51
|
Menzel L, Kleber L, Friedrich C, Hummel R, Dangel L, Winter J, Schmitz K, Tegeder I, Schäfer MKE. Progranulin protects against exaggerated axonal injury and astrogliosis following traumatic brain injury. Glia 2016; 65:278-292. [DOI: 10.1002/glia.23091] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 10/04/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Lutz Menzel
- Department of Anesthesiology; University Medical Center, Johannes Gutenberg-University, Mainz; Germany
| | - Lisa Kleber
- Department of Anesthesiology; University Medical Center, Johannes Gutenberg-University, Mainz; Germany
| | - Carina Friedrich
- Department of Anesthesiology; University Medical Center, Johannes Gutenberg-University, Mainz; Germany
| | - Regina Hummel
- Department of Anesthesiology; University Medical Center, Johannes Gutenberg-University, Mainz; Germany
| | - Larissa Dangel
- Department of Anesthesiology; University Medical Center, Johannes Gutenberg-University, Mainz; Germany
| | - Jennifer Winter
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg-University, Mainz; Germany
- Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University, Mainz; Germany
| | - Katja Schmitz
- Clinical Pharmacology; Goethe-University Hospital; Frankfurt Germany
| | - Irmgard Tegeder
- Clinical Pharmacology; Goethe-University Hospital; Frankfurt Germany
| | - Michael K. E. Schäfer
- Department of Anesthesiology; University Medical Center, Johannes Gutenberg-University, Mainz; Germany
- Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University, Mainz; Germany
| |
Collapse
|
52
|
Chiu CC, Liao YE, Yang LY, Wang JY, Tweedie D, Karnati HK, Greig NH, Wang JY. Neuroinflammation in animal models of traumatic brain injury. J Neurosci Methods 2016; 272:38-49. [PMID: 27382003 PMCID: PMC5201203 DOI: 10.1016/j.jneumeth.2016.06.018] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and morbidity worldwide. Neuroinflammation is prominent in the short and long-term consequences of neuronal injuries that occur after TBI. Neuroinflammation involves the activation of glia, including microglia and astrocytes, to release inflammatory mediators within the brain, and the subsequent recruitment of peripheral immune cells. Various animal models of TBI have been developed that have proved valuable to elucidate the pathophysiology of the disorder and to assess the safety and efficacy of novel therapies prior to clinical trials. These models provide an excellent platform to delineate key injury mechanisms that associate with types of injury (concussion, contusion, and penetration injuries) that occur clinically for the investigation of mild, moderate, and severe forms of TBI. Additionally, TBI modeling in genetically engineered mice, in particular, has aided the identification of key molecules and pathways for putative injury mechanisms, as targets for development of novel therapies for human TBI. This Review details the evidence showing that neuroinflammation, characterized by the activation of microglia and astrocytes and elevated production of inflammatory mediators, is a critical process occurring in various TBI animal models, provides a broad overview of commonly used animal models of TBI, and overviews representative techniques to quantify markers of the brain inflammatory process. A better understanding of neuroinflammation could open therapeutic avenues for abrogation of secondary cell death and behavioral symptoms that may mediate the progression of TBI.
Collapse
Affiliation(s)
- Chong-Chi Chiu
- Department of General Surgery, Chi Mei Medical Center, Tainan and Liouying, Taiwan
| | - Yi-En Liao
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Ling-Yu Yang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ya Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Hanuma K Karnati
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
53
|
Zhang Z, Bassam B, Thomas AG, Williams M, Liu J, Nance E, Rojas C, Slusher BS, Kannan S. Maternal inflammation leads to impaired glutamate homeostasis and up-regulation of glutamate carboxypeptidase II in activated microglia in the fetal/newborn rabbit brain. Neurobiol Dis 2016; 94:116-28. [PMID: 27326668 PMCID: PMC5394739 DOI: 10.1016/j.nbd.2016.06.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 06/05/2016] [Accepted: 06/16/2016] [Indexed: 12/12/2022] Open
Abstract
Astrocyte dysfunction and excessive activation of glutamatergic systems have been implicated in a number of neurologic disorders, including periventricular leukomalacia (PVL) and cerebral palsy (CP). However, the role of chorioamnionitis on glutamate homeostasis in the fetal and neonatal brains is not clearly understood. We have previously shown that intrauterine endotoxin administration results in intense microglial 'activation' and increased pro-inflammatory cytokines in the periventricular region (PVR) of the neonatal rabbit brain. In this study, we assessed the effect of maternal inflammation on key components of the glutamate pathway and its relationship to astrocyte and microglial activation in the fetal and neonatal New Zealand white rabbit brain. We found that intrauterine endotoxin exposure at gestational day 28 (G28) induced acute and prolonged glutamate elevation in the PVR of fetal (G29, 1day post-injury) and postnatal day 1 (PND1, 3days post-injury) brains along with prominent morphological changes in the astrocytes (soma hypertrophy and retracted processes) in the white matter tracts. There was a significant increase in glutaminase and N-Methyl-d-Aspartate receptor (NMDAR) NR2 subunit expression along with decreased glial L-glutamate transporter 1 (GLT-1) in the PVR at G29, that would promote acute dysregulation of glutamate homeostasis. This was accompanied with significantly decreased TGF-β1 at PND1 in CP kits indicating ongoing neuroinflammation. We also show for the first time that glutamate carboxypeptidase II (GCPII) was significantly increased in the activated microglia at the periventricular white matter area in both G29 and PND1 CP kits. This was confirmed by in vitro studies demonstrating that LPS activated primary microglia markedly upregulate GCPII enzymatic activity. These results suggest that maternal intrauterine endotoxin exposure results in early onset and long-lasting dysregulation of glutamate homeostasis, which may be mediated by impaired astrocyte function and GCPII upregulation in activated microglia.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Bassam Bassam
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Monica Williams
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Jinhuan Liu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Elizabeth Nance
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Barbara S Slusher
- Neurology, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA; Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA.
