to posttraumatic neuroinflammation and neurodegeneration. However, how TBI affects bone marrow (BM) hematopoietic stem/progenitor cells chronically and to what extent such changes may negatively impact innate immunity and neurological function has not been examined. METHODS To further understand the role of BM cell derivatives on TBI outcome, we generated BM chimeric mice by transplanting BM from chronically injured or sham (i.e., 90 days post-surgery) congenic donor mice into otherwise healthy, age-matched, irradiated CD45.2 C57BL/6 (WT) hosts. Immune changes were evaluated by flow cytometry, multiplex ELISA, and NanoString technology. Moderate-to-severe TBI was induced by controlled cortical impact injury and neurological function was measured using a battery of behavioral tests. RESULTS TBI induced chronic alterations in the transcriptome of BM lineage-c-Kit+Sca1+ (LSK+) cells in C57BL/6 mice, including modified epigenetic and senescence pathways. After 8 weeks of reconstitution, peripheral myeloid cells from TBI→WT mice showed significantly higher oxidative stress levels and reduced phagocytic activity. At eight months after reconstitution, TBI→WT chimeric mice were leukopenic, with continued alterations in phagocytosis and oxidative stress responses, as well as persistent neurological deficits. Gene expression analysis revealed BM-driven changes in neuroinflammation and neuropathology after 8 weeks and 8 months of reconstitution, respectively. Chimeric mice subjected to TBI at 8 weeks and 8 months post-reconstitution showed that longer reconstitution periods (i.e., time post-injury) were associated with increased microgliosis and leukocyte infiltration. Pre-treatment with a senolytic agent, ABT-263, significantly improved behavioral performance of aged C57BL/6 mice at baseline, although it did not attenuate neuroinflammation in the acutely injured brain. CONCLUSIONS TBI causes chronic activation and progressive dysfunction of the BM stem/progenitor cell pool, which drives long-term deficits in hematopoiesis, innate immunity, and neurological function, as well as altered sensitivity to subsequent brain injury.
Collapse
Affiliation(s)
- Rodney M Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Yun Li
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yun Jiao
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, Baltimore, MD, 21250, USA
| | - Sarah J Doran
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Niaz Khan
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kavitha Brunner
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Gregory L Szeto
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore County, Baltimore, MD, 21250, USA
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
20
|
Baucom MR, Price AD, England L, Schuster RM, Pritts TA, Goodman MD. Murine Traumatic Brain Injury Model Comparison: Closed Head Injury Versus Controlled Cortical Impact. J Surg Res 2024; 296:230-238. [PMID: 38295710 DOI: 10.1016/j.jss.2024.01.002] [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: 08/23/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 03/19/2024]
Abstract
INTRODUCTION Various murine models have been utilized to study TBI, including closed head injury (CHI) and controlled cortical impact (CCI), without direct comparison. The aim of our study was to evaluate these models to determine differences in neurological and behavioral outcomes postinjury. METHODS Male C57B/6 mice (9-10 wk) were separated into six groups including: untouched, sham craniotomy (4 mm), CCI 0.9 mm depth of impact, CCI 1.6 mm, CCI 2.2 mm, and CHI. CCI was performed using a 3 mm impact tip at a velocity of 5 m/s, dwell time of 250 ms, and depth as noted above. CHI was completed with a centered 400 g weight drop from 1 cm height. Mice were survived to 14-d (n = 5 per group) and 30-d (n = 5 per group) respectively for histological analysis of p-tau within the hippocampus. These mice underwent Morris Water Maze memory testing and Rotarod motor testing. Serum was collected from a separate cohort of mice (n = 5 per group) including untouched, isoflurane only, CCI 1.6 mm, CHI at 1, 4, 6, and 24 h for analysis of neuron specific enolase and glial fibrillary acidic protein (GFAP) via ELISA. Laser speckle contrast imaging was analyzed prior to and after impact in the CHI and CCI 1.6 mm groups. RESULTS There were no significant differences in Morris Water Maze or Rotarod testing times between groups at 14- or 30-d. P-tau was significantly elevated in all groups except CCI 1.6 mm contralateral and CCI 2.2 mm ipsilateral compared to untouched mice at 30-d. P-tau was also significantly elevated in the CHI group at 30 d compared to CCI 1.6 mm contralateral and CCI 2.2 mm on both sides. GFAP was significantly increased in mice undergoing CHI (9959 ± 91 pg/mL) compared to CCI (2299 ± 1288 pg/mL), isoflurane only (133 ± 75 pg/mL), and sham (86 ± 58 pg/mL) at 1-h post TBI (P < 0.0001). There were no differences in serum neuron specific enolase levels between groups. Laser doppler imaging demonstrated similar decreases in cerebral blood flow between CHI and CCI; however, CCI mice had a reduction in blood flow with craniotomy only that did not significantly decrease further with impact. CONCLUSIONS Based on our findings, CHI leads to increased serum GFAP levels and increased p-tau within the hippocampus at 30-d postinjury. While CCI allows the comparison of one cerebral hemisphere to the other, CHI may be a better model of TBI as it requires less technical expertise and has similar neurological outcomes in these murine models.
Collapse
Affiliation(s)
- Matthew R Baucom
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Adam D Price
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Lisa England
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | - Timothy A Pritts
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | |
Collapse
|
21
|
Snyder K, Dixon CE, Henchir J, Gorse K, Vagni VA, Janesko-Feldman K, Kochanek PM, Jackson TC. Gene knockout of RNA binding motif 5 in the brain alters RIMS2 protein homeostasis in the cerebellum and Hippocampus and exacerbates behavioral deficits after a TBI in mice. Exp Neurol 2024; 374:114690. [PMID: 38218585 PMCID: PMC11178365 DOI: 10.1016/j.expneurol.2024.114690] [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: 10/19/2023] [Revised: 12/28/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
RNA binding motif 5 (RBM5) is a tumor suppressor in cancer but its role in the brain is unclear. We used conditional gene knockout (KO) mice to test if RBM5 inhibition in the brain affects chronic cortical brain tissue survival or function after a controlled cortical impact (CCI) traumatic brain injury (TBI). RBM5 KO decreased baseline contralateral hemispheric volume (p < 0.0001) and exacerbated ipsilateral tissue loss at 21 d after CCI in male mice vs. wild type (WT) (p = 0.0019). CCI injury, but not RBM5 KO, impaired beam balance performance (0-5d post-injury) and swim speed on the Morris Water Maze (MWM) (19-20d) (p < 0.0001). RBM5 KO was associated with mild learning impairment in female mice (p = 0.0426), reflected as a modest increase in escape latency early in training (14-18d post-injury). However, KO did not affect spatial memory at 19d post-injury in male or in female mice but it was impaired by CCI in females (p = 0.0061). RBM5 KO was associated with impaired visual function in male mice on the visible platform test at 20d post-injury (p = 0.0256). To explore signaling disturbances in KOs related to behavior, we first cross-referenced known brain-specific RBM5-regulated gene targets with genes in the curated RetNet database that impact vision. We then performed a secondary literature search on RBM5-regulated genes with a putative role in hippocampal function. Regulating synaptic membrane exocytosis 2 (RIMS) 2 was identified as a gene of interest because it regulates both vision and hippocampal function. Immunoprecipitation and western blot confirmed protein expression of a novel ~170 kDa RIMS2 variant in the cerebellum, and in the hippocampus, it was significantly increased in KO vs WT (p < 0.0001), and in a sex-dependent manner (p = 0.0390). Furthermore, male KOs had decreased total canonical RIMS2 levels in the cerebellum (p = 0.0027) and hippocampus (p < 0.0001), whereas female KOs had increased total RIMS1 levels in the cerebellum (p = 0.0389). In summary, RBM5 modulates brain function in mammals. Future work is needed to test if RBM5 dependent regulation of RIMS2 splicing effects vision and cognition, and to verify potential sex differences on behavior in a larger cohort of mice.
Collapse
Affiliation(s)
- Kara Snyder
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL 33602, United States of America; University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, United States of America.
| | - C Edward Dixon
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Jeremy Henchir
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Kiersten Gorse
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL 33602, United States of America; University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, United States of America.
| | - Vincent A Vagni
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Travis C Jackson
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL 33602, United States of America; University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, United States of America.
| |
Collapse
|
22
|
Huerta de la Cruz S, Santiago-Castañeda C, Rodríguez-Palma EJ, Rocha L, Sancho M. Lateral fluid percussion injury: A rat model of experimental traumatic brain injury. Methods Cell Biol 2024; 185:197-224. [PMID: 38556449 DOI: 10.1016/bs.mcb.2024.02.011] [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] [Indexed: 04/02/2024]
Abstract
Traumatic brain injury (TBI) represents one of the leading causes of disability and death worldwide. The annual economic impact of TBI-including direct and indirect costs-is high, particularly impacting low- and middle-income countries. Despite extensive research, a comprehensive understanding of the primary and secondary TBI pathophysiology, followed by the development of promising therapeutic approaches, remains limited. These fundamental caveats in knowledge have motivated the development of various experimental models to explore the molecular mechanisms underpinning the pathogenesis of TBI. In this context, the Lateral Fluid Percussion Injury (LFPI) model produces a brain injury that mimics most of the neurological and systemic aspects observed in human TBI. Moreover, its high reproducibility makes the LFPI model one of the most widely used rodent-based TBI models. In this chapter, we provide a detailed surgical protocol of the LFPI model used to induce TBI in adult Wistar rats. We further highlight the neuroscore test as a valuable tool for the evaluation of TBI-induced sensorimotor consequences and their severity in rats. Lastly, we briefly summarize the current knowledge on the pathological aspects and functional outcomes observed in the LFPI-induced TBI model in rodents.
