1
|
Rigo YR, Benvenutti R, Portela LV, Strogulski NR. Neurogenic potential of NG2 in neurotrauma: a systematic review. Neural Regen Res 2024; 19:2673-2683. [PMID: 38595286 PMCID: PMC11168526 DOI: 10.4103/nrr.nrr-d-23-01031] [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: 06/21/2023] [Revised: 12/20/2023] [Accepted: 02/07/2024] [Indexed: 04/11/2024] Open
Abstract
Regenerative approaches towards neuronal loss following traumatic brain or spinal cord injury have long been considered a dogma in neuroscience and remain a cutting-edge area of research. This is reflected in a large disparity between the number of studies investigating primary and secondary injury as therapeutic targets in spinal cord and traumatic brain injuries. Significant advances in biotechnology may have the potential to reshape the current state-of-the-art and bring focus to primary injury neurotrauma research. Recent studies using neural-glial factor/antigen 2 (NG2) cells indicate that they may differentiate into neurons even in the developed brain. As these cells show great potential to play a regenerative role, studies have been conducted to test various manipulations in neurotrauma models aimed at eliciting a neurogenic response from them. In the present study, we systematically reviewed the experimental protocols and findings described in the scientific literature, which were peer-reviewed original research articles (1) describing preclinical experimental studies, (2) investigating NG2 cells, (3) associated with neurogenesis and neurotrauma, and (4) in vitro and/or in vivo, available in PubMed/MEDLINE, Web of Science or SCOPUS, from 1998 to 2022. Here, we have reviewed a total of 1504 papers, and summarized findings that ultimately suggest that NG2 cells possess an inducible neurogenic potential in animal models and in vitro. We also discriminate findings of NG2 neurogenesis promoted by different pharmacological and genetic approaches over functional and biochemical outcomes of traumatic brain injury and spinal cord injury models, and provide mounting evidence for the potential benefits of manipulated NG2 cell ex vivo transplantation in primary injury treatment. These findings indicate the feasibility of NG2 cell neurogenesis strategies and add new players in the development of therapeutic alternatives for neurotrauma.
Collapse
Affiliation(s)
- Yuri R. Rigo
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Radharani Benvenutti
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Luis V. Portela
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Zima L, Moore AN, Smolen P, Kobori N, Noble B, Robinson D, Hood KN, Homma R, Al Mamun A, Redell JB, Dash PK. The evolving pathophysiology of TBI and the advantages of temporally-guided combination therapies. Neurochem Int 2024; 180:105874. [PMID: 39366429 DOI: 10.1016/j.neuint.2024.105874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Several clinical and experimental studies have demonstrated that traumatic brain injury (TBI) activates cascades of biochemical, molecular, structural, and pathological changes in the brain. These changes combine to contribute to the various outcomes observed after TBI. Given the breadth and complexity of changes, combination treatments may be an effective approach for targeting multiple detrimental pathways to yield meaningful improvements. In order to identify targets for therapy development, the temporally evolving pathophysiology of TBI needs to be elucidated in detail at both the cellular and molecular levels, as it has been shown that the mechanisms contributing to cognitive dysfunction change over time. Thus, a combination of individual mechanism-based therapies is likely to be effective when maintained based on the time courses of the cellular and molecular changes being targeted. In this review, we will discuss the temporal changes of some of the key clinical pathologies of human TBI, the underlying cellular and molecular mechanisms, and the results from preclinical and clinical studies aimed at mitigating their consequences. As most of the pathological events that occur after TBI are likely to have subsided in the chronic stage of the disease, combination treatments aimed at attenuating chronic conditions such as cognitive dysfunction may not require the initiation of individual treatments at a specific time. We propose that a combination of acute, subacute, and chronic interventions may be necessary to maximally improve health-related quality of life (HRQoL) for persons who have sustained a TBI.
Collapse
Affiliation(s)
- Laura Zima
- Departments of Neurosurgery, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Anthony N Moore
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Paul Smolen
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Nobuhide Kobori
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Brian Noble
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Dustin Robinson
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Kimberly N Hood
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Ryota Homma
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Amar Al Mamun
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - John B Redell
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Pramod K Dash
- Departments of Neurosurgery, The University of Texas McGovern Medical School, Houston, TX, USA; Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
3
|
Niedowicz DM, Wang WX, Prajapati P, Zhong Y, Fister S, Rogers CB, Sompol P, Powell DK, Patel I, Norris CM, Saatman KE, Nelson PT. Nicorandil treatment improves survival and spatial learning in aged granulin knockout mice. Brain Pathol 2024:e13312. [PMID: 39438022 DOI: 10.1111/bpa.13312] [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: 05/29/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Mutations in the human granulin (GRN) gene are associated with multiple diseases, including dementia disorders such as frontotemporal dementia (FTD) and limbic-predominant age-related TDP-43 encephalopathy (LATE). We studied a Grn knockout (Grn-KO) mouse model in order to evaluate a potential therapeutic strategy for these diseases using nicorandil, a commercially available agonist for the ABCC9/Abcc9-encoded regulatory subunit of the "K+ATP" channel that is well-tolerated in humans. Aged (13 months) Grn-KO and wild-type (WT) mice were treated as controls or with nicorandil (15 mg/kg/day) in drinking water for 7 months, then tested for neurobehavioral performance, neuropathology, and gene expression. Mortality was significantly higher for aged Grn-KO mice (particularly females), but there was a conspicuous improvement in survival for both sexes treated with nicorandil. Grn-KO mice performed worse on some cognitive tests than WT mice, but Morris Water Maze performance was improved with nicorandil treatment. Neuropathologically, Grn-KO mice had significantly increased levels of glial fibrillary acidic protein (GFAP)-immunoreactive astrocytosis but not ionized calcium binding adaptor molecule 1 (IBA-1)-immunoreactive microgliosis, indicating cell-specific inflammation in the brain. Expression of several astrocyte-enriched genes, including Gfap, were also elevated in the Grn-KO brain. Nicorandil treatment was associated with a subtle shift in a subset of detected brain transcript levels, mostly related to attenuated inflammatory markers. Nicorandil treatment improved survival outcomes, cognition, and inflammation in aged Grn-KO mice.
Collapse
Affiliation(s)
- Dana M Niedowicz
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Wang-Xia Wang
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| | - Paresh Prajapati
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Yu Zhong
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Shuling Fister
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Colin B Rogers
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Pradoldej Sompol
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - David K Powell
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Indumati Patel
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Christopher M Norris
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Kathryn E Saatman
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Peter T Nelson
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
4
|
Rodolphi MS, Strogulski NR, Kopczynski A, Sartor M, Soares G, de Oliveira VG, Vinade L, Dal-Belo C, Portela JV, Geller CA, De Bastiani MA, Justus JS, Portela LOC, Smith DH, Portela LV. Nandrolone Abuse Prior to Head Trauma Mitigates Endoplasmic Reticulum Stress, Mitochondrial Bioenergetic Deficits, and Markers of Neurodegeneration. Mol Neurobiol 2024:10.1007/s12035-024-04488-8. [PMID: 39313656 DOI: 10.1007/s12035-024-04488-8] [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: 03/07/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
The abuse of synthetic steroids, such as nandrolone decanoate (ND), is often associated with violent behavior, increasing the risk of traumatic brain injury (TBI). After a TBI, proteins like APP, β-amyloid peptide-42 (Aβ42), and phosphorylated tau (pTau) accumulate and trigger endoplasmic reticulum (ER) stress associated with an unfolded protein response (UPR). The involvement of mitochondrial bioenergetics in this context remains unexplored. We interrogate whether the abuse of ND before TBI alters the responses of ER stress and mitochondrial bioenergetics in connection with neurodegeneration and memory processing in mice. Male CF1 adult mice were administered ND (15 mg/kg) or vehicle (VEH) s.c. for 19 days, coinciding with the peak day of aggressive behavior, and then underwent cortical controlled impact (CCI) or sham surgery. Spatial memory was assessed through the Morris water maze task (MWM) post-TBI. In synaptosome preparations, i) we challenged mitochondrial complexes (I, II, and V) in a respirometry assay, employing metabolic substrates, an uncoupler, and inhibitors; and ii) assessed molecular biomarkers through Western blot. TBI significantly increased APP, Aβ42, and pTauSer396 levels, along with ER-stress proteins, GRP78, ATF6, and CHOP, implying it primed apoptotic signaling. Concurrently, TBI reduced mitochondrial Ca2+ efflux in exchange with Na+, disturbed the formation/dissipation of membrane potential, increased H2O2 production, decreased biogenesis (PGC-1⍺ and TOM20), and ATP biosynthesis coupled with oxygen consumption. Unexpectedly, ND abuse before TBI attenuated the elevations in APP, Aβ42, and pTauSer396, accompanied by a decrease in GRP78, ATF6, and CHOP levels, and partial normalization of mitochondrial-related endpoints. A principal component analysis revealed a key hierarchical signature featuring mitochondrial Ca2+ efflux, CHOP, GRP78, TOM20, H2O2, and bioenergetic efficiency as a unique variable (PC1) able to explain the memory deficits caused by TBI, as well as the preservation of memory fitness induced by prior ND abuse.