| |
Collapse
|
54
|
Wang ML, Li WB. Cognitive impairment after traumatic brain injury: The role of MRI and possible pathological basis. J Neurol Sci 2016; 370:244-250. [PMID: 27772768 DOI: 10.1016/j.jns.2016.09.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 09/01/2016] [Accepted: 09/23/2016] [Indexed: 01/26/2023]
Abstract
Traumatic brain injury (TBI) is closely related to increased incidence of cognitive impairment from the acute phase to chronic phase. At present, the pathological mechanism leading to cognitive impairment after TBI is still not fully understood. We hypothesize that neuron loss, diffuse axonal injury, microbleed, and blood-brain barrier (BBB) disruption altogether contribute to the development of cognitive impairment. Furthermore, the disruption of structural and functional neural network related to the cognitive function might bring about the final step in the occurrence of cognitive impairment after TBI. In this review, we summarize the role of different MRI techniques in the assessment of the pathological changes related to cognitive impairment after TBI. These MRI techniques include T1-MPRAGE sequence reflecting neuron loss, diffusion tensor imaging reflecting diffuse axonal injury, diffusion kurtosis imaging reflecting diffuse axonal injury and reactive gliosis, susceptibility weighted imaging showing microbleed, arterial spin labeling showing blood flow and dynamic contrast enhanced MRI showing BBB disruption. In the future, correlational study of multi-MRI sequences scan, pathological examination, and cognitive tests will provide valuable information for understanding the mechanism of cognitive impairment after TBI and manage TBI patients.
Collapse
Affiliation(s)
- Ming-Liang Wang
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Wen-Bin Li
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Imaging center, Kashgar Prefecture Second People(')s Hospital, Kashgar 844000, China.
| |
Collapse
|
55
|
Abstract
There is a paucity of accurate and reliable biomarkers to detect traumatic brain injury, grade its severity, and model post-traumatic brain injury (TBI) recovery. This gap could be addressed via advances in brain mapping which define injury signatures and enable tracking of post-injury trajectories at the individual level. Mapping of molecular and anatomical changes and of modifications in functional activation supports the conceptual paradigm of TBI as a disorder of large-scale neural connectivity. Imaging approaches with particular relevance are magnetic resonance techniques (diffusion weighted imaging, diffusion tensor imaging, susceptibility weighted imaging, magnetic resonance spectroscopy, functional magnetic resonance imaging, and positron emission tomographic methods including molecular neuroimaging). Inferences from mapping represent unique endophenotypes which have the potential to transform classification and treatment of patients with TBI. Limitations of these methods, as well as future research directions, are highlighted.