Collapse
Affiliation(s)
- Saúl Huerta de la Cruz
- Department of Pharmacology, University of Vermont, Burlington, VT, United States; Departamento de Farmacobiología, Cinvestav Sede Sur, Ciudad de México, México.
| | | | - Erick J Rodríguez-Palma
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Sede Sur, Mexico City, Mexico
| | - Luisa Rocha
- Departamento de Farmacobiología, Cinvestav Sede Sur, Ciudad de México, México
| | - Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT, United States; Department of Physiology, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
23
|
Fringuello AR, Colbourn R, Goodman JH, Michelson HB, Ling DSF, Hrabetova S. Rapid volume pulsations of the extracellular space accompany epileptiform activity in trauma-injured neocortex and depend on the sodium-bicarbonate cotransporter NBCe1. Epilepsy Res 2024; 201:107337. [PMID: 38461594 DOI: 10.1016/j.eplepsyres.2024.107337] [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: 09/29/2023] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024]
Abstract
Post traumatic epilepsy (PTE) is a treatment-resistant consequence of traumatic brain injury (TBI). Recently, it has been revealed that epileptiform activity in acute chemoconvulsant seizure models is accompanied by transient shrinkages of extracellular space (ECS) called rapid volume pulsations (RVPs). Shrinkage of the ECS surrounding neurons and glia may contribute to ictogenic hyperexcitability and hypersynchrony during the chronic phase of TBI. Here, we identify the phenomenon of RVPs occurring spontaneously in rat neocortex at ≥ 3 weeks after injury in the controlled cortical impact (CCI) model for PTE. We further report that blocking the electrogenic action of the astrocytic cotransporter NBCe1 with 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS) eliminates both RVPs and epileptiform activity in ex-vivo CCI neocortical brain slices. We conclude that NBCe1-mediated extracellular volume shrinkage may represent a new target for therapeutic intervention in PTE.
Collapse
Affiliation(s)
- Anthony R Fringuello
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA; Neural and Behavioral Science Graduate Program, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Robert Colbourn
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA; Neural and Behavioral Science Graduate Program, SUNY Downstate Health Sciences University, Brooklyn, NY, USA; Present address: Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey H Goodman
- Department of Developmental Neurobiology, The New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA; Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA; The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Hillary B Michelson
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA; The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Douglas S F Ling
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA; The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Sabina Hrabetova
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA; The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Health Sciences University, Brooklyn, NY, USA.
| |
Collapse
|
24
|
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
|
25
|
Vita SM, Cruise SC, Gilpin NW, Molina PE. Histological comparison of repeated mild weight drop and lateral fluid percussion injury models of traumatic brain injury (TBI) in female and male rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578177. [PMID: 38352449 PMCID: PMC10862833 DOI: 10.1101/2024.01.31.578177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Traumatic brain injury (TBI) heterogeneity has led to the development of several preclinical models, each modeling a distinct subset of outcomes. Selection of an injury model should be guided by the research question and the specific outcome measures of interest. Consequently, there is a need for conducting direct comparisons of different TBI models. Here, we used immunohistochemistry to directly compare the outcomes from two common models, lateral fluid percussion (LFP) and repeat mild weight drop (rmWD), on neuropathology in adult female and male Wistar rats. Specifically, we used immunohistochemistry to measure the effects of LFP and rmWD on cerebrovascular and tight junction disruption, inflammatory markers, mature neurons and perineuronal nets in the cortical site of injury, cortex adjacent to injury, dentate gyrus, and the CA2/3 area of the hippocampus. Animals were randomized into either LFP or rmWD groups. The LFP group received a craniotomy prior to LFP (or corresponding sham procedure) three days later, while rmWD animals underwent either weight drop or sham (isoflurane only) on each of those four days. After a recovery period of 7 days, animals were euthanized, and brains were harvested for analysis of RECA-1, claudin-5, GFAP, Iba-1, CD-68, NeuN, and wisteria floribunda lectin. Overall, our observations revealed that the most significant disruptions were evident in response to LFP, followed by craniotomy-only, while rmWD animals showed the least residual changes compared to isoflurane-only controls. These findings support consideration of rmWD as a mild, transient injury. LFP leads to longer-lasting disruptions that are more closely associated with a moderate TBI. We further show that both craniotomy and LFP produced greater disruptions in females relative to males at 7 days post-injury. These findings support the inclusion of a time-matched experimentally-naïve or anesthesia-only control group in preclinical TBI research to enhance the validity of data interpretation and conclusions.
Collapse
|
26
|
Nespoli E, Hakani M, Hein TM, May SN, Danzer K, Wirth T, Baumann B, Dimou L. Glial cells react to closed head injury in a distinct and spatiotemporally orchestrated manner. Sci Rep 2024; 14:2441. [PMID: 38286816 PMCID: PMC10825139 DOI: 10.1038/s41598-024-52337-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 01/17/2024] [Indexed: 01/31/2024] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and disability worldwide. Acute neuroinflammation is a prominent reaction after TBI and is mostly initiated by brain-resident glial cells such as microglia, NG2-glia and astrocytes. The magnitude of this reaction paves the way for long-lasting consequences such as chronic neurological pathologies, for which therapeutic options remain limited. The neuroinflammatory response to TBI is mostly studied with craniotomy-based animal models that are very robust but also rather artificial. Here, we aimed to analyze the reaction of glial cells in a highly translational but variable closed head injury (CHI) model and were able to correlate the severity of the trauma to the degree of glial response. Furthermore, we could show that the different glial cell types react in a temporally and spatially orchestrated manner in terms of morphological changes, proliferation, and cell numbers in the first 15 days after the lesion. Interestingly, NG2-glia, the only proliferating cells in the healthy brain parenchyma, divided at a rate that was correlated with the size of the injury. Our findings describe the previously uncharacterized posttraumatic response of the major brain glial cell types in CHI in order to gain a detailed understanding of the course of neuroinflammatory events; such knowledge may open novel avenues for future therapeutic approaches in TBI.
Collapse
Affiliation(s)
- Ester Nespoli
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany
| | - Marsela Hakani
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany
| | | | | | - Karin Danzer
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DNZE), Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Leda Dimou
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany.
| |
Collapse
|
27
|
Wei S, Jiang A, Sun H, Zhu J, Jia S, Liu X, Xu Z, Zhang J, Shang Y, Fu X, Li G, Wang P, Xia Z, Jiang T, Cao A, Duan X. Shape-changing electrode array for minimally invasive large-scale intracranial brain activity mapping. Nat Commun 2024; 15:715. [PMID: 38267440 PMCID: PMC10808108 DOI: 10.1038/s41467-024-44805-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
Large-scale brain activity mapping is important for understanding the neural basis of behaviour. Electrocorticograms (ECoGs) have high spatiotemporal resolution, bandwidth, and signal quality. However, the invasiveness and surgical risks of electrode array implantation limit its application scope. We developed an ultrathin, flexible shape-changing electrode array (SCEA) for large-scale ECoG mapping with minimal invasiveness. SCEAs were inserted into cortical surfaces in compressed states through small openings in the skull or dura and fully expanded to cover large cortical areas. MRI and histological studies on rats proved the minimal invasiveness of the implantation process and the high chronic biocompatibility of the SCEAs. High-quality micro-ECoG activities mapped with SCEAs from male rodent brains during seizures and canine brains during the emergence period revealed the spatiotemporal organization of different brain states with resolution and bandwidth that cannot be achieved using existing noninvasive techniques. The biocompatibility and ability to map large-scale physiological and pathological cortical activities with high spatiotemporal resolution, bandwidth, and signal quality in a minimally invasive manner offer SCEAs as a superior tool for applications ranging from fundamental brain research to brain-machine interfaces.
Collapse
Affiliation(s)
- Shiyuan Wei
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Anqi Jiang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Hongji Sun
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Jingjun Zhu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
- National Biomedical Imaging Centre, Peking University, Beijing, 100871, China
| | - Shengyi Jia
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Xiaojun Liu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Zheng Xu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Jing Zhang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yuanyuan Shang
- Key Laboratory of Material Physics, Ministry of Education, School of Physics and Microelectronics, Zhengzhou University, Zhengzhou, 450052, China
| | - Xuefeng Fu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Gen Li
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Puxin Wang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Zhiyuan Xia
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Tianzi Jiang
- Brainnetome Centre, Institute of Automation, Chinese Academy of Sciences (CAS), Beijing, 100190, China
| | - Anyuan Cao
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Xiaojie Duan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China.
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
- National Biomedical Imaging Centre, Peking University, Beijing, 100871, China.
| |
Collapse
|
28
|
Alanezi ST, Almutairi WM, Cronin M, Gobbo O, O'Mara SM, Sheppard D, O'Connor WT, Gilchrist MD, Kleefeld C, Colgan N. Whole-brain traumatic controlled cortical impact to the left frontal lobe: Magnetic resonance image-based texture analysis. J Neuropathol Exp Neurol 2024; 83:94-106. [PMID: 38164986 DOI: 10.1093/jnen/nlad110] [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] [Indexed: 01/03/2024] Open
Abstract
This research assesses the capability of texture analysis (TA) derived from high-resolution (HR) T2-weighted magnetic resonance imaging to identify primary sequelae following 1-5 hours of controlled cortical impact mild or severe traumatic brain injury (TBI) to the left frontal cortex (focal impact) and secondary (diffuse) sequelae in the right frontal cortex, bilateral corpus callosum, and hippocampus in rats. The TA technique comprised first-order (histogram-based) and second-order statistics (including gray-level co-occurrence matrix, gray-level run length matrix, and neighborhood gray-level difference matrix). Edema in the left frontal impact region developed within 1 hour and continued throughout the 5-hour assessments. The TA features from HR images confirmed the focal injury. There was no significant difference among radiomics features between the left and right corpus callosum or hippocampus from 1 to 5 hours following a mild or severe impact. The adjacent corpus callosum region and the distal hippocampus region (s), showed no diffuse injury 1-5 hours after mild or severe TBI. These results suggest that combining HR images with TA may enhance detection of early primary and secondary sequelae following TBI.