Collapse
Affiliation(s)
- Marcelo S Rodolphi
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Nathan R Strogulski
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Leinster, Ireland
| | - Afonso Kopczynski
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Monia Sartor
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Gabriela Soares
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Vitoria G de Oliveira
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Lucia Vinade
- Laboratory of Neurobiology and Toxinology (LANETOX), Universidade Federal Do Pampa (UNIPAMPA), São Gabriel, RS, Brazil
| | - Chariston Dal-Belo
- Laboratory of Neurobiology and Toxinology (LANETOX), Universidade Federal Do Pampa (UNIPAMPA), São Gabriel, RS, Brazil
- Departamento Multidisciplinar - Escola Paulista de Política, Economia E Negócios (EPPEN), Universidade Federal de São Paulo (UNIFESP), Osasco, SP, Brazil
| | - Juliana V Portela
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Cesar A Geller
- Laboratory of Performance in Simulated Environment (LAPAS), Centro de Educação Física, Universidade Federal de Santa Maria - UFSM, Santa Maria, RS, Brazil
| | - Marco A De Bastiani
- Zimmer Neuroimaging Lab, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Jijo S Justus
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Luiz Osorio C Portela
- Laboratory of Performance in Simulated Environment (LAPAS), Centro de Educação Física, Universidade Federal de Santa Maria - UFSM, Santa Maria, RS, Brazil
| | - Douglas H Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Luis V Portela
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Universidade Federal Do Rio Grande Do Sul, UFRGS, Anexo, Rua Ramiro Barcelos 2600, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
5
|
Saccaro LF, Tassone M, Tozzi F, Rutigliano G. Proton magnetic resonance spectroscopy of N-acetyl aspartate in first depressive episode and chronic major depressive disorder: A systematic review and meta-analysis. J Affect Disord 2024; 355:265-282. [PMID: 38554884 DOI: 10.1016/j.jad.2024.03.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
N-acetyl aspartate (NAA) is a marker of neuronal integrity and metabolism. Deficiency in neuronal plasticity and hypometabolism are implicated in Major Depressive Disorder (MDD) pathophysiology. To test if cerebral NAA concentrations decrease progressively over the MDD course, we conducted a pre-registered meta-analysis of Proton Magnetic Resonance Spectroscopy (1H-MRS) studies comparing NAA concentrations in chronic MDD (n = 1308) and first episode of depression (n = 242) patients to healthy controls (HC, n = 1242). Sixty-two studies were meta-analyzed using a random-effect model for each brain region. NAA concentrations were significantly reduced in chronic MDD compared to HC within the frontal lobe (Hedges' g = -0.330; p = 0.018), the occipital lobe (Hedges' g = -0.677; p = 0.007), thalamus (Hedges' g = -0.673; p = 0.016), and frontal (Hedges' g = -0.471; p = 0.034) and periventricular white matter (Hedges' g = -0.478; p = 0.047). We highlighted a gap of knowledge regarding NAA levels in first episode of depression patients. Sensitivity analyses indicated that antidepressant treatment may reverse NAA alterations in the frontal lobe. We highlighted field strength and correction for voxel grey matter as moderators of NAA levels detection. Future studies should assess NAA alterations in the early stages of the illness and their longitudinal progression.
Collapse
Affiliation(s)
- Luigi F Saccaro
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Campus Biotech, 9 Chemin des Mines, 1202 Geneva, Switzerland; Department of Psychiatry, Geneva University Hospital, 1205 Geneva, Switzerland.
| | - Matteo Tassone
- Department of Pathology, University of Pisa, via Savi 10, 56126 Pisa, Italy
| | - Francesca Tozzi
- Bio@SNS laboratory, Scuola Normale Superiore, 56124 Pisa, Italy
| | - Grazia Rutigliano
- Department of Pathology, University of Pisa, via Savi 10, 56126 Pisa, Italy; Institute of Clinical Sciences, Imperial College London, MRI Steiner Unit, Hammersmith Hospital Campus, Du Cane Road, W12 0NN London, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
6
|
Vekaria HJ, Kalimon OJ, Prajapati P, Velmurugan GV, Sullivan PG. An efficient and high-throughput method for the evaluation of mitochondrial dysfunction in frozen brain samples after traumatic brain injury. Front Mol Biosci 2024; 11:1378536. [PMID: 38983247 PMCID: PMC11232470 DOI: 10.3389/fmolb.2024.1378536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/22/2024] [Indexed: 07/11/2024] Open
Abstract
Mitochondrial function analysis is a well-established method used in preclinical and clinical investigations to assess pathophysiological changes in various disease states, including traumatic brain injury (TBI). Although there are multiple approaches to assess mitochondrial function, one common method involves respirometric assays utilizing either Clark-type oxygen electrodes or fluorescent-based Seahorse analysis (Agilent). However, these functional analysis methods are typically limited to the availability of freshly isolated tissue samples due to the compromise of the electron transport chain (ETC) upon storage, caused by freeze-thaw-mediated breakdown of mitochondrial membranes. In this study, we propose and refine a method for evaluating electron flux through the ETC, encompassing complexes I, II, and IV, in frozen homogenates or mitochondrial samples within a single well of a Seahorse plate. Initially, we demonstrate the impact of TBI on freshly isolated mitochondria using the conventional oxidative phosphorylation protocol (OxPP), followed by a comparison with ETC analysis conducted on frozen tissue samples within the context of a controlled cortical impact (CCI) model of TBI. Additionally, we explore the effects of mitochondrial isolation from fresh versus snap-frozen brain tissues and their storage at -80°C, assessing its impact on electron transport chain protocol (ETCP) activity. Our findings indicate that while both sets of samples were frozen at a single time point, mitochondria from snap-frozen tissues exhibited reduced injury effects compared to preparations from fresh tissues, which were either homogenized or isolated into mitochondria and subsequently frozen for later use. Thus, we demonstrate that the preparation of homogenates or isolated mitochondria can serve as an appropriate method for storing brain samples, allowing for later analysis of mitochondrial function, following TBI using ETCP.
Collapse
Affiliation(s)
- Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
- Lexington VA Medical Center, United States Department of Veterans Affairs, Lexington, KY, United States
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Olivia J. Kalimon
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
- Lexington VA Medical Center, United States Department of Veterans Affairs, Lexington, KY, United States
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Paresh Prajapati
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Gopal V. Velmurugan
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
- Lexington VA Medical Center, United States Department of Veterans Affairs, Lexington, KY, United States
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
7
|
Kilgore MO, Hubbard WB. Effects of Low-Level Blast on Neurovascular Health and Cerebral Blood Flow: Current Findings and Future Opportunities in Neuroimaging. Int J Mol Sci 2024; 25:642. [PMID: 38203813 PMCID: PMC10779081 DOI: 10.3390/ijms25010642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Low-level blast (LLB) exposure can lead to alterations in neurological health, cerebral vasculature, and cerebral blood flow (CBF). The development of cognitive issues and behavioral abnormalities after LLB, or subconcussive blast exposure, is insidious due to the lack of acute symptoms. One major hallmark of LLB exposure is the initiation of neurovascular damage followed by the development of neurovascular dysfunction. Preclinical studies of LLB exposure demonstrate impairment to cerebral vasculature and the blood-brain barrier (BBB) at both early and long-term stages following LLB. Neuroimaging techniques, such as arterial spin labeling (ASL) using magnetic resonance imaging (MRI), have been utilized in clinical investigations to understand brain perfusion and CBF changes in response to cumulative LLB exposure. In this review, we summarize neuroimaging techniques that can further our understanding of the underlying mechanisms of blast-related neurotrauma, specifically after LLB. Neuroimaging related to cerebrovascular function can contribute to improved diagnostic and therapeutic strategies for LLB. As these same imaging modalities can capture the effects of LLB exposure in animal models, neuroimaging can serve as a gap-bridging diagnostic tool that permits a more extensive exploration of potential relationships between blast-induced changes in CBF and neurovascular health. Future research directions are suggested, including investigating chronic LLB effects on cerebral perfusion, exploring mechanisms of dysautoregulation after LLB, and measuring cerebrovascular reactivity (CVR) in preclinical LLB models.
Collapse
Affiliation(s)
- Madison O. Kilgore
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA;
| | - W. Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA;
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY 40502, USA
| |
Collapse
|
8
|
Harris JL, Wang X, Christian SK, Novikova L, Kalani A, Hui D, Ferren S, Barbay S, Ortiz JP, Nudo RJ, Brooks WM, Wilkins HM, Chalise P, Michaelis ML, Michaelis EK, Swerdlow RH. Traumatic Brain Injury Alters the Trajectory of Age-Related Mitochondrial Change. J Alzheimers Dis 2024; 97:1793-1806. [PMID: 38306050 DOI: 10.3233/jad-231237] [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: 02/03/2024]
Abstract
Background Some epidemiologic studies associate traumatic brain injury (TBI) with Alzheimer's disease (AD). Objective To test whether a TBI-induced acceleration of age-related mitochondrial change could potentially mediate the reported TBI-AD association. Methods We administered unilateral controlled cortical impact (CCI) or sham injuries to 5-month-old C57BL/6J and tau transgenic rTg4510 mice. In the non-transgenics, we assessed behavior (1-5 days, 1 month, and 15 months), lesion size (1 and 15 months), respiratory chain enzymes (1 and 15 months), and mitochondrial DNA copy number (mtDNAcn) (1 and 15 months) after CCI/sham. In the transgenics we quantified post-injury mtDNAcn and tangle burden. Results In the non-transgenics CCI caused acute behavioral deficits that improved or resolved by 1-month post-injury. Protein-normalized complex I and cytochrome oxidase activities were not significantly altered at 1 or 15 months, although complex I activity in the CCI ipsilesional cortex declined during that period. Hippocampal mtDNAcn was not altered by injury at 1 month, increased with age, and rose to the greatest extent in the CCI contralesional hippocampus. In the injured then aged transgenics, the ipsilesional hippocampus contained less mtDNA and fewer tangles than the contralesional hippocampus; mtDNAcn and tangle counts did not correlate. Conclusions As mice age their brains increase mtDNAcn as part of a compensatory response that preserves mitochondrial function, and TBI enhances this response. TBI may, therefore, increase the amount of compensation required to preserve late-life mitochondrial function. If TBI does modify AD risk, altering the trajectory or biology of aging-related mitochondrial changes could mediate the effect.