Collapse
|
56
|
Reeves TM, Trimmer PA, Colley BS, Phillips LL. Targeting Kv1.3 channels to reduce white matter pathology after traumatic brain injury. Exp Neurol 2016; 283:188-203. [PMID: 27302680 PMCID: PMC4992637 DOI: 10.1016/j.expneurol.2016.06.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/31/2016] [Accepted: 06/10/2016] [Indexed: 02/07/2023]
Abstract
Axonal injury is present in essentially all clinically significant cases of traumatic brain injury (TBI). While no effective treatment has been identified to date, experimental TBI models have shown promising axonal protection using immunosuppressants FK506 and Cyclosporine-A, with treatment benefits attributed to calcineurin inhibition or protection of mitochondrial function. However, growing evidence suggests neuroprotective efficacy of these compounds may also involve direct modulation of ion channels, and in particular Kv1.3. The present study tested whether blockade of Kv1.3 channels, using Clofazimine (CFZ), would alleviate TBI-induced white matter pathology in rodents. Postinjury CFZ administration prevented suppression of compound action potential (CAP) amplitude in the corpus callosum of adult rats following midline fluid percussion TBI, with injury and treatment effects primarily expressed in unmyelinated CAPs. Kv1.3 protein levels in callosal tissue extracts were significantly reduced postinjury, but this loss was prevented by CFZ treatment. In parallel, CFZ also attenuated the injury-induced elevation in pro-inflammatory cytokine IL1-β. The effects of CFZ on glial function were further studied using mixed microglia/astrocyte cell cultures derived from P3-5 mouse corpus callosum. Cultures of callosal glia challenged with lipopolysaccharide exhibited a dramatic increase in IL1-β levels, accompanied by reactive morphological changes in microglia, both of which were attenuated by CFZ treatment. These results support a cell specific role for Kv1.3 signaling in white matter pathology after TBI, and suggest a treatment approach based on the blockade of these channels. This therapeutic strategy may be especially efficacious for normalizing neuro-glial interactions affecting unmyelinated axons after TBI.
Collapse
Affiliation(s)
- Thomas M Reeves
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, United States
| | - Patricia A Trimmer
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, United States
| | - Beverly S Colley
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, United States
| | - Linda L Phillips
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, United States
| |
Collapse
|
57
|
Connectome-scale assessment of structural and functional connectivity in mild traumatic brain injury at the acute stage. NEUROIMAGE-CLINICAL 2016; 12:100-115. [PMID: 27408795 PMCID: PMC4932612 DOI: 10.1016/j.nicl.2016.06.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 06/08/2016] [Accepted: 06/14/2016] [Indexed: 01/16/2023]
Abstract
Mild traumatic brain injury (mTBI) accounts for over one million emergency visits each year in the United States. The large-scale structural and functional network connectivity changes of mTBI are still unknown. This study was designed to determine the connectome-scale brain network connectivity changes in mTBI at both structural and functional levels. 40 mTBI patients at the acute stage and 50 healthy controls were recruited. A novel approach called Dense Individualized and Common Connectivity-based Cortical Landmarks (DICCCOLs) was applied for connectome-scale analysis of both diffusion tensor imaging and resting state functional MRI data. Among 358 networks identified on DICCCOL analysis, 41 networks were identified as structurally discrepant between patient and control groups. The involved major white matter tracts include the corpus callosum, and superior and inferior longitudinal fasciculi. Functional connectivity analysis identified 60 connectomic signatures that differentiate patients from controls with 93.75% sensitivity and 100% specificity. Analysis of functional domains showed decreased intra-network connectivity within the emotion network and among emotion-cognition interactions, and increased interactions among action-emotion and action-cognition as well as within perception networks. This work suggests that mTBI may result in changes of structural and functional connectivity on a connectome scale at the acute stage.
Collapse
|
58
|
Muroi C, Kashiwagi Y, Rokugawa T, Tonomura M, Obata A, Nevzati E, Tsuboi A, Okuchi K, Mishima K, Abe K, Fujioka M. Evaluation of a filament perforation model for mouse subarachnoid hemorrhage using 7.0 Tesla MRI. J Clin Neurosci 2016; 28:141-7. [PMID: 27021225 DOI: 10.1016/j.jocn.2015.10.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 10/12/2015] [Accepted: 10/25/2015] [Indexed: 10/22/2022]
Abstract
The filament perforation model (FPM) in mice is becoming increasingly popular to elucidate the molecular pathogenesis of neuronal injury after subarachnoid hemorrhage (SAH). We evaluated brain MRI in a mouse FPM. A total of 28 male C57Bl/6J mice were used. Seventeen animals underwent SAH induction by FPM. In two animals, transient middle cerebral artery occlusion (MCAo) was induced. Nine mice served as controls. T1-weighted images (T1WI), T2-weighted images (T2WI), T2(∗)-weighted images (T2*WI) and apparent diffusion coefficient maps were acquired at day 0 and at various time points following SAH (range: day 1-6 after SAH). Cerebral blood flow (CBF) analysis by (14)C-iodoamphetamine ((14)C-IMP) autoradiography was conducted in nine animals. Hemorrhage could be best confirmed using T2*WI. The degree of hemorrhage varied. All animals evaluated for ⩾2days were hydrocephalic, which was best seen on T2WI. T2-hyperintensity of the corpus callosum and external capsule, indicating white matter (WM) injury, was present after SAH. Ventricle and WM injury volumes were statistically significantly higher at day 3 compared to day 0. Territorial ischemia was detectable in MCAo but not in SAH. Markedly hypointense cortical veins were visible in the hyperacute and delayed phase after SAH on T2*WI. The (14)C-IMP analysis indicated decreased CBF after SAH. MRI is feasible and useful in evaluating pathophysiological changes over time. T2*WI seems best for SAH detection and grading. The chronological change of hydrocephalus and WM injury could be analyzed. T2*WI illustrated specific signal changes of cortical veins, possibly caused by increased oxygen extraction fraction due to decreased CBF.