Collapse
Affiliation(s)
- Saleh T Alanezi
- Physics Department, Faculty of Science, Northern Border University, ArAr, Saudi Arabia
- School of Natural Sciences, College of Science and Engineering, University of Galway, Galway, Ireland
| | - Waleed M Almutairi
- Medical Imaging Department, King Abdullah bin Abdulaziz University Hospital, Riyadh, Saudi Arabia
- Department of Physics, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Michelle Cronin
- Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Oliviero Gobbo
- School of Pharmacy and Pharmaceutical Sciences & Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Shane M O'Mara
- Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Declan Sheppard
- Department of Radiology, University Hospital Galway, Galway, Ireland
| | - William T O'Connor
- University of Limerick School of Medicine, Castletroy, Limerick, Ireland
| | - Michael D Gilchrist
- School of Mechanical & Materials Engineering, University College Dublin, Belfield, Dublin, Ireland
| | - Christoph Kleefeld
- School of Natural Sciences, College of Science and Engineering, University of Galway, Galway, Ireland
| | - Niall Colgan
- School of Natural Sciences, College of Science and Engineering, University of Galway, Galway, Ireland
- Department of Engineering, Technological University of the Shannon, Athlone, Ireland
| |
Collapse
|
29
|
Grandizoli Saletti P, Casillas-Espinosa PM, Panagiotis Lisgaras C, Bi Mowrey W, Li Q, Liu W, Brady RD, Ali I, Silva J, Yamakawa G, Hudson M, Li C, Braine EL, Coles L, Cloyd JC, Jones NC, Shultz SR, Moshé SL, O'Brien TJ, Galanopoulou AS. Tau Phosphorylation Patterns in the Rat Cerebral Cortex After Traumatic Brain Injury and Sodium Selenate Effects: An Epibios4rx Project 2 Study. J Neurotrauma 2024; 41:222-243. [PMID: 36950806 PMCID: PMC11079442 DOI: 10.1089/neu.2022.0219] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
Sodium selenate (SS) activates protein phosphatase 2 (PP2A) and reduces phosphorylated tau (pTAU) and late post-traumatic seizures after lateral fluid percussion injury (LFPI). In EpiBioS4Rx Project 2, a multi-center international study for post-traumatic targets, biomarkers, and treatments, we tested the target relevance and modification by SS of pTAU forms and PP2A and in the LFPI model, at two sites: Einstein and Melbourne. In Experiment 1, adult male rats were assigned to LFPI and sham (both sites) and naïve controls (Einstein). Motor function was monitored by neuroscores. Brains were studied with immunohistochemistry (IHC), Western blots (WBs), or PP2A activity assay, from 2 days to 8 weeks post-operatively. In Experiment 2, LFPI rats received SS for 7 days (SS0.33: 0.33 mg/kg/day; SS1: 1 mg/kg/day, subcutaneously) or vehicle (Veh) post-LFPI and pTAU, PR55 expression, or PP2A activity were studied at 2 days and 1 week (on treatment), or 2 weeks (1 week off treatment). Plasma selenium and SS levels were measured. In Experiment 1 IHC, LFPI rats had higher cortical pTAU-Ser202/Thr205-immunoreactivity (AT8-ir) and pTAU-Ser199/202-ir at 2 days, and pTAU-Thr231-ir (AT180-ir) at 2 days, 2 weeks, and 8 weeks, ipsilaterally to LFPI, than controls. LFPI-2d rats also had higher AT8/total-TAU5-ir in cortical extracts ipsilateral to the lesion (WB). PP2A (PR55-ir) showed time- and region-dependent changes in IHC, but not in WB. PP2A activity was lower in LFPI-1wk than in sham rats. In Experiment 2, SS did not affect neuroscores or cellular AT8-ir, AT180-ir, or PR55-ir in IHC. In WB, total cortical AT8/total-TAU-ir was lower in SS0.33 and SS1 LFPI rats than in Veh rats (2 days, 1 week); total cortical PR55-ir (WB) and PP2A activity were higher in SS1 than Veh rats (2 days). SS dose dependently increased plasma selenium and SS levels. Concordant across-sites data confirm time and pTAU form-specific cortical increases ipsilateral to LFPI. The discordant SS effects may either suggest SS-induced reduction in the numbers of cells with increased pTAU-ir, need for longer treatment, or the involvement of other mechanisms of action.
Collapse
Affiliation(s)
- Patricia Grandizoli Saletti
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Pablo M. Casillas-Espinosa
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Christos Panagiotis Lisgaras
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Wenzhu Bi Mowrey
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx New York, USA
| | - Qianyun Li
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Wei Liu
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Rhys D. Brady
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Idrish Ali
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Juliana Silva
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Glenn Yamakawa
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Matt Hudson
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Crystal Li
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Emma L. Braine
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Lisa Coles
- University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - James C. Cloyd
- University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx New York, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx New York, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx New York, USA
| | - Terence J. O'Brien
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx New York, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx New York, USA
| |
Collapse
|
30
|
da Silva Fiorin F, Cunha do Espírito Santo C, Santos do Nascimento R, França AP, Freire Royes LF. Behavioral deficits after mild traumatic brain injury by fluid percussion in rats. Neurosci Lett 2024; 818:137550. [PMID: 37926292 DOI: 10.1016/j.neulet.2023.137550] [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: 09/04/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023]
Abstract
Mild traumatic brain injury (TBI) can lead to various disorders, encompassing cognitive and psychiatric complications. While pre-clinical studies have long investigated behavioral alterations, the fluid percussion injury (FPI) model still lacks a comprehensive behavioral battery that includes psychiatric-like disorders. To address this gap, we conducted multiple behavioral tasks over two months in adult male Wistar rats, focusing on mild FPI. Statistical analyses revealed that both naive and sham animals exhibited an increase in sweet liquid consumption over time. In contrast, the TBI group did not show any temporal changes, although mild FPI did induce a statistically significant decrease in sucrose consumption compared to control groups during the chronic phase. Additionally, social interaction tasks indicated reduced contact time in TBI animals. The elevated plus maze task demonstrated an increase in open-arm exploration following fluid percussion. Nonetheless, no significant differences were observed in the acute and chronic phases for the forced swim and light-dark box tasks. Evaluation of three distinct memory tasks in the chronic phase revealed that mild FPI led to long-term memory deficits, as assessed by the object recognition task, while the surgical procedure itself resulted in short-term spatial memory deficits, as evaluated by the Y-maze task. Conversely, working memory remained unaffected in the water maze task. Collectively, these findings provide a nuanced characterization of behavioral deficits induced by mild FPI.
Collapse
Affiliation(s)
- Fernando da Silva Fiorin
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Neurobiologia da Dor e Inflamação, Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Bioquímica do Exercício, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| | - Caroline Cunha do Espírito Santo
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Neurobiologia da Dor e Inflamação, Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Raphael Santos do Nascimento
- Instituto de Engenharia Biomédica, Departamento de Engenharia Elétrica e Eletrônica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Angela Patricia França
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Luiz Fernando Freire Royes
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Bioquímica do Exercício, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
31
|
Oshima K, Siddiqui N, Orfila JE, Carter D, Laing J, Han X, Zakharevich I, Iozzo RV, Ghasabyan A, Moore H, Zhang F, Linhardt RJ, Moore EE, Quillinan N, Schmidt EP, Herson PS, Hippensteel JA. A role for decorin in improving motor deficits after traumatic brain injury. Matrix Biol 2024; 125:88-99. [PMID: 38135163 PMCID: PMC10922985 DOI: 10.1016/j.matbio.2023.12.005] [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: 10/09/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability due to injury worldwide. Extracellular matrix (ECM) remodeling is known to significantly contribute to TBI pathophysiology. Glycosaminoglycans, which are long-chain, variably sulfated polysaccharides abundant within the ECM, have previously been shown to be substantially altered after TBI. In this study, we sought to delineate the dynamics of glycosaminoglycan alterations after TBI and discover the precise biologic processes responsible for observed glycosaminoglycan changes after injury. We performed state-of-the art mass spectrometry on brain tissues isolated from mice after TBI or craniotomy-alone. We observed dynamic changes in glycosaminoglycans at Day 1 and 7 post-TBI, with heparan sulfate, chondroitin sulfate, and hyaluronan remaining significantly increased after a week vis-à-vis craniotomy-alone tissues. We did not observe appreciable changes in circulating glycosaminoglycans in mice after experimental TBI compared to craniotomy-alone nor in patients with TBI and severe polytrauma compared to control patients with mild injuries, suggesting increases in injury site glycosaminoglycans are driven by local synthesis. We subsequently performed an unbiased whole genome transcriptomics analysis on mouse brain tissues 7 days post-TBI and discovered a significant induction of hyaluronan synthase 2, glypican-3, and decorin. The functional role of decorin after injury was further examined through multimodal behavioral testing comparing wild-type and Dcn-/- mice. We discovered that genetic ablation of Dcn led to an overall negative effect of TBI on function, exacerbating motor impairments after TBI. Collectively, our results provide a spatiotemporal characterization of post-TBI glycosaminoglycan alterations in the brain ECM and support an important adaptive role for decorin upregulation after TBI.