Collapse
Affiliation(s)
- Janna L Harris
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
- Departments of Cell Biology and Physiology, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Xiaowan Wang
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Sarah K Christian
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Lesya Novikova
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Anuradha Kalani
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Dongwei Hui
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Sadie Ferren
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Scott Barbay
- Departments of Physical Medicine and Rehabilitation, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Judit Perez Ortiz
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Randolph J Nudo
- Departments of Physical Medicine and Rehabilitation, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
| | - William M Brooks
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
- Departments of Neurology, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
- Departments of Neurology, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Prabhakar Chalise
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
- Departments of Biostatistics and Data Science, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Mary Lou Michaelis
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Elias K Michaelis
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
- Departments of Cell Biology and Physiology, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
- Departments of Neurology, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
- Departments of Biochemistry and Molecular Biology, University of Kansas Alzheimer's Disease Research Center, The University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
9
|
Hubbard WB, Vekaria HJ, Velmurugan GV, Kalimon OJ, Prajapati P, Brown E, Geisler JG, Sullivan PG. Mitochondrial Dysfunction After Repeated Mild Blast Traumatic Brain Injury Is Attenuated by a Mild Mitochondrial Uncoupling Prodrug. J Neurotrauma 2023; 40:2396-2409. [PMID: 37476976 PMCID: PMC10653072 DOI: 10.1089/neu.2023.0102] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Abstract
Mild traumatic brain injury (mTBI) results in impairment of brain metabolism, which is propagated by mitochondrial dysfunction in the brain. Mitochondrial dysfunction has been identified as a pathobiological therapeutic target to quell cellular dyshomeostasis. Further, therapeutic approaches targeting mitochondrial impairments, such as mild mitochondrial uncoupling, have been shown to alleviate behavioral alterations after TBI. To examine how mild mitochondrial uncoupling modulates acute mitochondrial outcomes in a military-relevant model of mTBI, we utilized repeated blast overpressure of 11 psi peak overpressure to model repeated mild blast traumatic brain injury (rmbTBI) in rats followed by assessment of mitochondrial respiration and mitochondrial-related oxidative damage at 2 days post-rmbTBI. Treatment groups were administered 8 or 80 mg/kg MP201, a prodrug of 2,4 dinitrophenol (DNP) that displays improved pharmacokinetics compared with its metabolized form. Synaptic and glia-enriched mitochondria were isolated using fractionated a mitochondrial magnetic separation technique. There was a consistent physiological response, decreased heart rate, following mbTBI among experimental groups. Although there was a lack of injury effect in mitochondrial respiration of glia-enriched mitochondria, there were impairments in mitochondrial respiration in synaptic mitochondria isolated from the prefrontal cortex (PFC) and the amygdala/entorhinal/piriform cortex (AEP) region. Impairments in synaptic mitochondrial respiration were rescued by oral 80 mg/kg MP201 treatment after rmbTBI, which may be facilitated by increases in complex II and complex IV activity. Mitochondrial oxidative damage in glia-enriched mitochondria was increased in the PFC and hippocampus after rmbTBI. MP201 treatment alleviated elevated glia-enriched mitochondrial oxidative damage following rmbTBI. However, there was a lack of injury-associated differences in oxidative damage in synaptic mitochondria. Overall, our report demonstrates that rmbTBI results in mitochondrial impairment diffusely throughout the brain and mild mitochondrial uncoupling can restore mitochondrial bioenergetics and oxidative balance.
Collapse
Affiliation(s)
- W. Brad Hubbard
- Lexington Veterans' Affairs Healthcare System, Lexington, Kentucky, USA
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Hemendra J. Vekaria
- Lexington Veterans' Affairs Healthcare System, Lexington, Kentucky, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Gopal V. Velmurugan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Olivia J. Kalimon
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Paresh Prajapati
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Emily Brown
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - John G. Geisler
- Mitochon Pharmaceuticals, Inc., Blue Bell, Pennsylvania, USA
| | - Patrick G. Sullivan
- Lexington Veterans' Affairs Healthcare System, Lexington, Kentucky, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
10
|
Strogulski NR, Portela LV, Polster BM, Loane DJ. Fundamental Neurochemistry Review: Microglial immunometabolism in traumatic brain injury. J Neurochem 2023; 167:129-153. [PMID: 37759406 PMCID: PMC10655864 DOI: 10.1111/jnc.15959] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Traumatic brain injury (TBI) is a devastating neurological disorder caused by a physical impact to the brain that promotes diffuse damage and chronic neurodegeneration. Key mechanisms believed to support secondary brain injury include mitochondrial dysfunction and chronic neuroinflammation. Microglia and brain-infiltrating macrophages are responsible for neuroinflammatory cytokine and reactive oxygen species (ROS) production after TBI. Their production is associated with loss of homeostatic microglial functions such as immunosurveillance, phagocytosis, and immune resolution. Beyond providing energy support, mitochondrial metabolic pathways reprogram the pro- and anti-inflammatory machinery in immune cells, providing a critical immunometabolic axis capable of regulating immunologic response to noxious stimuli. In the brain, the capacity to adapt to different environmental stimuli derives, in part, from microglia's ability to recognize and respond to changes in extracellular and intracellular metabolite levels. This capacity is met by an equally plastic metabolism, capable of altering immune function. Microglial pro-inflammatory activation is associated with decreased mitochondrial respiration, whereas anti-inflammatory microglial polarization is supported by increased oxidative metabolism. These metabolic adaptations contribute to neuroimmune responses, placing mitochondria as a central regulator of post-traumatic neuroinflammation. Although it is established that profound neurometabolic changes occur following TBI, key questions related to metabolic shifts in microglia remain unresolved. These include (a) the nature of microglial mitochondrial dysfunction after TBI, (b) the hierarchical positions of different metabolic pathways such as glycolysis, pentose phosphate pathway, glutaminolysis, and lipid oxidation during secondary injury and recovery, and (c) how immunometabolism alters microglial phenotypes, culminating in chronic non-resolving neuroinflammation. In this basic neurochemistry review article, we describe the contributions of immunometabolism to TBI, detail primary evidence of mitochondrial dysfunction and metabolic impairments in microglia and macrophages, discuss how major metabolic pathways contribute to post-traumatic neuroinflammation, and set out future directions toward advancing immunometabolic phenotyping in TBI.
Collapse
Affiliation(s)
- Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Luis V. Portela
- Neurotrauma and Biomarkers Laboratory, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Brian M. Polster
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David J. Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Allen J, Pham L, Bond ST, O’Brien WT, Spitz G, Shultz SR, Drew BG, Wright DK, McDonald SJ. Acute effects of single and repeated mild traumatic brain injury on levels of neurometabolites, lipids, and mitochondrial function in male rats. Front Mol Neurosci 2023; 16:1208697. [PMID: 37456524 PMCID: PMC10338885 DOI: 10.3389/fnmol.2023.1208697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Mild traumatic brain injuries (mTBIs) are the most common form of acquired brain injury. Symptoms of mTBI are thought to be associated with a neuropathological cascade, potentially involving the dysregulation of neurometabolites, lipids, and mitochondrial bioenergetics. Such alterations may play a role in the period of enhanced vulnerability that occurs after mTBI, such that a second mTBI will exacerbate neuropathology. However, it is unclear whether mTBI-induced alterations in neurometabolites and lipids that are involved in energy metabolism and other important cellular functions are exacerbated by repeat mTBI, and if such alterations are associated with mitochondrial dysfunction. Methods In this experiment, using a well-established awake-closed head injury (ACHI) paradigm to model mTBI, male rats were subjected to a single injury, or five injuries delivered 1 day apart, and injuries were confirmed with a beam-walk task and a video observation protocol. Abundance of several neurometabolites was evaluated 24 h post-final injury in the ipsilateral and contralateral hippocampus using in vivo proton magnetic resonance spectroscopy (1H-MRS), and mitochondrial bioenergetics were evaluated 30 h post-final injury, or at 24 h in place of 1H-MRS, in the rostral half of the ipsilateral hippocampus. Lipidomic evaluations were conducted in the ipsilateral hippocampus and cortex. Results We found that behavioral deficits in the beam task persisted 1- and 4 h after the final injury in rats that received repetitive mTBIs, and this was paralleled by an increase and decrease in hippocampal glutamine and glucose, respectively, whereas a single mTBI had no effect on sensorimotor and metabolic measurements. No group differences were observed in lipid levels and mitochondrial bioenergetics in the hippocampus, although some lipids were altered in the cortex after repeated mTBI. Discussion The decrease in performance in sensorimotor tests and the presence of more neurometabolic and lipidomic abnormalities, after repeated but not singular mTBI, indicates that multiple concussions in short succession can have cumulative effects. Further preclinical research efforts are required to understand the underlying mechanisms that drive these alterations to establish biomarkers and inform treatment strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Josh Allen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Louise Pham
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Simon T. Bond
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - William T. O’Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Gershon Spitz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Monash-Epworth Rehabilitation Research Centre, Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Health Sciences, Vancouver Island University, Nanaimo, BC, Canada
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Brian G. Drew
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - David K. Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Reeder EL, O'Connell CJ, Collins SM, Traubert OD, Norman SV, Cáceres RA, Sah R, Smith DW, Robson MJ. Increased Carbon Dioxide Respiration Prevents the Effects of Acceleration/Deceleration Elicited Mild Traumatic Brain Injury. Neuroscience 2023; 509:20-35. [PMID: 36332692 DOI: 10.1016/j.neuroscience.2022.10.016] [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: 08/12/2022] [Revised: 09/30/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022]
Abstract
Acceleration/deceleration forces are a common component of various causes of mild traumatic brain injury (mTBI) and result in strain and shear forces on brain tissue. A small quantifiable volume dubbed the compensatory reserve volume (CRV) permits energy transmission to brain tissue during acceleration/deceleration events. The CRV is principally regulated by cerebral blood flow (CBF) and CBF is primarily determined by the concentration of inspired carbon dioxide (CO2). We hypothesized that experimental hypercapnia (i.e. increased inspired concentration of CO2) may act to prevent and mitigate the actions of acceleration/deceleration-induced TBI. To determine these effects C57Bl/6 mice underwent experimental hypercapnia whereby they were exposed to medical-grade atmospheric air or 5% CO2 immediately prior to an acceleration/deceleration-induced mTBI paradigm. mTBI results in significant increases in righting reflex time (RRT), reductions in core body temperature, and reductions in general locomotor activity-three hours post injury (hpi). Experimental hypercapnia immediately preceding mTBI was found to prevent mTBI-induced increases in RRT and reductions in core body temperature and general locomotor activity. Ribonucleic acid (RNA) sequencing conducted four hpi revealed that CO2 exposure prevented mTBI-induced transcriptional alterations of several targets related to oxidative stress, immune, and inflammatory signaling. Quantitative real-time PCR analysis confirmed the prevention of mTBI-induced increases in mitogen-activated protein kinase kinase kinase 6 and metallothionein-2. These initial proof of concept studies reveal that increases in inspired CO2 mitigate the detrimental contributions of acceleration/deceleration events in mTBI and may feasibly be translated in the future to humans using a medical device seeking to prevent mTBI among high-risk groups.