Collapse
Affiliation(s)
- Carl Muroi
- Institute of Aging and Brain Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan; Department of Neurosurgery, Kantonsspital Aarau, Aarau, Switzerland.
| | - Yuto Kashiwagi
- Department of Drug Metabolism and Pharmacokinetics, Research Laboratory for Development, Shionogi & Co, Osaka, Japan
| | - Takemi Rokugawa
- Department of Drug Metabolism and Pharmacokinetics, Research Laboratory for Development, Shionogi & Co, Osaka, Japan
| | - Misato Tonomura
- Department of Drug Metabolism and Pharmacokinetics, Research Laboratory for Development, Shionogi & Co, Osaka, Japan
| | - Atsushi Obata
- Department of Drug Metabolism and Pharmacokinetics, Research Laboratory for Development, Shionogi & Co, Osaka, Japan
| | - Edin Nevzati
- Department of Neurosurgery, Kantonsspital Aarau, Aarau, Switzerland
| | - Akio Tsuboi
- Laboratory for Molecular Biology of Neural System, Advanced Medical Research Center, Nara Medical University, Kashihara, Nara, Japan
| | - Kazuo Okuchi
- Department of Emergency and Critical Care Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Kenichi Mishima
- Institute of Aging and Brain Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kohji Abe
- Department of Drug Metabolism and Pharmacokinetics, Research Laboratory for Development, Shionogi & Co, Osaka, Japan
| | - Masayuki Fujioka
- Institute of Aging and Brain Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan; Laboratory for Molecular Biology of Neural System, Advanced Medical Research Center, Nara Medical University, Kashihara, Nara, Japan; Department of Emergency and Critical Care Medicine, Nara Medical University, Kashihara, Nara, Japan; Department of Neurosurgery, Saiseikai Senri Hospital, Suita, Osaka, Japan
| |
Collapse
|
59
|
Vállez García D, Otte A, Dierckx RAJO, Doorduin J. Three Month Follow-Up of Rat Mild Traumatic Brain Injury: A Combined [ 18F]FDG and [ 11C]PK11195 Positron Emission Study. J Neurotrauma 2016; 33:1855-1865. [PMID: 26756169 DOI: 10.1089/neu.2015.4230] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is the most common cause of head trauma. The time course of functional pathology is not well defined, however. The purpose of this study was to evaluate the consequences of mTBI in rats over a period of 3 months by determining the presence of neuroinflammation ([11C]PK11195) and changes in brain metabolism ([18F]FDG) with positron emission tomography (PET) imaging. Male Sprague-Dawley rats were divided in mTBI (n = 8) and sham (n = 8) groups. In vivo PET imaging and behavioral tests (open field, object recognition, and Y-maze) were performed at different time points after induction of the trauma. Differences between groups in PET images were explored using volume-of-interest and voxel-based analysis. mTBI did not result in death, skull fracture, or suppression of reflexes. Weight gain was reduced (p = 0.003) in the mTBI group compared with the sham-treated group. No statistical differences were found in the behavioral tests at any time point. Volume-of-interest analysis showed neuroinflammation limited to the subacute phase (day 12) involving amygdala, globus pallidus, hypothalamus, pons, septum, striatum, and thalamus (p < 0.03, d > 1.2). Alterations in glucose metabolism were detected over the 3 month period, with increased uptake in the medulla (p < 0.04, d ≥ 1.2), and decreased uptake in the globus pallidus, striatum, and thalamus (p < 0.04, d ≤ 1.2). Similar findings were observed in the voxel-based analysis (p < 0.05 at corrected cluster level). As a consequence of the mTBI, and in the absence of apparent behavioral alterations, relative brain glucose metabolism was found altered in several brain regions, which mostly correspond with those presenting neuroinflammation in the subacute stage.