Collapse
Affiliation(s)
- Kaori Oshima
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Noah Siddiqui
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James E Orfila
- Department of Neurosurgery, The Ohio State University, College of Medicine, Columbus, Ohio, USA
| | - Danelle Carter
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Justin Laing
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Xiaorui Han
- Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA; Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Igor Zakharevich
- Department of Biochemistry, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Renato V Iozzo
- Department of Pathology and Genomic Medicine and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Arsen Ghasabyan
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, University of Colorado, Denver, Colorado, USA
| | - Hunter Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, University of Colorado, Denver, Colorado, USA
| | - Fuming Zhang
- Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Robert J Linhardt
- Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Ernest E Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, University of Colorado, Denver, Colorado, USA
| | - Nidia Quillinan
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Eric P Schmidt
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paco S Herson
- Department of Neurosurgery, The Ohio State University, College of Medicine, Columbus, Ohio, USA
| | - Joseph A Hippensteel
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
32
|
Wu N, Li L. A review on wound management strategies in enhanced recovery after craniotomy: An in-depth analysis of their influence on patient recovery and surgical outcomes. Int Wound J 2024; 21:e14595. [PMID: 38272808 PMCID: PMC10789584 DOI: 10.1111/iwj.14595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/27/2024] Open
Abstract
Craniotomy, an essential neurosurgical operation, poses distinct difficulties in the realm of post-operative care, specifically with regard to the management of wounds. Efficient wound management is critical in order to optimize the surgical outcomes, reduce complications and facilitate a speedier recovery. The purpose of this comprehensive review was to assess contemporary wound management approaches as they pertain to improved recovery following craniotomy. This was achieved by contrasting conventional methods with more recent and innovative techniques and analysing the effects of these approaches on patient recovery and surgical results. An exhaustive literature search was undertaken, comprising narrative reviews, clinical studies, peer-reviewed articles and expert opinions. The emphasis was on the evolution of wound management strategies and techniques utilized after cranial section, as well as their contributions to patient recovery. The analysis reveals that while conventional wound management methods, including suturing and antiseptics, continue to be essential, innovative strategies such as negative pressure wound therapy, skin adhesives and advanced pain management protocols are becoming increasingly recognized. It has been demonstrated that these novel approaches improve recovery by decreasing the incidence of infections, enhancing patient comfort and producing superior cosmetic results. Nevertheless, obstacles continue to endure, including patient-specific variables, technological and financial considerations and the enduring consequences of recovery. Thus the treatment of wounds during craniotomy recuperation necessitates an integrated strategy that incorporates conventional techniques alongside contemporary advancements. Progress in this domain necessitates the customization of approaches to suit the unique requirements of each patient, the resolution of identified obstacles and an emphasis on ongoing investigation and interdisciplinary cooperation. The ever-changing terrain of wound management approaches underscores the ever-changing character of neurosurgical treatment and the continuous endeavour to enhance patient results following cranial resection.
Collapse
Affiliation(s)
- Nan Wu
- Nursing Department, Sir Run Run Shaw HospitalZhejiang University School of MedicineZhejiangHangzhouChina
| | - Luping Li
- Nursing Department, Sir Run Run Shaw HospitalZhejiang University School of MedicineZhejiangHangzhouChina
| |
Collapse
|
33
|
Freeman-Jones E, Miller WH, Work LM, Fullerton JL. Polypathologies and Animal Models of Traumatic Brain Injury. Brain Sci 2023; 13:1709. [PMID: 38137157 PMCID: PMC10741988 DOI: 10.3390/brainsci13121709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Traumatic brain injury (TBI) is an important health issue for the worldwide population, as it causes long-term pathological consequences for a diverse group of individuals. We are yet to fully elucidate the significance of TBI polypathologies, such as neuroinflammation and tau hyperphosphorylation, and their contribution to the development of chronic traumatic encephalopathy (CTE) and other neurological conditions. To advance our understanding of TBI, it is necessary to replicate TBI in preclinical models. Commonly used animal models include the weight drop model; these methods model human TBI in various ways and in different animal species. However, animal models have not demonstrated their clinical utility for identifying therapeutic interventions. Many interventions that were successful in improving outcomes for animal models did not translate into clinical benefit for patients. It is important to review current animal models and discuss their strengths and limitations within a TBI context. Modelling human TBI in animals encounters numerous challenges, yet despite these barriers, the TBI research community is working to overcome these difficulties. Developments include advances in biomarkers, standardising, and refining existing models. This progress will improve our ability to model TBI in animals and, therefore, enhance our understanding of TBI and, potentially, how to treat it.
Collapse
Affiliation(s)
- Erin Freeman-Jones
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow G12 8QQ, UK; (E.F.-J.); (W.H.M.)
| | - William H. Miller
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow G12 8QQ, UK; (E.F.-J.); (W.H.M.)
| | - Lorraine M. Work
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK;
| | - Josie L. Fullerton
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK;
| |
Collapse
|
34
|
Rodríguez JA, Gutiérrez MI, Vera A, Hernández DA, Gutiérrez JM, Martínez-Fong D, Leija L. Protocol to Induce the Temporary Opening of the Blood-Brain Barrier with Short-Time Focused Ultrasound in Rats. Pharmaceutics 2023; 15:2733. [PMID: 38140074 PMCID: PMC10748005 DOI: 10.3390/pharmaceutics15122733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Brain neurodegenerative diseases are central nervous system (CNS) affections typically common in older adults. A new therapeutic approach for them consists of providing specific drugs to the CNS through blood circulation; however, the Blood-Brain Barrier (BBB) prevents almost 100% of neurotherapeutics from reaching the brain. There are indications that Focused Ultrasound (FUS), temporarily placed in the BBB, can achieve a controlled increase in temperature at its focus, allowing temporary, localized, and reversible opening of this barrier, which facilitates the temporary delivery of specific drugs. This work presents a FUS-based protocol for the local, temporary, and reversible opening of the BBB in Wistar rats. The proposed protocol specifies certain power, treatment times, and duty cycle to controllably increase the temperature at the region of interest, i.e., the substantia nigra. Numerical simulations using commercial software based on the finite element method were carried out to determine the optimal size of the craniotomies for nearly full-acoustic transmission. Experiments in rats were performed with the parameters used during computational simulations to determine the adequate opening of the BBB. For this, craniotomies of different sizes were made at coordinates of the substantia nigra, and FUS was applied from the exterior. The opening of the BBB was evaluated using Evans Blue (EB) as an indicator of the crossing of the dye from the blood vessels to brain tissue. Numerical simulations demonstrated a major distance reached by the ultrasound focus with a bigger diameter. Experimental results show the local, temporary, and reversible opening of the BBB through a 10 mm diameter craniotomy, which effectively allowed placing the ultrasound focus over the substantia nigra, unlike a 6 mm diameter craniotomy in which there is a deviation of the focus through that window. Moreover, from these results, it was also determined that the disruption of the BBB was reversible, with an opening duration of 6 h after FUS application. The experimental work developed in this study resulted in a minimally invasive method for the temporary opening of the BBB.
Collapse
Affiliation(s)
- Jorge A. Rodríguez
- Bioelectronics Section, Electrical Engineering Department, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico; (J.A.R.); (D.A.H.); (J.M.G.); (L.L.)
| | - Mario I. Gutiérrez
- Subdirección de Investigación Tecnológica, Consejo Nacional de Humanidades, Ciencias y Tecnologías-Instituto Nacional de Rehabilitación LGII, División de Investigación en Ingeniería Médica, Mexico City 14389, Mexico;
| | - Arturo Vera
- Bioelectronics Section, Electrical Engineering Department, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico; (J.A.R.); (D.A.H.); (J.M.G.); (L.L.)
| | - Daniel A. Hernández
- Bioelectronics Section, Electrical Engineering Department, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico; (J.A.R.); (D.A.H.); (J.M.G.); (L.L.)
| | - Juan M. Gutiérrez
- Bioelectronics Section, Electrical Engineering Department, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico; (J.A.R.); (D.A.H.); (J.M.G.); (L.L.)
| | - Daniel Martínez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, Programa de Nanociencias y Nanotecnología, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico;
| | - Lorenzo Leija
- Bioelectronics Section, Electrical Engineering Department, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico; (J.A.R.); (D.A.H.); (J.M.G.); (L.L.)