Collapse
Affiliation(s)
- Evan L Reeder
- University of Cincinnati James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH 45267, USA
| | - Christopher J O'Connell
- University of Cincinnati James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH 45267, USA
| | - Sean M Collins
- University of Cincinnati James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH 45267, USA
| | - Owen D Traubert
- University of Cincinnati College of Arts and Sciences, Department of Biological Sciences, Cincinnati, OH 45221, USA
| | - Sophia V Norman
- University of Cincinnati College of Arts and Sciences, Department of Biological Sciences, Cincinnati, OH 45221, USA
| | - Román A Cáceres
- University of Cincinnati College of Medicine, Department of Cancer and Cell Biology Cincinnati, OH 45267, USA
| | - Renu Sah
- University of Cincinnati College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH 45267, USA
| | | | - Matthew J Robson
- University of Cincinnati James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH 45267, USA.
| |
Collapse
|
13
|
Yanckello LM, Chang YH, Sun M, Chlipala G, Green SJ, Lei Z, Ericsson AC, Xing X, Hammond TC, Bachstetter AD, Lin AL. Inulin supplementation prior to mild traumatic brain injury mitigates gut dysbiosis, and brain vascular and white matter deficits in mice. FRONTIERS IN MICROBIOMES 2022; 1:986951. [PMID: 36756543 PMCID: PMC9903356 DOI: 10.3389/frmbi.2022.986951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Introduction Mild traumatic brain injury (mTBI) has been shown to negatively alter bacterial diversity and composition within the gut, known as dysbiosis, in rodents and humans. These changes cause secondary consequences systemically through decreased bacterial metabolites such as short chain fatty acids (SCFAs) which play a role in inflammation and metabolism. The goal of the study was to identify if giving prebiotic inulin prior to closed head injury (CHI) could mitigate gut dysbiosis, increase SCFAs, and improve recovery outcomes, including protecting cerebral blood flow (CBF) and white matter integrity (WMI) in young mice. Methods We fed mice at 2 months of age with either inulin or control diet (with cellulose as fiber source) for two months before the CHI and continued till the end of the study. We analyzed gut microbiome composition and diversity, determined SCFAs levels, and measured CBF and WMI using MRI. We compared the results with Naïve and Sham-injury mice at 24 hours, 1.5 months, and 3-4 months post-injury. Results We found that both CHI and Sham mice had time-dependent changes in gut composition and diversity after surgery. Inulin significantly reduced the abundance of pathobiont bacteria, such as E. coli, Desulfovibrio spp and Pseudomonas aeruginosa, in Sham and CHI mice compared to mice fed with control diet. On the other hand, inulin increased SCFAs-producing bacteria, such as Bifidobacterium spp and Lactobacillus spp, increased levels of SCFAs, including butyrate and propionate, and significantly altered beta diversity as early as 24 hours post-injury, which lasted up to 3-4 months post-injury. The mitigation of dysbiosis is associated with protection of WMI in fimbria, internal and external capsule, and CBF in the right hippocampus of CHI mice, suggesting protection of memory and cognitive functions. Discussion The results indicate that giving inulin prior to CHI could promote recovery outcome through gut microbiome modulation. As inulin, microbiome analysis, and MRI are readily to be used in humans, the findings from the study may pave a way for a cost-effective, accessible intervention for those at risk of sustaining a head injury, such as military personnel or athletes in contact sports.
Collapse
Affiliation(s)
- Lucille M. Yanckello
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Ya-Hsuan Chang
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - McKenna Sun
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - George Chlipala
- Research Informatics Core, University of Illinois Chicago, Chicago, IL, United States
| | - Stefan J. Green
- Genomics and Microbiome Core Facility, Rush University, Chicago, IL, United States
| | - Zhentian Lei
- Metabolomics Center, University of Missouri, Columbia, MO, United States
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Xin Xing
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Computer Science, University of Kentucky, Lexington, KY, United States
| | - Tyler C. Hammond
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Adam D. Bachstetter
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, KY, United States
| | - Ai-Ling Lin
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
- Department of Radiology, University of Missouri, Columbia, MO, United States
- Institute for Data Science &Informatics, University of Missouri, Columbia, MO, United States
- Department of Biological Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
14
|
Macheda T, Roberts K, Bachstetter AD. Electromagnetic Controlled Closed-Head Model of Mild Traumatic Brain Injury in Mice. J Vis Exp 2022:10.3791/64556. [PMID: 36279529 PMCID: PMC10550048 DOI: 10.3791/64556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Highly reproducible animal models of traumatic brain injury (TBI), with well-defined pathologies, are needed for testing therapeutic interventions and understanding the mechanisms of how a TBI alters brain function. The availability of multiple animal models of TBI is necessary to model the different aspects and severities of TBI seen in people. This manuscript describes the use of a midline closed head injury (CHI) to develop a mouse model of mild TBI. The model is considered mild because it does not produce structural brain lesions based on neuroimaging or gross neuronal loss. However, a single impact creates enough pathology that cognitive impairment is measurable at least 1 month after injury. A step-by-step protocol to induce a CHI in mice using a stereotaxically guided electromagnetic impactor is defined in the paper. The benefits of the mild midline CHI model include the reproducibility of the injury-induced changes with low mortality. The model has been temporally characterized up to 1 year after the injury for neuroimaging, neurochemical, neuropathological, and behavioral changes. The model is complementary to open skull models of controlled cortical impact using the same impactor device. Thus, labs can model both mild diffuse TBI and focal moderate-to-severe TBI with the same impactor.
Collapse
Affiliation(s)
- Teresa Macheda
- Spinal Cord & Brain Injury Research Center, University of Kentucky
| | - Kelly Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky
| | - Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky; Department of Neuroscience, University of Kentucky; Sanders-Brown Center on Aging, University of Kentucky;
| |
Collapse
|
15
|
Strogulski NR, Stefani MA, Böhmer AE, Hansel G, Rodolphi MS, Kopczynski A, de Oliveira VG, Stefani ET, Portela JV, Schmidt AP, Oses JP, Smith DH, Portela LV. Cerebrospinal fluid purinomics as a biomarker approach to predict outcome after severe traumatic brain injury. J Neurochem 2022; 161:173-186. [PMID: 35157328 PMCID: PMC9035090 DOI: 10.1111/jnc.15590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/19/2022] [Accepted: 01/31/2022] [Indexed: 11/27/2022]
Abstract
Severe traumatic brain injury (TBI) is associated with high rates of mortality and long-term disability linked to neurochemical abnormalities. Although purine-derivatives play important roles in TBI pathogenesis in preclinical models, little is known about potential changes in purine levels and their implications in human TBI. We assessed cerebrospinal fluid (CSF) levels of purines in severe TBI patients as potential biomarkers that predict mortality and long-term dysfunction. This was a cross-sectional study performed in 17 severe TBI patients (Glasgow Coma Scale < 8) and 51 controls. Two to four hours after admission to ICU, patients were submitted to ventricular drainage and CSF collection for quantification of adenine and guanine purine-derivatives by HPLC. TBI patients survival was followed up to 3 days from admission. A neurofunctional assessment was performed through the modified Rankin Scale (mRS) two years after ICU admission. Purine levels were compared between control and TBI patients, and between surviving and non-surviving patients. Relative to controls, TBI patients presented increased CSF levels of GDP, guanosine, adenosine, inosine, hypoxanthine, and xanthine. Further, GTP, GDP, IMP, and xanthine levels were different between surviving and non-surviving patients. Among the purines, guanosine was associated with improved mRS (p=0.042; r= -0.506). Remarkably, GTP displayed predictive value (AUC=0.841, p=0.024) for discriminating survival vs. non-survival patients up to three days from admission. These results support TBI-specific purine signatures, suggesting GTP as a promising biomarker of mortality, and guanosine as an indicator of long-term functional disability.