Collapse
Affiliation(s)
- David Vállez García
- 1 Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| | - Andreas Otte
- 2 Division of Biomedical Engineering, Department of Electrical Engineering and Information Technology, Offenburg University , Offenburg, Germany
| | - Rudi A J O Dierckx
- 1 Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| | - Janine Doorduin
- 1 Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| |
Collapse
|
60
|
Tajiri N, De La Peña I, Acosta SA, Kaneko Y, Tamir S, Landesman Y, Carlson R, Shacham S, Borlongan CV. A Nuclear Attack on Traumatic Brain Injury: Sequestration of Cell Death in the Nucleus. CNS Neurosci Ther 2016; 22:306-15. [PMID: 26842647 PMCID: PMC5067638 DOI: 10.1111/cns.12501] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/25/2015] [Accepted: 11/27/2015] [Indexed: 11/28/2022] Open
Abstract
Background Exportin 1 (XPO1/CRM1) plays prominent roles in the regulation of nuclear protein export. Selective inhibitors of nuclear export (SINE) are small orally bioavailable molecules that serve as drug‐like inhibitors of XPO1, with potent anti‐cancer properties. Traumatic brain injury (TBI) presents with a secondary cell death characterized by neuroinflammation that is putatively regulated by nuclear receptors. Aims and Results Here, we report that the SINE compounds (KPT‐350 or KPT‐335) sequestered TBI‐induced neuroinflammation‐related proteins (NF‐kB, AKT, FOXP1) within the nucleus of cultured primary rat cortical neurons, which coincided with protection against TNF‐α (20 ng/mL)‐induced neurotoxicity as shown by at least 50% and 100% increments in preservation of cell viability and cellular enzymatic activity, respectively, compared to non‐treated neuronal cells (P's < 0.05). In parallel, using an in vivo controlled cortical impact (CCI) model of TBI, we demonstrate that adult Sprague‐Dawley rats treated post‐injury with SINE compounds exhibited significant reductions in TBI‐induced behavioral and histological deficits. Animals that received KPT‐350 orally starting at 2 h post‐TBI and once a day thereafter over the next 4 days exhibited significantly better motor coordination, and balance in the rotorod test and motor asymmetry test by 100–200% improvements, as early as 4 h after initial SINE compound injection that was sustained during subsequent KPT‐350 dosing, and throughout the 18‐day post‐TBI study period compared to vehicle treatment (P's < 0.05). Moreover, KPT‐350 reduced cortical core impact area and peri‐impact cell death compared to vehicle treatment (P's < 0.05). Conclusions Both in vitro and in vivo experiments revealed that KPT‐350 increased XPO1, AKT, and FOXP1 nuclear expression and relegated NF‐kB expression within the neuronal nuclei. Altogether, these findings advance the utility of SINE compounds to stop trafficking of cell death proteins within the nucleus as an efficacious treatment for TBI.
Collapse
Affiliation(s)
- Naoki Tajiri
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Ike De La Peña
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sandra A Acosta
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | | | | | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
61
|
Regulating effect of activated NF-κB on edema induced by traumatic brain injury of rats. ASIAN PAC J TROP MED 2016; 9:274-7. [PMID: 26972401 DOI: 10.1016/j.apjtm.2016.01.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 12/20/2015] [Accepted: 12/30/2015] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To observe the effect of nuclear transcription factor-κB (NF-κB) on cerebral edema in rats with traumatic brain injury (TBI). METHODS Male SD rats with fluid percussion injury (FPI) were selected. After separation and culture, rats' astrocytes all suffered FPI. The expression of NF-κB and the water content were detected at the animal and cellular levels, while the activity of NOX was evaluated at the cellular level. RESULTS According to the results, the positive expression of NF-κB and expression of mRNA were significantly increased and the water content was increased for rats after TBI, while NF-κB inhibitor BAY11-7082 could significantly reduce the effect of TBI. 1 and 3 h after FPI of astrocytes, the activation of NF-κB was increased and BAY 11-7082 could significantly improve the injury-induced swelling of astrocytes. After the injury of astrocytes, the activity of NOX was also increased, while BAY 11-7082 could reduce the activity of NOX. CONCLUSIONS The results show that the activation of NF-κB in astrocytes is a key factor in the process of cerebral edema after TBI of rats.