| |
Collapse
|
35
|
Szczygielski J, Hubertus V, Kruchten E, Müller A, Albrecht LF, Schwerdtfeger K, Oertel J. Prolonged course of brain edema and neurological recovery in a translational model of decompressive craniectomy after closed head injury in mice. Front Neurol 2023; 14:1308683. [PMID: 38053795 PMCID: PMC10694459 DOI: 10.3389/fneur.2023.1308683] [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: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
Background The use of decompressive craniectomy in traumatic brain injury (TBI) remains a matter of debate. According to the DECRA trial, craniectomy may have a negative impact on functional outcome, while the RescueICP trial revealed a positive effect of surgical decompression, which is evolving over time. This ambivalence of craniectomy has not been studied extensively in controlled laboratory experiments. Objective The goal of the current study was to investigate the prolonged effects of decompressive craniectomy (both positive and negative) in an animal model. Methods Male mice were assigned to the following groups: sham, decompressive craniectomy, TBI and TBI followed by craniectomy. The analysis of functional outcome was performed at time points 3d, 7d, 14d and 28d post trauma according to the Neurological Severity Score and Beam Balance Score. At the same time points, magnetic resonance imaging was performed, and brain edema was analyzed. Results Animals subjected to both trauma and craniectomy presented the exacerbation of the neurological impairment that was apparent mostly in the early course (up to 7d) after injury. Decompressive craniectomy also caused a significant increase in brain edema volume (initially cytotoxic with a secondary shift to vasogenic edema and gliosis). Notably, delayed edema plus gliosis appeared also after decompression even without preceding trauma. Conclusion In prolonged outcomes, craniectomy applied after closed head injury in mice aggravates posttraumatic brain edema, leading to additional functional impairment. This effect is, however, transient. Treatment options that reduce brain swelling after decompression may accelerate neurological recovery and should be explored in future experiments.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Instutute of Neuropathology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Institute of Medical Sciences, University of Rzeszów, Rzeszow, Poland
| | - Vanessa Hubertus
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Department of Neurosurgery, Charité University Medicine, Berlin, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| | - Eduard Kruchten
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Institute of Interventional and Diagnostic Radiology, Karlsruhe, Germany
| | - Andreas Müller
- Department of Radiology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Lisa Franziska Albrecht
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Karsten Schwerdtfeger
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| |
Collapse
|
36
|
Mejia-Bautista A, Michelson HB, Sanjana A, Famuyiwa O, Goodman JH, Ling DSF. 4-AP challenge reveals that early intervention with brivaracetam prevents posttraumatic epileptogenesis in rats. Epilepsy Res 2023; 196:107217. [PMID: 37619297 DOI: 10.1016/j.eplepsyres.2023.107217] [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: 06/30/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
PURPOSE There are currently no clinical treatments to prevent posttraumatic epilepsy (PTE). Recently, our group has shown that administration of levetiracetam (LEV) or brivaracetam (BRV) shortly after cortical neurotrauma prevents the development of epileptiform activity in rats, as measured ex vivo in neocortical slices. Due to the low incidence of spontaneous seizures in rodent-based models of traumatic brain injury (TBI), chemoconvulsants have been used to test injured animals for seizure susceptibility. We used a low dose of the voltage-gated potassium channel blocker 4-aminopyridine (4-AP) to evaluate posttraumatic epileptogenesis after controlled cortical impact (CCI) injury. We then used this assessment to further investigate the efficacy of BRV as an antiepileptogenic treatment. METHODS Sprague-Dawley rats aged P24-35 were subjected to severe CCI injury. Following trauma, one group received BRV-21 mg/kg (IP) at 0-2 min after injury and the other BRV-100 mg/kg (IP) at 30 min after injury. Four to eight weeks after injury, animals were given a single, low dose of 4-AP (3.0-3.5 mg/kg, IP) and then monitored up to 90 min for stage 4/5 seizures. RESULTS The chemoconvulsant challenge revealed that within four to eight weeks, CCI injury led to a two-fold increase in percentage of rats with 4-AP induced stage 4-5 seizures relative to sham-injured controls. Administration of a single dose of BRV within 30 min after trauma significantly reduced injury-induced seizure susceptibility, bringing the proportion of CCI-rats that exhibited evoked seizures down to control levels. CONCLUSIONS This study is the first to use a low dose of 4-AP as a chemoconvulsant challenge to test epileptogenicity within the first two months after CCI injury in rats. Our findings show that a single dose of BRV administered within 30 min after TBI prevents injury-induced increases in seizure susceptibility. This supports our hypothesis that early intervention with BRV may prevent PTE.
Collapse
Affiliation(s)
- Ana Mejia-Bautista
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Program in Neural and Behaviroal Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA.
| | - Hillary B Michelson
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Program in Neural and Behaviroal Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Anika Sanjana
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Program in Neural and Behaviroal Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Oluwafunmilayo Famuyiwa
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Jeffrey H Goodman
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Department of Neurology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Program in Neural and Behaviroal Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Department of Developmental Neurobiology, The New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
| | - Douglas S F Ling
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Program in Neural and Behaviroal Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA; Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| |
Collapse
|
37
|
Smith JA, Nguyen T, Davis BC, Lahiri DK, Hato T, Obukhov AG, White FA. Propranolol treatment during repetitive mild traumatic brain injuries induces transcriptomic changes in the bone marrow of mice. Front Neurosci 2023; 17:1219941. [PMID: 37817806 PMCID: PMC10561692 DOI: 10.3389/fnins.2023.1219941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/25/2023] [Indexed: 10/12/2023] Open
Abstract
Introduction There are 1.5 million new mild traumatic brain injuries (mTBI) annually in the US, with many of the injured experiencing long-term consequences lasting months after the injury. Although the post injury mechanisms are not well understood, current knowledge indicates peripheral immune system activation as a causal link between mTBI and long-term side effects. Through a variety of mechanisms, peripheral innate immune cells are recruited to the CNS after TBI to repair and heal the injured tissue; however, the recruitment and activation of these cells leads to further inflammation. Emerging evidence suggests sympathetic nervous system (SNS) activity plays a substantial role in the recruitment of immune cells post injury. Methods We sought to identify the peripheral innate immune response after repeated TBIs in addition to repurposing the nonselective beta blocker propranolol as a novel mTBI therapy to limit SNS activity and mTBI pathophysiology in the mouse. Mice underwent repetitive mTBI or sham injury followed by i.p. saline or propranolol. Isolated mRNA derived from femur bone marrow of mice was assayed for changes in gene expression at one day, one week, and four weeks using Nanostring nCounter® stem cell characterization panel. Results Differential gene expression analysis for bone marrow uncovered significant changes in many genes following drug alone, mTBI alone and drug combined with mTBI. Discussion Our data displays changes in mRNA at various timepoints, most pronounced in the mTBI propranolol group, suggesting a single dose propranolol injection as a viable future mTBI therapy in the acute setting.
Collapse
Affiliation(s)
- Jared A. Smith
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Tyler Nguyen
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brittany C. Davis
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Debomoy K. Lahiri
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Takashi Hato
- Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Alexander G. Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Fletcher A. White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
38
|
Bjorklund GR, Wong J, Brafman D, Bowser R, Stabenfeldt SE. Traumatic brain injury induces TDP-43 mislocalization and neurodegenerative effects in tissue distal to the primary injury site in a non-transgenic mouse. Acta Neuropathol Commun 2023; 11:137. [PMID: 37608352 PMCID: PMC10463884 DOI: 10.1186/s40478-023-01625-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/18/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury (TBI) initiates tissue and cellular damage to the brain that is immediately followed by secondary injury sequalae with delayed and continual damage. This secondary damage includes pathological processes that may contribute to chronic neurodegeneration and permanent functional and cognitive deficits. TBI is also associated with an increased risk of developing neurodegenerative diseases such as Alzheimer's disease (AD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS) as indicated by shared pathological features. For example, abnormalities in the TAR DNA-binding Protein 43 (TDP-43) that includes cytoplasmic mislocalization, cytosolic aggregation, and an increase in phosphorylation and ubiquitination are seen in up to 50% of FTD cases, up to 70% of AD cases, and is considered a hallmark pathology of ALS occurring in > 97% of cases. Yet the prevalence of TDP-43 pathology post-TBI has yet to be fully characterized. Here, we employed a non-transgenic murine controlled cortical injury model of TBI and observed injury-induced hallmark TDP-43 pathologies in brain and spinal cord tissue distal to the primary injury site and did not include the focally damaged tissue within the primary cortical injury site. Analysis revealed a temporal-dependent and significant increase in neuronal TDP-43 mislocalization in the cortical forebrain rostral to and distant from the primary injury site up to 180 days post injury (DPI). TDP-43 mislocalization was also detected in neurons located in the ventral horns of the cervical spinal cord following a TBI. Moreover, a cortical layer-dependent affect was identified, increasing from superficial to deeper cortical layers over time from 7 DPI up to 180 DPI. Lastly, RNAseq analysis confirmed an injury-induced misregulation of several key biological processes implicated in neurons that increased over time. Collectively, this study demonstrates a connection between a single moderate TBI event and chronic neurodegenerative processes that are not limited to the primary injury site and broadly distributed throughout the cortex and corticospinal tract.
Collapse
Affiliation(s)
- George R Bjorklund
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jennifer Wong
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - David Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Robert Bowser
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
39
|
Wong KR, Wright DK, Sgro M, Salberg S, Bain J, Li C, Sun M, McDonald SJ, Mychasiuk R, Brady RD, Shultz SR. Persistent Changes in Mechanical Nociception in Rats With Traumatic Brain Injury Involving Polytrauma. THE JOURNAL OF PAIN 2023; 24:1383-1395. [PMID: 36958460 DOI: 10.1016/j.jpain.2023.03.009] [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: 09/29/2022] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
Traumatic brain injury (TBI) survivors often experience debilitating consequences. Due to the high impact nature of TBI, patients often experience concomitant peripheral injuries (ie, polytrauma). A common, yet often overlooked, comorbidity of TBI is chronic pain. Therefore, this study investigated how common concomitant peripheral injuries (ie, femoral fracture and muscle crush) can affect long-term behavioral and structural TBI outcomes with a particular focus on nociception. Rats were randomly assigned to 1 of 4 groups: polytrauma (POLY; ie, fracture + muscle crush + TBI), peripheral injury (PERI; ie, fracture + muscle crush + sham TBI), TBI (ie, sham fracture + sham muscle crush + TBI), and sham-injured (SHAM; ie, sham fracture + sham muscle crush + sham TBI). Rats underwent behavioral testing at 3-, 6-, and 11-weeks postinjury, and were then euthanized for postmortem magnetic resonance imaging (MRI). POLY rats had a persisting increase in pain sensitivity compared to all groups on the von Frey test. MRI revealed that POLY rats also had abnormalities in the cortical and subcortical brain structures involved in nociceptive processing. These findings have important implications and provide a foundation for future studies to determine the underlying mechanisms and potential treatment strategies for chronic pain in TBI survivors. PERSPECTIVE: Rats with TBI and concomitant peripheral trauma displayed chronic nociceptive pain and MRI images also revealed damaged brain structures/pathways that are involved in chronic pain development. This study highlights the importance of polytrauma and the affected brain regions for developing chronic pain.