Collapse
Affiliation(s)
- Nathan R Strogulski
- Laboratory of Neurotrauma e Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Marco Antonio Stefani
- Laboratory of Neuroanatomy, Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Ana Elisa Böhmer
- Laboratory of Neurotrauma e Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Gisele Hansel
- Neuroinflammation and Neuroimmunology Laboratory, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Marcelo S Rodolphi
- Laboratory of Neurotrauma e Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Afonso Kopczynski
- Laboratory of Neurotrauma e Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Vitória G de Oliveira
- Laboratory of Neurotrauma e Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Eduarda T Stefani
- Laboratory of Neuroanatomy, Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Juliana V Portela
- Laboratory of Neurotrauma e Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - André P Schmidt
- Laboratory of Neurotrauma e Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Department of Anesthesia and Perioperative Medicine, Hospital de Clínicas de Porto Alegre (HCPA), UFRGS, Porto Alegre, RS, Brazil. Department of Anesthesia, Santa Casa de Porto Alegre, Universidade Federal de Ciências Médicas de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil. Department of Anesthesia, Hospital Nossa Senhora da Conceição, Porto Alegre, RS, Brazil
| | - Jean Pierre Oses
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, RS, Brazil
| | - Douglas H Smith
- Penn Center for Brain Injury and Repair and Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Luis V Portela
- Laboratory of Neurotrauma e Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| |
Collapse
|
16
|
Yanckello LM, Fanelli B, McCulloch S, Xing X, Sun M, Hammond TC, Colwell R, Gu Z, Ericsson AC, Chang YH, Bachstetter AD, Lin AL. Inulin Supplementation Mitigates Gut Dysbiosis and Brain Impairment Induced by Mild Traumatic Brain Injury during Chronic Phase. JOURNAL OF CELLULAR IMMUNOLOGY 2022; 4:50-64. [PMID: 35611116 PMCID: PMC9126115 DOI: 10.33696/immunology.4.132] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mild traumatic brain injury (mTBI) has been shown to acutely alter the gut microbiome diversity and composition, known as dysbiosis, which can further exacerbate metabolic and vascular changes in the brain in both humans and rodents. However, it remains unknown how mTBI affects the gut microbiome in the chronic phase recovery (past one week post injury). It is also unknown if injury recovery can be improved by mitigating dysbiosis. The goal of the study is to fill the knowledge gap. First, we aim to understand how mTBI alters the gut microbiome through the chronic period of recovery (3 months post injury). In addition, as the gut microbiome can be modulated by diet, we also investigated if prebiotic inulin, a fermentable fiber that promotes growth of beneficial bacteria and metabolites, would mitigate dysbiosis, improve systemic metabolism, and protect brain structural and vascular integrity when administered after 3 months post closed head injury (CHI). We found that CHI given to male mice at 4 months of age induced gut dysbiosis which peaked at 1.5 months post injury, reduced cerebral blood flow (CBF) and altered brain white matter integrity. Interestingly, we also found that Sham mice had transient dysbiosis, which peaked 24 hours after injury and then normalized. After 8 weeks of inulin feeding, CHI mice had increased abundance of beneficial/anti-inflammatory bacteria, reduced abundance of pathogenic bacteria, enriched levels of short-chain fatty acids, and restored CBF in both hippocampi and left thalamus, compared to the CHI-control fed and Sham groups. Using machine learning, we further identified top bacterial species that separate Sham and CHI mice with and without the diet. Our results indicate that there is an injury- and time-dependent dysbiosis between CHI and Sham mice; inulin is effective to mitigate dysbiosis and improve brain injury recovery in the CHI mice. As there are currently no effective treatments for mTBI, the study may have profound implications for developing therapeutics or preventive interventions in the future.
Collapse
Affiliation(s)
- Lucille M. Yanckello
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States of America
| | - Brian Fanelli
- CosmosID Inc., Rockville, MD, United States of America
| | | | - Xin Xing
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Computer Science, University of Kentucky, Lexington, KY, United States of America
| | - McKenna Sun
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Tyler C. Hammond
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Rita Colwell
- CosmosID Inc., Rockville, MD, United States of America
| | - Zezong Gu
- Department of Pathology & Anatomical Sciences, University of Missouri, Columbia, MO, United States of America
- Harry S. Truman Memorial Veteran Hospital, Columbia, MO, United States of America
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States of America
| | - Ya-Hsuan Chang
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States of America
| | - Adam D. Bachstetter
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
- Spinal Cord and Brain Injury Research Center, University of Kentucky, KY, United States of America
| | - Ai-Ling Lin
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States of America
- Department of Radiology, University of Missouri, Columbia, MO, United States of America
- Institute for Data Science &Informatics, University of Missouri, Columbia, MO United States of America
| |
Collapse
|
17
|
Hubbard WB, Spry ML, Gooch JL, Cloud AL, Vekaria HJ, Burden S, Powell DK, Berkowitz BA, Geldenhuys WJ, Harris NG, Sullivan PG. Clinically relevant mitochondrial-targeted therapy improves chronic outcomes after traumatic brain injury. Brain 2021; 144:3788-3807. [PMID: 34972207 PMCID: PMC8719838 DOI: 10.1093/brain/awab341] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 11/14/2022] Open
Abstract
Pioglitazone, an FDA-approved compound, has been shown to target the novel mitochondrial protein mitoNEET and produce short-term neuroprotection and functional benefits following traumatic brain injury. To expand on these findings, we now investigate the dose- and time-dependent effects of pioglitazone administration on mitochondrial function after experimental traumatic brain injury. We then hypothesize that optimal pioglitazone dosing will lead to ongoing neuroprotection and cognitive benefits that are dependent on pioglitazone-mitoNEET signalling pathways. We show that delayed intervention is significantly more effective than early intervention at improving acute mitochondrial bioenergetics in the brain after traumatic brain injury. In corroboration, we demonstrate that mitoNEET is more heavily expressed, especially near the cortical contusion, in the 18 h following traumatic brain injury. To explore whether these findings relate to ongoing pathological and behavioural outcomes, mice received controlled cortical impact followed by initiation of pioglitazone treatment at either 3 or 18 h post-injury. Mice with treatment initiation at 18 h post-injury exhibited significantly improved behaviour and tissue sparing compared to mice with pioglitazone initiated at 3 h post-injury. Further using mitoNEET knockout mice, we show that this therapeutic effect is dependent on mitoNEET. Finally, we demonstrate that delayed pioglitazone treatment improves serial motor and cognitive performance in conjunction with attenuated brain atrophy after traumatic brain injury. This study illustrates that mitoNEET is the critical target for delayed pioglitazone intervention after traumatic brain injury, mitochondrial-targeting is highly time-dependent after injury and there is an extended therapeutic window to effectively treat mitochondrial dysfunction after brain injury.
Collapse
Affiliation(s)
- W Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA
- Lexington VA Healthcare System, Lexington, KY 40502, USA
| | - Malinda L Spry
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Jennifer L Gooch
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Amber L Cloud
- College of Medicine, University of Kentucky, Lexington, KY 40508, USA
| | - Hemendra J Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Shawn Burden
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - David K Powell
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508, USA
| | - Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Neil G Harris
- UCLA Brain Injury Research Center, Department of Neurosurgery, and Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508, USA
- Lexington VA Healthcare System, Lexington, KY 40502, USA
| |
Collapse
|
18
|
Mester JR, Bazzigaluppi P, Dorr A, Beckett T, Burke M, McLaurin J, Sled JG, Stefanovic B. Attenuation of tonic inhibition prevents chronic neurovascular impairments in a Thy1-ChR2 mouse model of repeated, mild traumatic brain injury. Am J Cancer Res 2021; 11:7685-7699. [PMID: 34335958 PMCID: PMC8315057 DOI: 10.7150/thno.60190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: Mild traumatic brain injury (mTBI), the most common type of brain trauma, frequently leads to chronic cognitive and neurobehavioral deficits. Intervening effectively is impeded by our poor understanding of its pathophysiological sequelae. Methods: To elucidate the long-term neurovascular sequelae of mTBI, we combined optogenetics, two-photon fluorescence microscopy, and intracortical electrophysiological recordings in mice to selectively stimulate peri-contusional neurons weeks following repeated closed-head injury and probe individual vessel's function and local neuronal reactivity. Results: Compared to sham-operated animals, mTBI mice showed doubled cortical venular speeds (115 ± 25%) and strongly elevated cortical venular reactivity (53 ± 17%). Concomitantly, the pericontusional neurons exhibited attenuated spontaneous activity (-57 ± 79%) and decreased reactivity (-47 ± 28%). Post-mortem immunofluorescence revealed signs of peri-contusional senescence and DNA damage, in the absence of neuronal loss or gliosis. Alteration of neuronal and vascular functioning was largely prevented by chronic, low dose, systemic administration of a GABA-A receptor inverse agonist (L-655,708), commencing 3 days following the third impact. Conclusions: Our findings indicate that repeated mTBI leads to dramatic changes in the neurovascular unit function and that attenuation of tonic inhibition can prevent these alterations. The sustained disruption of the neurovascular function may underlie the concussed brain's long-term susceptibility to injury, and calls for development of better functional assays as well as of neurovascularly targeted interventions.
Collapse
|
19
|
Baker TL, Agoston DV, Brady RD, Major B, McDonald SJ, Mychasiuk R, Wright DK, Yamakawa GR, Sun M, Shultz SR. Targeting the Cerebrovascular System: Next-Generation Biomarkers and Treatment for Mild Traumatic Brain Injury. Neuroscientist 2021; 28:594-612. [PMID: 33966527 DOI: 10.1177/10738584211012264] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The diagnosis, prognosis, and treatment of mild traumatic brain injuries (mTBIs), such as concussions, are significant unmet medical issues. The kinetic forces that occur in mTBI adversely affect the cerebral vasculature, making cerebrovascular injury (CVI) a pathophysiological hallmark of mTBI. Given the importance of a healthy cerebrovascular system in overall brain function, CVI is likely to contribute to neurological dysfunction after mTBI. As such, CVI and related pathomechanisms may provide objective biomarkers and therapeutic targets to improve the clinical management and outcomes of mTBI. Despite this potential, until recently, few studies have focused on the cerebral vasculature in this context. This article will begin by providing a brief overview of the cerebrovascular system followed by a review of the literature regarding how mTBI can affect the integrity and function of the cerebrovascular system, and how this may ultimately contribute to neurological dysfunction and neurodegenerative conditions. We then discuss promising avenues of research related to mTBI biomarkers and interventions that target CVI, and conclude that a clinical approach that takes CVI into account could result in substantial improvements in the care and outcomes of patients with mTBI.