Collapse
|
62
|
Bailey ZS, Hubbard WB, VandeVord PJ. Cellular Mechanisms and Behavioral Outcomes in Blast-Induced Neurotrauma: Comparing Experimental Setups. Methods Mol Biol 2016; 1462:119-138. [PMID: 27604716 DOI: 10.1007/978-1-4939-3816-2_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Blast-induced neurotrauma (BINT) has increased in incidence over the past decades and can result in cognitive issues that have debilitating consequences. The exact primary and secondary mechanisms of injury have not been elucidated and appearance of cellular injury can vary based on many factors, such as blast overpressure magnitude and duration. Many methodologies to study blast neurotrauma have been employed, ranging from open-field explosives to experimental shock tubes for producing free-field blast waves. While there are benefits to the various methods, certain specifications need to be accounted for in order to properly examine BINT. Primary cell injury mechanisms, occurring as a direct result of the blast wave, have been identified in several studies and include cerebral vascular damage, blood-brain barrier disruption, axonal injury, and cytoskeletal damage. Secondary cell injury mechanisms, triggered subsequent to the initial insult, result in the activation of several molecular cascades and can include, but are not limited to, neuroinflammation and oxidative stress. The collective result of these secondary injuries can lead to functional deficits. Behavioral measures examining motor function, anxiety traits, and cognition/memory problems have been utilized to determine the level of injury severity. While cellular injury mechanisms have been identified following blast exposure, the various experimental models present both concurrent and conflicting results. Furthermore, the temporal response and progression of pathology after blast exposure have yet to be detailed and remain unclear due to limited resemblance of methodologies. This chapter summarizes the current state of blast neuropathology and emphasizes the need for a standardized preclinical model of blast neurotrauma.
Collapse
Affiliation(s)
- Zachary S Bailey
- School of Biomedical Engineering and Sciences, Virginia Tech, 447 Kelly Hall, 325 Stanger Street, Blacksburg, VA, 24061, USA
| | - W Brad Hubbard
- School of Biomedical Engineering and Sciences, Virginia Tech, 447 Kelly Hall, 325 Stanger Street, Blacksburg, VA, 24061, USA
| | - Pamela J VandeVord
- School of Biomedical Engineering and Sciences, Virginia Tech, 447 Kelly Hall, 325 Stanger Street, Blacksburg, VA, 24061, USA.
| |
Collapse
|
63
|
Koutsoudaki PN, Papastefanaki F, Stamatakis A, Kouroupi G, Xingi E, Stylianopoulou F, Matsas R. Neural stem/progenitor cells differentiate into oligodendrocytes, reduce inflammation, and ameliorate learning deficits after transplantation in a mouse model of traumatic brain injury. Glia 2015; 64:763-79. [PMID: 26712314 DOI: 10.1002/glia.22959] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 11/30/2015] [Accepted: 12/03/2015] [Indexed: 01/09/2023]
Abstract
The central nervous system has limited capacity for regeneration after traumatic injury. Transplantation of neural stem/progenitor cells (NPCs) has been proposed as a potential therapeutic approach while insulin-like growth factor I (IGF-I) has neuroprotective properties following various experimental insults to the nervous system. We have previously shown that NPCs transduced with a lentiviral vector for IGF-I overexpression have an enhanced ability to give rise to neurons in vitro but also in vivo, upon transplantation in a mouse model of temporal lobe epilepsy. Here we studied the regenerative potential of NPCs, IGF-I-transduced or not, in a mouse model of hippocampal mechanical injury. NPC transplantation, with or without IGF-I transduction, rescued the injury-induced spatial learning deficits as revealed in the Morris Water Maze. Moreover, it had beneficial effects on the host tissue by reducing astroglial activation and microglial/macrophage accumulation while enhancing generation of endogenous oligodendrocyte precursor cells. One or two months after transplantation the grafted NPCs had migrated towards the lesion site and in the neighboring myelin-rich regions. Transplanted cells differentiated toward the oligodendroglial, but not the neuronal or astrocytic lineages, expressing the early and late oligodendrocyte markers NG2, Olig2, and CNPase. The newly generated oligodendrocytes reached maturity and formed myelin internodes. Our current and previous observations illustrate the high plasticity of transplanted NPCs which can acquire injury-dependent phenotypes within the host CNS, supporting the fact that reciprocal interactions between transplanted cells and the host tissue are an important factor to be considered when designing prospective cell-based therapies for CNS degenerative conditions.