Collapse
Affiliation(s)
- Ker Rui Wong
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Marissa Sgro
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Jesse Bain
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Crystal Li
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Department of Nursing, Health and Human Services, Vancouver Island University, Nanaimo, BC, Canada.
| |
Collapse
|
40
|
Saletti PG, Mowrey WB, Liu W, Li Q, McCullough J, Aniceto R, Lin I, Eklund M, Casillas‐Espinosa PM, Ali I, Santana‐Gomez C, Coles L, Shultz SR, Jones N, Staba R, O'Brien TJ, Moshé SL, Agoston DV, Galanopoulou AS, for the EpiBioS4Rx Study Group. Early preclinical plasma protein biomarkers of brain trauma are influenced by early seizures and levetiracetam. Epilepsia Open 2023; 8:586-608. [PMID: 37026764 PMCID: PMC10235584 DOI: 10.1002/epi4.12738] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
OBJECTIVE We used the lateral fluid percussion injury (LFPI) model of moderate-to-severe traumatic brain injury (TBI) to identify early plasma biomarkers predicting injury, early post-traumatic seizures or neuromotor functional recovery (neuroscores), considering the effect of levetiracetam, which is commonly given after severe TBI. METHODS Adult male Sprague-Dawley rats underwent left parietal LFPI, received levetiracetam (200 mg/kg bolus, 200 mg/kg/day subcutaneously for 7 days [7d]) or vehicle post-LFPI, and were continuously video-EEG recorded (n = 14/group). Sham (craniotomy only, n = 6), and naïve controls (n = 10) were also used. Neuroscores and plasma collection were done at 2d or 7d post-LFPI or equivalent timepoints in sham/naïve. Plasma protein biomarker levels were determined by reverse phase protein microarray and classified according to injury severity (LFPI vs. sham/control), levetiracetam treatment, early seizures, and 2d-to-7d neuroscore recovery, using machine learning. RESULTS Low 2d plasma levels of Thr231 -phosphorylated tau protein (pTAU-Thr231 ) and S100B combined (ROC AUC = 0.7790) predicted prior craniotomy surgery (diagnostic biomarker). Levetiracetam-treated LFPI rats were differentiated from vehicle treated by the 2d-HMGB1, 2d-pTAU-Thr231 , and 2d-UCHL1 plasma levels combined (ROC AUC = 0.9394) (pharmacodynamic biomarker). Levetiracetam prevented the seizure effects on two biomarkers that predicted early seizures only among vehicle-treated LFPI rats: pTAU-Thr231 (ROC AUC = 1) and UCHL1 (ROC AUC = 0.8333) (prognostic biomarker of early seizures among vehicle-treated LFPI rats). Levetiracetam-resistant early seizures were predicted by high 2d-IFNγ plasma levels (ROC AUC = 0.8750) (response biomarker). 2d-to-7d neuroscore recovery was best predicted by higher 2d-S100B, lower 2d-HMGB1, and 2d-to-7d increase in HMGB1 or decrease in TNF (P < 0.05) (prognostic biomarkers). SIGNIFICANCE Antiseizure medications and early seizures need to be considered in the interpretation of early post-traumatic biomarkers.
Collapse
Affiliation(s)
- Patricia G. Saletti
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Wenzhu B. Mowrey
- Department of Epidemiology & Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Wei Liu
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Qianyun Li
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Jesse McCullough
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Roxanne Aniceto
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - I‐Hsuan Lin
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Michael Eklund
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Pablo M. Casillas‐Espinosa
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Idrish Ali
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | | | - Lisa Coles
- University of Minnesota Twin CitiesMinneapolisMinnesotaUSA
| | - Sandy R. Shultz
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Nigel Jones
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | | | - Terence J. O'Brien
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
- Isabelle Rapin Division of Child NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Dominick P Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Denes V. Agoston
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
- Isabelle Rapin Division of Child NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Dominick P Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
| | | |
Collapse
|
41
|
El-Demerdash N, Pan T, Choi O, Saraswati M, Koehler RC, Robertson CL, Savonenko A. Importance of Control Groups for Evaluating Long-Term Behavioral and Cognitive Outcomes of Controlled Cortical Impact in Immature Rats. J Neurotrauma 2023; 40:1197-1215. [PMID: 36416234 PMCID: PMC10259614 DOI: 10.1089/neu.2021.0376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Therapies are limited for pediatric traumatic brain injury (TBI), especially for the very young who can experience long-term consequences to learning, memory, and social behavior. Animal models of pediatric TBI have yielded mechanistic insights, but demonstration of clinically relevant long-term behavioral and/or cognitive deficits has been challenging. We characterized short- and long-term outcomes in a controlled cortical impact (CCI) model of pediatric TBI using a panel of tests between 2 weeks and ∼4 months after injury. Male rats with CCI at postnatal Day (PND) 10 were compared with three control groups: Naïve, Anesthesia, and Craniotomy. Motor testing (PND 25-33), novel object recognition (NOR; PND 40-50), and multiple tasks in water maze (WM; PND 65-100) were followed by social interaction tests (PND 120-140). Anesthesia rats performed the same as Naïve rats in all tasks. TBI rats, when compared with Naïve controls, had functional impairments across most tests studied. The most sensitive cognitive processes affected by TBI included those that required fast one-trial learning (NOR, WM), flexibility of acquired memory traces (reversals in WM), response strategies (WM), or recognition memory in the setting of reciprocal social interactions. Both TBI and Craniotomy groups demonstrated increased rates of decision making across several WM tasks, suggesting disinhibition of motor responses. When the TBI group was compared with the Craniotomy group, however, deficits were detected in a limited number of outcomes. The latter included learning speed (WM), cognitive flexibility (WM), and social recognition memory. Notably, effects of craniotomy, when compared with Naïve controls, spanned across multiple tasks, and in some tasks, could reach the effect sizes observed in TBI. These results highlight the importance of appropriate control groups in pediatric CCI models. In addition, the study demonstrates the high sensitivity of comprehensive cognitive testing to detect long-term effects of early-age craniotomy and TBI and provides a template for future testing of experimental therapies.
Collapse
Affiliation(s)
- Nagat El-Demerdash
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | - Tiffany Pan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | - Olivia Choi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | - Manda Saraswati
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | - Raymond C. Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | - Courtney L. Robertson
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | - Alena Savonenko
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Song S, Fallegger F, Trouillet A, Kim K, Lacour SP. Deployment of an electrocorticography system with a soft robotic actuator. Sci Robot 2023; 8:eadd1002. [PMID: 37163609 DOI: 10.1126/scirobotics.add1002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Electrocorticography (ECoG) is a minimally invasive approach frequently used clinically to map epileptogenic regions of the brain and facilitate lesion resection surgery and increasingly explored in brain-machine interface applications. Current devices display limitations that require trade-offs among cortical surface coverage, spatial electrode resolution, aesthetic, and risk consequences and often limit the use of the mapping technology to the operating room. In this work, we report on a scalable technique for the fabrication of large-area soft robotic electrode arrays and their deployment on the cortex through a square-centimeter burr hole using a pressure-driven actuation mechanism called eversion. The deployable system consists of up to six prefolded soft legs, and it is placed subdurally on the cortex using an aqueous pressurized solution and secured to the pedestal on the rim of the small craniotomy. Each leg contains soft, microfabricated electrodes and strain sensors for real-time deployment monitoring. In a proof-of-concept acute surgery, a soft robotic electrode array was successfully deployed on the cortex of a minipig to record sensory cortical activity. This soft robotic neurotechnology opens promising avenues for minimally invasive cortical surgery and applications related to neurological disorders such as motor and sensory deficits.
Collapse
Affiliation(s)
- Sukho Song
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
- Laboratory of Sustainability Robotics, Swiss Federal Laboratories for Materials Science and Technology (Empa), 8600 Dübendorf, Switzerland
| | - Florian Fallegger
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
| | - Alix Trouillet
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
| | - Kyungjin Kim
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Stéphanie P Lacour
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
| |
Collapse
|
43
|
Gee CC, Steffen R, Kievit FM. An updated Barnes maze protocol for assessing the outcome of controlled cortical impact mouse models of traumatic brain injury. J Neurosci Methods 2023; 392:109866. [PMID: 37116622 PMCID: PMC10205663 DOI: 10.1016/j.jneumeth.2023.109866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/06/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
BACKGROUND The Barnes Maze (BM) is a common method of testing cognitive deficits in rodents. Adapting BM protocols for specific neurological disorders could potentially aid in more effective testing, reduce research time, and help decrease variability between studies. NEW METHOD We tested differences an updated, shortened BM consisting of 6 days, 3 trials per day, only covering the equivalent of the spatial acquisition week BM protocol and a probe trial day consisting of one trial (7 total days). RESULTS Kaplan-Meier plots of escape percentage as a function of total latency showed a significant difference between control and CCI mice in the updated protocol on days 3 through 6. Additionally, probe trial data showed significant differences in primary latency, primary errors, and returns to goal. COMPARISON WITH EXISTING METHODS We tested differences between a traditional 5 days per week, 2 trials per day, spatial acquisition and reversal weeks BM protocol (12 total days with probe trials) with an updated 6-day BM protocol (7 total days with probe trial). In the probe trial, the updated protocol control mice showed an over 5-fold decrease in primary latency and primary errors and a 4.6-fold increase in returns to goal compared to the traditional protocol. Additionally, mice in both protocols performed similarly on a trial-by-trial basis suggesting that the changes made for the updated protocol increased learning and memory and was not simply an easier task. CONCLUSION The updated BM protocol showed an improved ability to distinguish between control and CCI mice and promoted improved and more consistent learning for both the control and CCI groups.