Collapse
Affiliation(s)
- Tamara L Baker
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Brendan Major
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
20
|
Wiegand TLT, Sollmann N, Bonke EM, Umeasalugo KE, Sobolewski KR, Plesnila N, Shenton ME, Lin AP, Koerte IK. Translational neuroimaging in mild traumatic brain injury. J Neurosci Res 2021; 100:1201-1217. [PMID: 33789358 DOI: 10.1002/jnr.24840] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/09/2021] [Accepted: 03/17/2021] [Indexed: 01/26/2023]
Abstract
Traumatic brain injuries (TBIs) are common with an estimated 27.1 million cases per year. Approximately 80% of TBIs are categorized as mild TBI (mTBI) based on initial symptom presentation. While in most individuals, symptoms resolve within days to weeks, in some, symptoms become chronic. Advanced neuroimaging has the potential to characterize brain morphometric, microstructural, biochemical, and metabolic abnormalities following mTBI. However, translational studies are needed for the interpretation of neuroimaging findings in humans with respect to the underlying pathophysiological processes, and, ultimately, for developing novel and more targeted treatment options. In this review, we introduce the most commonly used animal models for the study of mTBI. We then summarize the neuroimaging findings in humans and animals after mTBI and, wherever applicable, the translational aspects of studies available today. Finally, we highlight the importance of translational approaches and outline future perspectives in the field of translational neuroimaging in mTBI.
Collapse
Affiliation(s)
- Tim L T Wiegand
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nico Sollmann
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| | - Elena M Bonke
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
| | - Kosisochukwu E Umeasalugo
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Kristen R Sobolewski
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| | - Martha E Shenton
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander P Lin
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Clinical Spectroscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Inga K Koerte
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Ludwig-Maximilians-Universität, Munich, Germany
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Hubbard WB, Banerjee M, Vekaria H, Prakhya KS, Joshi S, Wang QJ, Saatman KE, Whiteheart SW, Sullivan PG. Differential Leukocyte and Platelet Profiles in Distinct Models of Traumatic Brain Injury. Cells 2021; 10:cells10030500. [PMID: 33652745 PMCID: PMC7996744 DOI: 10.3390/cells10030500] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/04/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) affects over 3 million individuals every year in the U.S. There is growing appreciation that TBI can produce systemic modifications, which are in part propagated through blood–brain barrier (BBB) dysfunction and blood–brain cell interactions. As such, platelets and leukocytes contribute to mechanisms of thromboinflammation after TBI. While these mechanisms have been investigated in experimental models of contusion brain injury, less is known regarding acute alterations following mild closed head injury. To investigate the role of platelet dynamics and bioenergetics after TBI, we employed two distinct, well-established models of TBI in mice: the controlled cortical impact (CCI) model of contusion brain injury and the closed head injury (CHI) model of mild diffuse brain injury. Hematology parameters, platelet-neutrophil aggregation, and platelet respirometry were assessed acutely after injury. CCI resulted in an early drop in blood leukocyte counts, while CHI increased blood leukocyte counts early after injury. Platelet-neutrophil aggregation was altered acutely after CCI compared to sham. Furthermore, platelet bioenergetic coupling efficiency was transiently reduced at 6 h and increased at 24 h post-CCI. After CHI, oxidative phosphorylation in intact platelets was reduced at 6 h and increased at 24 h compared to sham. Taken together, these data demonstrate that brain trauma initiates alterations in platelet-leukocyte dynamics and platelet metabolism, which may be time- and injury-dependent, providing evidence that platelets carry a peripheral signature of brain injury. The unique trend of platelet bioenergetics after two distinct types of TBI suggests the potential for utilization in prognosis.
Collapse
Affiliation(s)
- William Brad Hubbard
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA; (W.B.H.); (H.V.); (K.E.S.)
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508, USA
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY 40502, USA;
| | - Meenakshi Banerjee
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA; (M.B.); (K.S.P.); (S.J.)
| | - Hemendra Vekaria
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA; (W.B.H.); (H.V.); (K.E.S.)
| | | | - Smita Joshi
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA; (M.B.); (K.S.P.); (S.J.)
| | - Qing Jun Wang
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA; (W.B.H.); (H.V.); (K.E.S.)
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA
| | - Sidney W. Whiteheart
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY 40502, USA;
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA; (M.B.); (K.S.P.); (S.J.)
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA; (W.B.H.); (H.V.); (K.E.S.)
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508, USA
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY 40502, USA;
- Correspondence: ; Tel.: +1-859-323-4684
| |
Collapse
|
22
|
Proton Magnetic Resonance Spectroscopy of N-acetyl Aspartate in Chronic Schizophrenia, First Episode of Psychosis and High-Risk of Psychosis: A Systematic Review and Meta-Analysis. Neurosci Biobehav Rev 2020; 119:255-267. [PMID: 33068555 DOI: 10.1016/j.neubiorev.2020.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/01/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
N-acetyl-aspartate (NAA) is a readily measured marker of neuronal metabolism. Previous analyses in schizophrenia have shown NAA levels are low in frontal, temporal and thalamic regions, but may be underpowered to detect effects in other regions, in high-risk states and in first episode psychosis. We searched for magnetic resonance spectroscopy studies comparing NAA in chronic schizophrenia, first episode psychosis and high risk of psychosis to controls. 182 studies were included and meta-analysed using a random-effects model for each region and illness stage. NAA levels were significantly lower than controls in the frontal lobe [Hedge's g = -0.36, p < 0.001], hippocampus [-0.52, p < 0.001], temporal lobe [-0.35, p = 0.031], thalamus [-0.32, p = 0.012] and parietal lobe [-0.25, p = 0.028] in chronic schizophrenia, and lower than controls in the frontal lobe [-0.26, p = 0.002], anterior cingulate cortex [-0.24, p = 0.016] and thalamus [-0.28, p = 0.028] in first episode psychosis. NAA was lower in high-risk of psychosis in the hippocampus [-0.20, p = 0.049]. In schizophrenia, NAA alterations appear to begin in hippocampus, frontal cortex and thalamus, and extend later to many other regions.
Collapse
|
23
|
Shultz SR, McDonald SJ, Corrigan F, Semple BD, Salberg S, Zamani A, Jones NC, Mychasiuk R. Clinical Relevance of Behavior Testing in Animal Models of Traumatic Brain Injury. J Neurotrauma 2020; 37:2381-2400. [DOI: 10.1089/neu.2018.6149] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Frances Corrigan
- Department of Anatomy, University of South Australia, Adelaide, South Australia, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
24
|
Pham L, Wright DK, O'Brien WT, Bain J, Huang C, Sun M, Casillas-Espinosa PM, Shah AD, Schittenhelm RB, Sobey CG, Brady RD, O'Brien TJ, Mychasiuk R, Shultz SR, McDonald SJ. Behavioral, axonal, and proteomic alterations following repeated mild traumatic brain injury: Novel insights using a clinically relevant rat model. Neurobiol Dis 2020; 148:105151. [PMID: 33127468 DOI: 10.1016/j.nbd.2020.105151] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/07/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022] Open
Abstract
A history of mild traumatic brain injury (mTBI) is linked to a number of chronic neurological conditions, however there is still much unknown about the underlying mechanisms. To provide new insights, this study used a clinically relevant model of repeated mTBI in rats to characterize the acute and chronic neuropathological and neurobehavioral consequences of these injuries. Rats were given four sham-injuries or four mTBIs and allocated to 7-day or 3.5-months post-injury recovery groups. Behavioral analysis assessed sensorimotor function, locomotion, anxiety, and spatial memory. Neuropathological analysis included serum quantification of neurofilament light (NfL), mass spectrometry of the hippocampal proteome, and ex vivo magnetic resonance imaging (MRI). Repeated mTBI rats had evidence of acute cognitive deficits and prolonged sensorimotor impairments. Serum NfL was elevated at 7 days post injury, with levels correlating with sensorimotor deficits; however, no NfL differences were observed at 3.5 months. Several hippocampal proteins were altered by repeated mTBI, including those associated with energy metabolism, neuroinflammation, and impaired neurogenic capacity. Diffusion MRI analysis at 3.5 months found widespread reductions in white matter integrity. Taken together, these findings provide novel insights into the nature and progression of repeated mTBI neuropathology that may underlie lingering or chronic neurobehavioral deficits.
Collapse
Affiliation(s)
- Louise Pham
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - William T O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Jesse Bain
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Cheng Huang
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Anup D Shah
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; Monash Bioinformatics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
25
|
Macheda T, Roberts KN, Morganti JM, Braun DJ, Bachstetter AD. Optimization and validation of a modified radial-arm water maze protocol using a murine model of mild closed head traumatic brain injury. PLoS One 2020; 15:e0232862. [PMID: 32810143 PMCID: PMC7433858 DOI: 10.1371/journal.pone.0232862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/27/2020] [Indexed: 11/25/2022] Open
Abstract
Cognitive impairments can be a significant problem after a traumatic brain injury (TBI), which affects millions worldwide each year. There is a need for establish reproducible cognitive assays in rodents to better understand disease mechanisms and to develop therapeutic interventions towards treating TBI-induced impairments. Our goal was to validate and standardize the radial arm water maze (RAWM) test as an assay to screen for cognitive impairments caused by TBI. RAWM is a visuo-spatial learning test, originally designed for use with rats, and later adapted for mice. The present study investigates whether test procedures, such us the presence of extra-maze cues influences learning and memory performance. C57BL/6 mice were tested in an 8-arm RAWM using a four-day protocol. We demonstrated that two days of training, exposing the mice to extra-maze cues and a visible platform, influenced learning and memory performance. Mice that did not receive training performed poorer compared to mice trained. To further validate our RAWM protocol, we used scopolamine. We, also, demonstrated that a single mild closed head injury (CHI) caused deficits in this task at two weeks post-CHI. Our data supported the use of 7 trials per day and a spaced training protocol as key factor to unmask memory impairment following CHI. Here, we provide a detailed standard operating procedure for RAWM test, which can be applied to a variety of mouse models including neurodegenerative diseases and pathology, as well as when pharmacological approaches are used.