Collapse
Affiliation(s)
- Paraskevi N Koutsoudaki
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, 11521, Greece
| | - Florentia Papastefanaki
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, 11521, Greece
| | - Antonios Stamatakis
- Biology-Biochemistry Laboratory, Department of Basic Sciences, Faculty of Nursing, University of Athens, Athens, 11527, Greece
| | - Georgia Kouroupi
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, 11521, Greece
| | - Evangelia Xingi
- Light Microscopy Unit, Hellenic Pasteur Institute, Athens, 11521, Greece
| | - Fotini Stylianopoulou
- Biology-Biochemistry Laboratory, Department of Basic Sciences, Faculty of Nursing, University of Athens, Athens, 11527, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, 11521, Greece
| |
Collapse
|
64
|
Compensation through Functional Hyperconnectivity: A Longitudinal Connectome Assessment of Mild Traumatic Brain Injury. Neural Plast 2015; 2016:4072402. [PMID: 26819765 PMCID: PMC4706919 DOI: 10.1155/2016/4072402] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/07/2015] [Accepted: 08/11/2015] [Indexed: 11/18/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is a major public health concern. Functional MRI has reported alterations in several brain networks following mTBI. However, the connectome-scale brain network changes are still unknown. In this study, sixteen mTBI patients were prospectively recruited from an emergency department and followed up at 4-6 weeks after injury. Twenty-four healthy controls were also scanned twice with the same time interval. Three hundred fifty-eight brain landmarks that preserve structural and functional correspondence of brain networks across individuals were used to investigate longitudinal brain connectivity. Network-based statistic (NBS) analysis did not find significant difference in the group-by-time interaction and time effects. However, 258 functional pairs show group differences in which mTBI patients have higher functional connectivity. Meta-analysis showed that "Action" and "Cognition" are the most affected functional domains. Categorization of connectomic signatures using multiview group-wise cluster analysis identified two patterns of functional hyperconnectivity among mTBI patients: (I) between the posterior cingulate cortex and the association areas of the brain and (II) between the occipital and the frontal lobes of the brain. Our results demonstrate that brain concussion renders connectome-scale brain network connectivity changes, and the brain tends to be hyperactivated to compensate the pathophysiological disturbances.
Collapse
|
65
|
Lucke-Wold BP, Logsdon AF, Smith KE, Turner RC, Alkon DL, Tan Z, Naser ZJ, Knotts CM, Huber JD, Rosen CL. Bryostatin-1 Restores Blood Brain Barrier Integrity following Blast-Induced Traumatic Brain Injury. Mol Neurobiol 2015; 52:1119-1134. [PMID: 25301233 PMCID: PMC5000781 DOI: 10.1007/s12035-014-8902-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/24/2014] [Indexed: 02/08/2023]
Abstract
Recent wars in Iraq and Afghanistan have accounted for an estimated 270,000 blast exposures among military personnel. Blast traumatic brain injury (TBI) is the 'signature injury' of modern warfare. Blood brain barrier (BBB) disruption following blast TBI can lead to long-term and diffuse neuroinflammation. In this study, we investigate for the first time the role of bryostatin-1, a specific protein kinase C (PKC) modulator, in ameliorating BBB breakdown. Thirty seven Sprague-Dawley rats were used for this study. We utilized a clinically relevant and validated blast model to expose animals to moderate blast exposure. Groups included: control, single blast exposure, and single blast exposure + bryostatin-1. Bryostatin-1 was administered i.p. 2.5 mg/kg after blast exposure. Evan's blue, immunohistochemistry, and western blot analysis were performed to assess injury. Evan's blue binds to albumin and is a marker for BBB disruption. The single blast exposure caused an increase in permeability compared to control (t = 4.808, p < 0.05), and a reduction back toward control levels when bryostatin-1 was administered (t = 5.113, p < 0.01). Three important PKC isozymes, PKCα, PKCδ, and PKCε, were co-localized primarily with endothelial cells but not astrocytes. Bryostatin-1 administration reduced toxic PKCα levels back toward control levels (t = 4.559, p < 0.01) and increased the neuroprotective isozyme PKCε (t = 6.102, p < 0.01). Bryostatin-1 caused a significant increase in the tight junction proteins VE-cadherin, ZO-1, and occludin through modulation of PKC activity. Bryostatin-1 ultimately decreased BBB breakdown potentially due to modulation of PKC isozymes. Future work will examine the role of bryostatin-1 in preventing chronic neurodegeneration following repetitive neurotrauma.
Collapse
Affiliation(s)
- Brandon P Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Aric F Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Kelly E Smith
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Ryan C Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Daniel L Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, 26506, USA
| | - Zhenjun Tan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Zachary J Naser
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Office of Professional Studies in Health Sciences, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Chelsea M Knotts
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Jason D Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Charles L Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Department of Neurosurgery, West Virginia University School of Medicine, One Medical Center Drive, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV, 26506-9183, USA.