Collapse
Affiliation(s)
- Connor C Gee
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 262 Morrison Center, 4240 Fair St, Lincoln, NE 68583, USA
| | - Rylie Steffen
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 262 Morrison Center, 4240 Fair St, Lincoln, NE 68583, USA
| | - Forrest M Kievit
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 262 Morrison Center, 4240 Fair St, Lincoln, NE 68583, USA.
| |
Collapse
|
44
|
Babb JA, Zuberer A, Heinrichs S, Rumbika KK, Alfiler L, Lakis GA, Leite-Morris KA, Kaplan GB. Disturbances in fear extinction learning after mild traumatic brain injury in mice are accompanied by alterations in dendritic plasticity in the medial prefrontal cortex and basolateral nucleus of the amygdala. Brain Res Bull 2023; 198:15-26. [PMID: 37031792 DOI: 10.1016/j.brainresbull.2023.04.001] [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: 01/10/2023] [Revised: 03/25/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Mild traumatic brain injury (mTBI) and post-traumatic stress disorder (PTSD) have emerged as the signature injuries of the U.S. veterans who served in Iraq and Afghanistan, and frequently co-occur in both military and civilian and populations. To better understand how fear learning and underlying neural systems might be altered after mTBI we examined the acquisition of cued fear conditioning and its extinction along with brain morphology and dendritic plasticity in a mouse model of mTBI. To induce mTBI in adult male C57BL/6J mice, a lateral fluid percussive injury (LFP 1.7) was produced using a fluid pulse of 1.7 atmosphere force to the right parietal lobe. Behavior in LFP 1.7 mice was compared to behavior in mice from two separate control groups: mice subjected to craniotomy without LFP injury (Sham) and mice that did not undergo surgery (Unoperated). Following behavioral testing, neural endpoints (dendritic structural plasticity and neuronal volume) were assessed in the basolateral nucleus of the amygdala (BLA), which plays a critical sensory role in fear learning, and medial prefrontal cortex (mPFC), responsible for executive functions and inhibition of fear behaviors. No gross motor abnormalities or increased anxiety-like behaviors were observed in LFP or Sham mice after surgery compared to Unoperated mice. We found that all mice acquired fear behavior, assessed as conditioned freezing to auditory cue in a single session of 6 trials, and acquisition was similar across treatment groups. Using a linear mixed effects analysis, we showed that fear behavior decreased overall over 6 days of extinction training with no effect of treatment group across extinction days. However, a significant interaction was demonstrated between the treatment groups during within-session freezing behavior (5 trials per day) during extinction training. Specifically, freezing behavior increased across within-session extinction trials in LFP 1.7 mice, whereas freezing behavior in control groups did not change on extinction test days, reflecting a dissociation between within-trial and between-trial fear extinction. Additionally, LFP mice demonstrated bilateral increases in dendritic spine density in the BLA and decreases in dendritic complexity in the PFC. The translational implications are that individuals with TBI undergoing fear extinction therapy may demonstrate within-session aberrant learning that could be targeted for more effective treatment interventions.
Collapse
Affiliation(s)
- Jessica A Babb
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Mental Health Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Department of Psychiatry, Harvard Medical School, Boston, MA, 02115 USA.
| | - Agnieszka Zuberer
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; Department of Psychiatry and Psychotherapy, Jena University Hospital, 07743 Jena, Germany.
| | - Stephen Heinrichs
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA.
| | - Kendra K Rumbika
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA.
| | - Lauren Alfiler
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA.
| | - Gabrielle A Lakis
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02218 USA.
| | - Kimberly A Leite-Morris
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118 USA.
| | - Gary B Kaplan
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Mental Health Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118 USA; Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118 USA.
| |
Collapse
|
45
|
Yadikar H, Johnson C, Pafundi N, Nguyen L, Kurup M, Torres I, Al-Enezy A, Yang Z, Yost R, Kobeissy FH, Wang KKW. Neurobiochemical, Peptidomic, and Bioinformatic Approaches to Characterize Tauopathy Peptidome Biomarker Candidates in Experimental Mouse Model of Traumatic Brain Injury. Mol Neurobiol 2023; 60:2295-2319. [PMID: 36635478 DOI: 10.1007/s12035-022-03165-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 12/10/2022] [Indexed: 01/14/2023]
Abstract
Traumatic brain injury (TBI) is a multidimensional damage, and currently, no FDA-approved medicine is available. Multiple pathways in the cell are triggered through a head injury (e.g., calpain and caspase activation), which truncate tau and generate variable fragment sizes (MW 400-45,000 K). In this study, we used an open-head TBI mouse model generated by controlled cortical impact (CCI) and collected ipsilateral (IC) and contralateral (CC) mice htau brain cortices at one (D1) three (D3), and seven (D7) days post-injury. We implemented immunological (antibody-based detection) and peptidomic approaches (nano-reversed-phase liquid chromatography/tandem mass spectrometry) to investigate proteolytic tau peptidome (low molecular weight (LMW) < 10 K)) and pathological phosphorylation sites (high-molecular-weight (HMW); > 10 K) derived from CCI-TBI animal models. Our immunoblotting analysis verified tau hyperphosphorylation, HMW, and HMW breakdown products (HMW-BDP) formation of tau (e.g., pSer202, pThr181, pThr231, pSer396, and pSer404), following CCI-TBI. Peptidomic data revealed unique sequences of injury-dependent proteolytic peptides generated from human tau protein. Among the N-terminal tau peptides, EIPEGTTAEEAGIGDTPSLEDEAAGHVTQA (a.a. 96-125) and AQPHTEIPEGTTAEEAGIGDTPSLEDEAAGHVTQARM (a.a. 91-127). Examples of tau C-terminal peptides identified include NVSSTGSIDMVDSPQLATLADEVSASLAKQGL (a.a. 410-441) and QLATLADEVSASLAKQGL (a.a. 424-441). Our peptidomic bioinformatic tools showed the association of proteases, such as CAPN1, CAPN2, and CTSL; CASP1, MMP7, and MMP9; and ELANE, GZMA, and MEP1A, in CCI-TBI tau peptidome. In clinical trials for novel TBI treatments, it might be useful to monitor a subset of tau peptidome as targets for biomarker utility and use them for a "theranostic" approach.
Collapse
Affiliation(s)
- Hamad Yadikar
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait.
| | - Connor Johnson
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Niko Pafundi
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Lynn Nguyen
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Milin Kurup
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Isabel Torres
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Albandery Al-Enezy
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Zhihui Yang
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Richard Yost
- Department of Chemistry, Chemistry Laboratory Building, University of Florida, Gainesville, FL, 32611, USA
| | - Firas H Kobeissy
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA. .,Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon. .,Morehouse School of Medicine, Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), 720 Westview Dr. SW, Atlanta, GA, 30310, USA.
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA. .,Morehouse School of Medicine, Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), 720 Westview Dr. SW, Atlanta, GA, 30310, USA. .,Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, Gainesville, FL, 32608, USA.
| |
Collapse
|
46
|
Heiskanen M, Das Gupta S, Mills JD, van Vliet EA, Manninen E, Ciszek R, Andrade P, Puhakka N, Aronica E, Pitkänen A. Discovery and Validation of Circulating microRNAs as Biomarkers for Epileptogenesis after Experimental Traumatic Brain Injury-The EPITARGET Cohort. Int J Mol Sci 2023; 24:ijms24032823. [PMID: 36769143 PMCID: PMC9918096 DOI: 10.3390/ijms24032823] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Traumatic brain injury (TBI) causes 10-20% of structural epilepsies and 5% of all epilepsies. The lack of prognostic biomarkers for post-traumatic epilepsy (PTE) is a major obstacle to the development of anti-epileptogenic treatments. Previous studies revealed TBI-induced alterations in blood microRNA (miRNA) levels, and patients with epilepsy exhibit dysregulation of blood miRNAs. We hypothesized that acutely altered plasma miRNAs could serve as prognostic biomarkers for brain damage severity and the development of PTE. To investigate this, epileptogenesis was induced in adult male Sprague Dawley rats by lateral fluid-percussion-induced TBI. Epilepsy was defined as the occurrence of at least one unprovoked seizure during continuous 1-month video-electroencephalography monitoring in the sixth post-TBI month. Cortical pathology was analyzed by magnetic resonance imaging on day 2 (D2), D7, and D21, and by histology 6 months post-TBI. Small RNA sequencing was performed from tail-vein plasma samples on D2 and D9 after TBI (n = 16, 7 with and 9 without epilepsy) or sham operation (n = 4). The most promising miRNA biomarker candidates were validated by droplet digital polymerase chain reaction in a validation cohort of 115 rats (8 naïve, 17 sham, and 90 TBI rats [21 with epilepsy]). These included 7 brain-enriched plasma miRNAs (miR-434-3p, miR-9a-3p, miR-136-3p, miR-323-3p, miR-124-3p, miR-212-3p, and miR-132-3p) that were upregulated on D2 post-TBI (p < 0.001 for all compared with naïve rats). The acute post-TBI plasma miRNA profile did not predict the subsequent development of PTE or PTE severity. Plasma miRNA levels, however, predicted the cortical pathology severity on D2 (Spearman ρ = 0.345-0.582, p < 0.001), D9 (ρ = 0.287-0.522, p < 0.001-0.01), D21 (ρ = 0.269-0.581, p < 0.001-0.05) and at 6 months post-TBI (ρ = 0.230-0.433, p < 0.001-0.05). We found that the levels of 6 of 7 miRNAs also reflected mild brain injury caused by the craniotomy during sham operation (ROC AUC 0.76-0.96, p < 0.001-0.05). In conclusion, our findings revealed that increased levels of neuronally enriched miRNAs in the blood circulation after TBI reflect the extent of cortical injury in the brain but do not predict PTE development.