Collapse
Affiliation(s)
- Teresa Macheda
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Kelly N. Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Josh M. Morganti
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - David J. Braun
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Adam D. Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- * E-mail:
| |
Collapse
|
26
|
Wu L, Niu Z, Hu X, Liu H, Li S, Chen L, Zheng D, Liu Z, Liu T, Xu F, Manyande A, Wang J, Xia H. Regional cerebral metabolic levels and turnover in awake rats after acute or chronic spinal cord injury. FASEB J 2020; 34:10547-10559. [PMID: 32592196 DOI: 10.1096/fj.202000447r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Liang Wu
- Department of Neurosurgery General Hospital of Ningxia Medical University Yinchuan P.R. China
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- Ningxia Key Laboratory of Cerebrocranial Diseases Yinchuan P.R. China
- School of Clinical Medicine Ningxia Medical University Yinchuan P.R. China
| | - Zhanfeng Niu
- Department of Neurosurgery General Hospital of Ningxia Medical University Yinchuan P.R. China
| | - Xulei Hu
- Department of Neurosurgery General Hospital of Ningxia Medical University Yinchuan P.R. China
- Ningxia Key Laboratory of Cerebrocranial Diseases Yinchuan P.R. China
- School of Clinical Medicine Ningxia Medical University Yinchuan P.R. China
| | - Huili Liu
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Shuang Li
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
| | - Lei Chen
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
| | - Danhao Zheng
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Zhuang Liu
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Taotao Liu
- Department of Anesthesiology Peking University Third Hospital Beijing P.R. China
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Anne Manyande
- School of Human and Social Sciences University of West London London UK
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
- Hebei Provincial Key Laboratory of Basic Medicine for Diabetes 2nd Hospital of Shijiazhuang Shijiazhuang P.R. China
| | - Hechun Xia
- Department of Neurosurgery General Hospital of Ningxia Medical University Yinchuan P.R. China
- Ningxia Human Stem Cell Research Institute General Hospital of Ningxia Medical University Yinchuan P.R. China
| |
Collapse
|
27
|
An active avoidance behavioral paradigm for use in a mild closed head model of traumatic brain injury in mice. J Neurosci Methods 2020; 343:108831. [PMID: 32592717 DOI: 10.1016/j.jneumeth.2020.108831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND A mild traumatic brain injury (TBI) occurs to millions of people each year. Translational approaches to understanding the pathogenesis of neurological diseases and the testing of the effectiveness of interventions typically require cognitive function assays in rodents. NEW METHODS Our goal was to validate the active avoidance task using the GEMINI avoidance system in a mouse model of mild closed head injury (CHI). RESULTS We found that shock intensity had only a marginal effect on the test. We found that sex was an important biological variable, as female mice learned the task better than male mice. We demonstrate that a single mild CHI in mice caused deficits in the task at four weeks post-injury. COMPARISON WITH EXISTING METHODS Active avoidance is a classical conditioning test in which mice must pair the presence of a conditioned stimulus with moving between two chambers to avoid an electric shock. External conditions (i.e., apparatus), as well as inherent differences in the mice, which may not be directly linked to the model of the disease (i.e., sensory differences), can affect the reproducibility of a behavioral assay. Before our study, there was a lack of standard operating procedures and validated methods for the active avoidance behavior for phenotyping mouse models of injury and disease. CONCLUSION We offer a method for validating the active avoidance test, and a standard operating procedure, which will be useful in other models of neurological injury and disease.
Collapse
|
28
|
Formoterol, a β 2-adrenoreceptor agonist, induces mitochondrial biogenesis and promotes cognitive recovery after traumatic brain injury. Neurobiol Dis 2020; 140:104866. [PMID: 32289370 DOI: 10.1016/j.nbd.2020.104866] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/12/2020] [Accepted: 04/05/2020] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) leads to acute necrosis at the site of injury followed by a sequence of secondary events lasting from hours to weeks and often years. Targeting mitochondrial impairment following TBI has shown improvements in brain mitochondrial bioenergetics and neuronal function. Recently formoterol, a highly selective β2-adrenoreceptor agonist, was found to induce mitochondrial biogenesis (MB) via Gβγ-Akt-eNOS-sGC pathway. Activation of MB is a novel approach that has been shown to restore mitochondrial function in several disease and injury models. We hypothesized that activation of MB as a target of formoterol after TBI would mitigate mitochondrial dysfunction, enhance neuronal function and improve behavioral outcomes. TBI-injured C57BL/6 male mice were injected (i.p.) with vehicle (normal saline) or formoterol (0.3 mg/kg) at 15 min, 8 h, 16 h, 24 h and then daily after controlled cortical impact (CCI) until euthanasia. After CCI, mitochondrial copy number and bioenergetic function were decreased in the ipsilateral cortex of the CCI-vehicle group. Compared to CCI-vehicle, cortical and hippocampal mitochondrial respiration rates as well as cortical mitochondrial DNA copy number were increased in the CCI-formoterol group. Mitochondrial Ca2+ buffering capacity in the hippocampus was higher in the CCI-formoterol group compared to CCI-vehicle group. Both assessments of cognitive performance, novel object recognition (NOR) and Morris water maze (MWM), decreased following CCI and were restored in the CCI-formoterol group. Although no changes were seen in the amount of cortical tissue spared between CCI-formoterol and CCI-vehicle groups, elevated levels of hippocampal neurons and improved white matter sparing in the corpus callosum were observed in CCI-formoterol group. Collectively, these results indicate that formoterol-mediated MB activation may be a potential therapeutic target to restore mitochondrial bioenergetics and promote functional recovery after TBI.
Collapse
|
29
|
Co-localized impaired regional cerebrovascular reactivity in chronic concussion is associated with BOLD activation differences during a working memory task. Brain Imaging Behav 2020; 14:2438-2449. [PMID: 31903527 DOI: 10.1007/s11682-019-00194-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The purpose of this study was to quantify differences in blood oxygen level dependent (BOLD) activation on a working memory task, baseline cerebral blood flow (CBF0), and cerebrovascular reactivity (CVR) between participants with and without a history of concussion. A dual-echo pseudo-continuous arterial spin labelling (pCASL) sequence was performed on a group of 10 subjects with a previous concussion (126 ± 15 days prior) and on a control group (n = 10) during a visual working memory protocol. A separate dual-echo pCASL sequence was used to derive CVR and CBF0 measurements from a boxcar hypercapnic breathing protocol. Brain areas with significant activation differences on the working memory task between groups were identified and combined as an aggregate region of interest for CBF and CVR analyses. Areas of reduced BOLD activation during the working memory task in the concussed group included the ventral anterior cingulate cortex (ACC), the medial temporal gyrus (MTG), and the lateral occipital cortex in two loci. A single area of increased activation was located in the parietal operculum. Further analyses of CBF0 and CVR in these regions revealed reduced CVR in the concussed group in the MTG and ACC, while CBF0 did not differ. The differences in CVR between the two groups in these regions suggest that concussive injury may result in microvascular dysfunction. In turn, the decreased BOLD response during the task could be due to altered neurovascular coupling, rather than an impairment in neural activation alone. However, in other regions associated with working memory, unchanged CBF0 and CVR suggests that neural injury also persists after concussion. In the future, BOLD results should be normalized to CVR in order achieve a clearer understanding of the neural and vascular contributions to the differences in the signal.
Collapse
|
30
|
Pioglitazone improves working memory performance when administered in chronic TBI. Neurobiol Dis 2019; 132:104611. [PMID: 31513844 DOI: 10.1016/j.nbd.2019.104611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 09/02/2019] [Accepted: 09/06/2019] [Indexed: 01/26/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability in the United States. Even in comparatively mild injuries, cognitive and behavioral symptoms can persist for years, and there are currently no established strategies for mitigating symptoms in chronic injury. A key feature of TBI-induced damage in acute and chronic injury is disruption of metabolic pathways. As neurotransmission, and therefore cognition, are highly dependent on the supply of energy, we hypothesized that modulating metabolic activity could help restore behavioral performance even when treatment was initiated weeks after TBI. We treated rats with pioglitazone, a FDA-approved drug for diabetes, beginning 46 days after lateral fluid percussion injury and tested working memory performance in the radial arm maze (RAM) after 14 days of treatment. Pioglitazone treated TBI rats performed significantly better in the RAM test than untreated TBI rats, and similarly to control animals. While hexokinase activity in hippocampus was increased by pioglitazone treatment, there was no upregulation of either the neuronal glucose transporter or hexokinase enzyme expression. Expression of glial markers GFAP and Iba-1 were also not influenced by pioglitazone treatment. These studies suggest that targeting brain metabolism, in particular hippocampal metabolism, may be effective in alleviating cognitive symptoms in chronic TBI.
Collapse
|
31
|
Abstract
Next to cancer, Alzheimer's disease (AD) and dementia is probably the most worrying health problem facing the Western world today. A large number of clinical trials have failed to show any benefit of the tested drugs in stabilizing or reversing the steady decline in cognitive function that is suffered by dementia patients. Although the pathological features of AD consisting of beta-amyloid plaques and tau tangles are well established, considerable debate exists concerning the genetic or lifestyle factors that predispose individuals to developing dementia. Photobiomodulation (PBM) describes the therapeutic use of red or near-infrared light to stimulate healing, relieve pain and inflammation, and prevent tissue from dying. In recent years PBM has been applied for a diverse range of brain disorders, frequently applied in a non-invasive manner by shining light on the head (transcranial PBM). The present review discusses the mechanisms of action of tPBM in the brain, and summarizes studies that have used tPBM to treat animal models of AD. The results of a limited number of clinical trials that have used tPBM to treat patients with AD and dementia are discussed.
Collapse
Affiliation(s)
- Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| |
Collapse
|
32
|
Gupte R, Brooks W, Vukas R, Pierce J, Harris J. Sex Differences in Traumatic Brain Injury: What We Know and What We Should Know. J Neurotrauma 2019; 36:3063-3091. [PMID: 30794028 PMCID: PMC6818488 DOI: 10.1089/neu.2018.6171] [Citation(s) in RCA: 287] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is growing recognition of the problem of male bias in neuroscience research, including in the field of traumatic brain injury (TBI) where fewer women than men are recruited to clinical trials and male rodents have predominantly been used as an experimental injury model. Despite TBI being a leading cause of mortality and disability worldwide, sex differences in pathophysiology and recovery are poorly understood, limiting clinical care and successful drug development. Given growing interest in sex as a biological variable affecting injury outcomes and treatment efficacy, there is a clear need to summarize sex differences in TBI. This scoping review presents an overview of current knowledge of sex differences in TBI and a comparison of human and animal studies. We found that overall, human studies report worse outcomes in women than men, whereas animal studies report better outcomes in females than males. However, closer examination shows that multiple factors including injury severity, sample size, and experimental injury model may differentially interact with sex to affect TBI outcomes. Additionally, we explore how sex differences in mitochondrial structure and function might contribute to possible sex differences in TBI outcomes. We propose recommendations for future investigations of sex differences in TBI, which we hope will lead to improved patient management, prognosis, and translation of therapies from bench to bedside.
Collapse
Affiliation(s)
- Raeesa Gupte
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - William Brooks
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas
- Hoglund Brain Center, University of Kansas Medical Center, Kansas City, Kansas
- The University of Kansas Clinical and Translational Sciences Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Rachel Vukas
- School of Medicine, Dykes Library of Health Sciences, University of Kansas Medical Center, Kansas City, Kansas
| | - Janet Pierce
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Janna Harris
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
- Hoglund Brain Center, University of Kansas Medical Center, Kansas City, Kansas
- Address correspondence to: Janna Harris, PhD, Hoglund Brain Imaging Center, MS 1052, 3901 Rainbow Boulevard, Kansas City, KS 66160
| |
Collapse
|
33
|
Sta Maria NS, Sargolzaei S, Prins ML, Dennis EL, Asarnow RF, Hovda DA, Harris NG, Giza CC. Bridging the gap: Mechanisms of plasticity and repair after pediatric TBI. Exp Neurol 2019; 318:78-91. [PMID: 31055004 DOI: 10.1016/j.expneurol.2019.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/09/2019] [Accepted: 04/25/2019] [Indexed: 01/25/2023]
Abstract
Traumatic brain injury is the leading cause of death and disability in the United States, and may be associated with long lasting impairments into adulthood. The multitude of ongoing neurobiological processes that occur during brain maturation confer both considerable vulnerability to TBI but may also provide adaptability and potential for recovery. This review will examine and synthesize our current understanding of developmental neurobiology in the context of pediatric TBI. Delineating this biology will facilitate more targeted initial care, mechanism-based therapeutic interventions and better long-term prognostication and follow-up.
Collapse
Affiliation(s)
- Naomi S Sta Maria
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, 1501 San Pablo Street, ZNI115, Los Angeles, CA 90033, United States of America.
| | - Saman Sargolzaei
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America.
| | - Mayumi L Prins
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Steve Tisch BrainSPORT Program, University of California at Los Angeles, Los Angeles, CA, United States of America.
| | - Emily L Dennis
- Brigham and Women's Hospital/Harvard University and Department of Psychology, Stanford University, 1249 Boylston Street, Boston, MA 02215, United States of America.
| | - Robert F Asarnow
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Box 951759, 760 Westwood Plaza, 48-240C Semel Institute, Los Angeles, CA 90095-1759, United States of America.
| | - David A Hovda
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Department of Medical and Molecular Pharmacology, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562 & Semel 18-228A, Los Angeles, CA 90095-6901, United States of America.
| | - Neil G Harris
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States of America.
| | - Christopher C Giza
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Steve Tisch BrainSPORT Program, University of California at Los Angeles, Los Angeles, CA, United States of America; Division of Pediatric Neurology, Mattel Children's Hospital - UCLA, Los Angeles, CA, United States of America.
| |
Collapse
|
34
|
Chitturi J, Santhakumar V, Kannurpatti SS. Beneficial Effects of Kaempferol after Developmental Traumatic Brain Injury Is through Protection of Mitochondrial Function, Oxidative Metabolism, and Neural Viability. J Neurotrauma 2019; 36:1264-1278. [PMID: 30430900 PMCID: PMC6479259 DOI: 10.1089/neu.2018.6100] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Oxidative energy metabolism is depressed after mild/moderate traumatic brain injury (TBI) during early development, accompanied by behavioral debilitation and secondary neuronal death. A TBI metabolome analysis revealed broad effects with a striking impact on energy metabolism. Our studies on mitochondrial modulators and their effects on brain function have shown that kaempferol, a stimulator of the mitochondrial Ca2+ uniporter channel (mCU), enhanced neural and neurovascular activity in the normal brain and improved stimulus-induced brain activation and behavior after TBI during early development. Because kaempferol enhances mitochondrial Ca2+ uptake and cycling, with protective effects after TBI, we tested the hypothesis that kaempferol treatment during the acute/subacute stage after TBI (0-72 h) acted on mitochondria in improving TBI outcome. Developmental age rats (P31) underwent TBI and were treated with vehicle or kaempferol (1 mg/kg intraperitoneally) in three doses at 1, 24, and 48 h after TBI. Brains were harvested at 72 h and subjected to liquid chromatography mass spectrometric measurements. Decrease in pyruvate and tricarboxylic acid (TCA) cycle flux were observed in the untreated and vehicle-treated group, consistent with previously established energy metabolic decline after TBI. Kaempferol improved TCA cycle flux, maintained mitochondrial functional integrity as observed by decreased acyl carnitines, improved neural viability as evidenced by higher N-acetyl aspartate levels. The positive outcomes of kaempferol on metabolic profile corresponded with improved sensorimotor behavior.
Collapse
Affiliation(s)
- Jyothsna Chitturi
- Department of Radiology, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey
- Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California
| | | |
Collapse
|
35
|
Maynard ME, Underwood EL, Redell JB, Zhao J, Kobori N, Hood KN, Moore AN, Dash PK. Carnosic Acid Improves Outcome after Repetitive Mild Traumatic Brain Injury. J Neurotrauma 2019; 36:2147-2152. [PMID: 30672378 DOI: 10.1089/neu.2018.6155] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In the majority of cases, the cognitive and behavioral impairments resulting from a mild traumatic brain injury (TBI) (also referred to as concussion) wane within days to weeks. In contrast, these impairments can persist for months to years after repetitive mild TBI (rmTBI). The cellular and molecular mechanisms underlying these impairments are not well understood. In the present study, we examined the consequences of rmTBI (three weight drops each separated by 72 h) on brain tissue respiration, pathology, and cognitive performance in mice. The transcription factor nuclear factor-erythroid 2-realted factor 2 (Nrf2) has been demonstrated to enhance the expression of numerous cytoprotective genes. Carnosic acid (CA) has been shown to activate Nrf2 and suppress the proinflammatory transcription factor nuclear factor kappa B (NF-κB). Because contemporaneous activation of cytoprotective genes and inhibition of proinflammatory genes can be beneficial, we questioned whether CA can be used to mitigate the pathobiology of rmTBI. The rmTBI increased hippocampal adenosine triphosphate-linked tissue respiration and proton leak that were unaffected by CA treatment. The rmTBI also caused significant motor and cognitive dysfunction, as tested using the foot fault, Barnes maze, and novel object recognition tasks. These impairments occurred in the absence of visible neuronal or dendritic loss. Post-rmTBI administration of CA significantly improved motor and cognitive function, and decreased Gfap and Iba1 immunoreactivities within white matter tracks. Taken together, these results show that rmTBI can cause cognitive impairments in the absence of overt neuronal pathologies, and post-injury treatment with CA can lessen some of these impairments.
Collapse
Affiliation(s)
- Mark E Maynard
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Erica L Underwood
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - John B Redell
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Jing Zhao
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Nobuhide Kobori
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Kimberly N Hood
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| |
Collapse
|
36
|
Hubbard WB, Joseph B, Spry M, Vekaria HJ, Saatman KE, Sullivan PG. Acute Mitochondrial Impairment Underlies Prolonged Cellular Dysfunction after Repeated Mild Traumatic Brain Injuries. J Neurotrauma 2018; 36:1252-1263. [PMID: 30417732 DOI: 10.1089/neu.2018.5990] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mild traumatic brain injuries (mTBI), accounting for more than 80% of TBIs, can cause cognitive and behavioral impairments, the severity and duration of which increase after additional mTBIs. While mTBI does not cause widespread neuronal death, the mechanisms underlying increased cellular susceptibility to subsequent head impacts remain unknown. To investigate the hypothesis that altered mitochondrial bioenergetics underlie cellular vulnerability to repeated insults, we employed a mouse model of mild closed head injury (CHI) to examine mitochondrial function and oxidative stress, because these mechanisms are often intertwined. Mitochondrial respiration was assayed (Seahorse XFe24 Flux Analyzer) from cortex and hippocampus collected at 6 h, 24 h, 48 h, and 96 h post-injury. State III (adenosine diphosphate [ADP]-mediated) respiration was significantly decreased in the hippocampal mitochondria of the CHI group compared with sham at 48 h post-injury. Further, cortex-derived mitochondria exhibited a decrease in State III respiration at 24 h and 48 h post-injury. No significant differences were observed at 6 h or 96 h post-injury in either region of interest. A second CHI repeated either 48 h or 96 h after the first did not worsen State III respiration at 48 h after the final injury compared with a single CHI, but CHI repeated at a 48 h interval prolonged cortical mitochondrial dysfunction to 96 h after the final injury. Markers of oxidative stress were significantly elevated after two CHIs delivered 48 h apart, but not after single CHI or two CHI delivered 96 h apart. This study establishes that mTBI results in early mitochondrial dysfunction, which may be a determinant for cellular vulnerability to repeated head impacts. Thus, therapies targeting mitochondrial impairment could improve outcomes after repeated mTBI.
Collapse
Affiliation(s)
- W Brad Hubbard
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Binoy Joseph
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Malinda Spry
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Hemendra J Vekaria
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kathryn E Saatman
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky.,2 Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Patrick G Sullivan
- 1 Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky.,3 Department of Neuroscience, University of Kentucky, Lexington, Kentucky.,4 Lexington VAMC, Lexington, Kentucky
| |
Collapse
|