Collapse
Affiliation(s)
- Mette Heiskanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Shalini Das Gupta
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - James D. Mills
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Chalfont Centre for Epilepsy, Buckinghamshire SL9 0RJ, UK
| | - Erwin A. van Vliet
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Eppu Manninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Robert Ciszek
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Pedro Andrade
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Noora Puhakka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland, 2103 SW Heemstede, The Netherlands
| | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Correspondence:
| |
Collapse
|
47
|
Jeon S, Baik J, Kim J, Lee J, Do W, Kim E, Lee HJ, Kim H. Intrathecal dexmedetomidine attenuates mechanical allodynia through the downregulation of brain-derived neurotrophic factor in a mild traumatic brain injury rat model. Korean J Anesthesiol 2023; 76:56-66. [PMID: 35760392 PMCID: PMC9902181 DOI: 10.4097/kja.22209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND This study evaluated the effects of dexmedetomidine and propofol on brain-derived neurotrophic factor level in the cerebrospinal fluid (c-BDNF) and mechanical allodynia in a mild traumatic brain injury (TBI) rat model. METHODS After fixing the rat's skull on a stereotactic frame under general anesthesia, craniotomy was performed. After impact, 10 µl of drug was injected into the cisterna magna (group S: sham, group D: dexmedetomidine 5 μg/kg, group P: propofol 500 μg/kg, and group T: untreated TBI). The 50% mechanical withdrawal threshold (50% MWT) and c-BDNF level were measured on postoperative days (PODs) 1, 7, and 14. RESULTS The 50% MWT measured on PODs 1, 7, and 14 was lower and the c-BDNF level on POD 1 was higher in group T than in group S. In group D, the c-BDNF level on POD 1 was lower than that in group T and was comparable with that in group S during the whole study period. The 50% MWT of group D was higher than that of group T throughout the postoperative period. In group P, there were no significant differences in the 50% MWT during the entire postoperative period compared with group T; the c-BDNF level was higher than that in group T on POD 1. CONCLUSIONS Intrathecal administration of dexmedetomidine may attenuate TBI-induced mechanical allodynia for up to two weeks post-injury through immediate suppression of c-BDNF in mild TBI rats. The inhibition of c-BDNF expression in the acute phase reduced the occurrence of TBI-induced chronic neuropathic pain.
Collapse
Affiliation(s)
- Soeun Jeon
- Department of Anesthesiology and Pain Medicine, School of Dentistry, Institute for Translational Research in Dentistry, Kyungpook National University, Daegu, Korea
| | - Jiseok Baik
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Pusan National University School of Medicine, Busan, Korea
| | - Jisu Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Jiyoon Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Wangseok Do
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Eunsoo Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Pusan National University School of Medicine, Busan, Korea
| | - Hyeon Jeong Lee
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Pusan National University School of Medicine, Busan, Korea
| | - Haekyu Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Pusan National University School of Medicine, Busan, Korea
| |
Collapse
|
48
|
Jovanović N, Suchánková Š, Kang M, Melichar A, Bureš Z, Tureček R. Altered hearing function in mice with implanted cranial windows. Neurosci Lett 2023; 792:136969. [PMID: 36402256 DOI: 10.1016/j.neulet.2022.136969] [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: 06/01/2022] [Revised: 11/02/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
The cranial window technique has proven to be an effective method for in vivo imaging of cortical activity. However, given the invasive nature of this procedure, possible side effects could be expected in the nervous system. In this study, we evaluated the effects of unilateral cranial window surgery on auditory function in C57BL6 mice using electrophysiological and behavioral approaches. We found that one week after implantation, mice exhibited both increased thresholds and decreased amplitudes of their auditory brainstem responses. These changes were accompanied by a decrease in distortion product otoacoustic emissions, indicating a deterioration in cochlear function. In addition, behavioral testing of these mice revealed reduced suppression of their acoustic startle response by gap prepulse, suggesting a deficit in auditory processing or possibly the presence of tinnitus. The changes in auditory function appeared to be only partially reversible within four weeks after surgery. Thus, our findings suggest that cranial window implantation causes long-term functional changes in the auditory system that should be considered when interpreting data from optical imaging techniques.
Collapse
Affiliation(s)
- Nataša Jovanović
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220 Prague 4 - Krč, Czech Republic; Second Faculty of Medicine, Charles University, 150 06 Prague, Czech Republic
| | - Štěpánka Suchánková
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220 Prague 4 - Krč, Czech Republic
| | - Minseok Kang
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220 Prague 4 - Krč, Czech Republic
| | - Adolf Melichar
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220 Prague 4 - Krč, Czech Republic; Second Faculty of Medicine, Charles University, 150 06 Prague, Czech Republic
| | - Zbyněk Bureš
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220 Prague 4 - Krč, Czech Republic; Department of Cognitive Systems and Neurosciences, Czech Institute of Informatics, Robotics and Cybernetics, Czech Technical University, Jugoslávských, partyzánů 1580/3, 160 00 Prague 6, Czech Republic
| | - Rostislav Tureček
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220 Prague 4 - Krč, Czech Republic.
| |
Collapse
|
49
|
Heiskanen M, Jääskeläinen O, Manninen E, Das Gupta S, Andrade P, Ciszek R, Gröhn O, Herukka SK, Puhakka N, Pitkänen A. Plasma Neurofilament Light Chain (NF-L) Is a Prognostic Biomarker for Cortical Damage Evolution but Not for Cognitive Impairment or Epileptogenesis Following Experimental TBI. Int J Mol Sci 2022; 23:ijms232315208. [PMID: 36499527 PMCID: PMC9736117 DOI: 10.3390/ijms232315208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/08/2022] Open
Abstract
Plasma neurofilament light chain (NF-L) levels were assessed as a diagnostic biomarker for traumatic brain injury (TBI) and as a prognostic biomarker for somatomotor recovery, cognitive decline, and epileptogenesis. Rats with severe TBI induced by lateral fluid-percussion injury (n = 26, 13 with and 13 without epilepsy) or sham-operation (n = 8) were studied. During a 6-month follow-up, rats underwent magnetic resonance imaging (MRI) (day (D) 2, D7, and D21), composite neuroscore (D2, D6, and D14), Morris-water maze (D35−D39), and a 1-month-long video-electroencephalogram to detect unprovoked seizures during the 6th month. Plasma NF-L levels were assessed using a single-molecule assay at baseline (i.e., naïve animals) and on D2, D9, and D178 after TBI or a sham operation. Plasma NF-L levels were 483-fold higher on D2 (5072.0 ± 2007.0 pg/mL), 89-fold higher on D9 (930.3 ± 306.4 pg/mL), and 3-fold higher on D176 32.2 ± 8.9 pg/mL after TBI compared with baseline (10.5 ± 2.6 pg/mL; all p < 0.001). Plasma NF-L levels distinguished TBI rats from naïve animals at all time-points examined (area under the curve [AUC] 1.0, p < 0.001), and from sham-operated controls on D2 (AUC 1.0, p < 0.001). Plasma NF-L increases on D2 were associated with somatomotor impairment severity (ρ = −0.480, p < 0.05) and the cortical lesion extent in MRI (ρ = 0.401, p < 0.05). Plasma NF-L increases on D2 or D9 were associated with the cortical lesion extent in histologic sections at 6 months post-injury (ρ = 0.437 for D2; ρ = 0.393 for D9, p < 0.05). Plasma NF-L levels, however, did not predict somatomotor recovery, cognitive decline, or epileptogenesis (p > 0.05). Plasma NF-L levels represent a promising noninvasive translational diagnostic biomarker for acute TBI and a prognostic biomarker for post-injury somatomotor impairment and long-term structural brain damage.
Collapse
Affiliation(s)
- Mette Heiskanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Olli Jääskeläinen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Eppu Manninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Shalini Das Gupta
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Pedro Andrade
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Robert Ciszek
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Olli Gröhn
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
- Department of Neurology, Kuopio University Hospital, P.O. Box 1777, 70211 Kuopio, Finland
| | - Noora Puhakka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
- Correspondence:
| |
Collapse
|
50
|
Boshra R, Eradath M, Dougherty K, Wu B, Morea BM, Harris M, Pinsk MA, Kastner S. Case studies in neuroscience: reversible signatures of edema following electric and piezoelectric craniotomy drilling in macaques. J Neurophysiol 2022; 128:919-926. [PMID: 36043799 PMCID: PMC9550573 DOI: 10.1152/jn.00108.2022] [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: 03/21/2022] [Revised: 07/28/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
In vivo electrophysiology requires direct access to brain tissue, necessitating the development and refinement of surgical procedures and techniques that promote the health and well-being of animal subjects. Here, we report a series of findings noted on structural magnetic resonance imaging (MRI) scans in monkeys with MRI-compatible implants following small craniotomies that provide access for intracranial electrophysiology. We found distinct brain regions exhibiting hyperintensities in T2-weighted scans that were prominent underneath the sites at which craniotomies had been performed. We interpreted these hyperintensities as edema of the neural tissue and found that they were predominantly present following electric and piezoelectric drilling, but not when manual, hand-operated drills were used. Furthermore, the anomalies subsided within 2-3 wk following surgery. Our report highlights the utility of MRI-compatible implants that promote clinical examination of the animal's brain, sometimes revealing findings that may go unnoticed when incompatible implants are used. We show replicable differences in outcome when using electric versus mechanical devices, both ubiquitous in the field. If electric drills are used, our report cautions against electrophysiological recordings from tissue directly underneath the craniotomy for the first 2-3 wk following the procedure due to putative edema.NEW & NOTEWORTHY Close examination of structural MRI in eight nonhuman primates following craniotomy surgeries for intracranial electrophysiology highlights a prevalence of hyperintensities on T2-weighted scans following surgeries conducted using electric and piezoelectric drills, but not when using mechanical, hand-operated drills. We interpret these anomalies as edema of neural tissue that resolved 2-3 wk postsurgery. This finding is especially of interest as electrophysiological recordings from compromised tissue may directly influence the integrity of collected data immediately following surgery.
Collapse
Affiliation(s)
- Rober Boshra
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Manoj Eradath
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Kacie Dougherty
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Bichan Wu
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Britney M Morea
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Michael Harris
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Mark A Pinsk
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Sabine Kastner
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
- Department of Psychology, Princeton University, Princeton, New Jersey
| |
Collapse
|