| |
Collapse
|
66
|
Liu G, Ni J, Mao L, Yan M, Pang T, Liao H. Expression of Nogo receptor 1 in microglia during development and following traumatic brain injury. Brain Res 2015; 1627:41-51. [PMID: 26367446 DOI: 10.1016/j.brainres.2015.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/26/2015] [Accepted: 09/05/2015] [Indexed: 01/18/2023]
Abstract
As the receptor of myelin associated inhibitory factors Nogo receptor 1 (NgR1) plays an important role in central nervous system (CNS) injury and regeneration. It is found that NgR1 complex acts in neurons to transduce the signals intracelluarly including induction of growth cone collapse, inhibition of axonal regeneration and regulation of nerve inflammation. In recent studies, NgR1 has also been found to be expressed in the microglia. However, NgR1 expressed in microglia in the developing nervous systems and following CNS injury have not been widely investigated. In this study, we detected the expression and cellular localization of NgR1 in microglia during development and following traumatic brain injury (TBI) in mice. The results showed that NgR1 was mainly expressed in microglia during embryonic and postnatal periods. The expression levels peaked at P4 and decreased thereafter into adulthood, while increased significantly with aging representatively at 17 mo. On the other hand, there was no significant difference in the number of double positive NgR1(+)Iba1(+) cells between normal and TBI group. In summary, we first detected the expression of NgR1 in microglia during development and found that NgR1 protein expression increased significantly in microglia with aging. These findings will contribute to make a foundation for subsequent study about the role of NgR1 expressed in microglia on the CNS disorders.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Brain/cytology
- Brain/embryology
- Brain/growth & development
- Brain Injuries/pathology
- Calcium-Binding Proteins/metabolism
- Disease Models, Animal
- Embryo, Mammalian
- Functional Laterality
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Expression Regulation, Developmental/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Microfilament Proteins/metabolism
- Microglia/metabolism
- Myelin Proteins/genetics
- Myelin Proteins/metabolism
- Nogo Receptor 1
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
Collapse
Affiliation(s)
- Gaoxiang Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Jie Ni
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Mao
- Department of Neurosurgery, Jinling Hospital, Nanjing 210000, China
| | - Ming Yan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Pang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Liao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
67
|
Zhang B, Wang B, Cao S, Wang Y. Epigallocatechin-3-Gallate (EGCG) Attenuates Traumatic Brain Injury by Inhibition of Edema Formation and Oxidative Stress. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:491-7. [PMID: 26557015 PMCID: PMC4637351 DOI: 10.4196/kjpp.2015.19.6.491] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/12/2015] [Accepted: 05/01/2015] [Indexed: 11/15/2022]
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and long-term disability, which can decrease quality of life. In spite of numerous studies suggesting that Epigallocatechin-3-gallate (EGCG) has been used as a therapeutic agent for a broad range of disorders, the effect of EGCG on TBI remains unknown. In this study, a weight drop model was established to evaluate the therapeutic potential of EGCG on TBI. Rats were administered with 100 mg/kg EGCG or PBS intraperitoneally. At different times following trauma, rats were sacrificed for analysis. It was found that EGCG (100 mg/kg, i.p.) treatment significantly reduced brain water content and vascular permeability at 12, 24, 48, 72 hour after TBI. Real-time PCR results revealed that EGCG inhibited TBI-induced IL-1β and TNF-α mRNA expression. Importantly, CD68 mRNA expression decreasing in the brain suggested that EGCG inhibited microglia activation. Western blotting and immunohistochemistry results showed that administering of EGCG significantly inhibited the levels of aquaporin-4 (AQP4) and glial fibrillary acidic protein (GFAP) expression. TBI-induced oxidative stress was remarkably impaired by EGCG treatment, which elevated the activities of SOD and GSH-PX. Conversely, EGCG significantly reduced the contents of MDA after TBI. In addition, EGCG decreased TBI-induced NADPH oxidase activation through inhibition of p47phox translocation from cytoplasm to plasma membrane. These data demonstrate that EGCG treatment may be an effective therapeutic strategy for TBI and the underlying mechanism involves inhibition of oxidative stress.
Collapse
Affiliation(s)
- Bo Zhang
- Intensive Care Unit, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin 300060, PR China. ; Intensive Care Unit, Tianjin First Center Hospital, Tianjin Institute of Emergency Medicine, Tianjin 300192, PR China
| | - Bing Wang
- Intensive Care Unit, Tianjin First Center Hospital, Tianjin Institute of Emergency Medicine, Tianjin 300192, PR China
| | - Shuhua Cao
- Intensive Care Unit, Tianjin First Center Hospital, Tianjin Institute of Emergency Medicine, Tianjin 300192, PR China
| | - Yongqiang Wang
- Intensive Care Unit, Tianjin First Center Hospital, Tianjin Institute of Emergency Medicine, Tianjin 300192, PR China
| |
Collapse
|
68
|
Ultrastructural Changes in the White and Gray Matter of Mice at Chronic Time Points After Repeated Concussive Head Injury. J Neuropathol Exp Neurol 2015; 74:1012-35. [DOI: 10.1097/nen.0000000000000247] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
69
|
Bergold PJ. Treatment of traumatic brain injury with anti-inflammatory drugs. Exp Neurol 2015; 275 Pt 3:367-380. [PMID: 26112314 DOI: 10.1016/j.expneurol.2015.05.024] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/31/2022]
Abstract
Traumatic brain injury rapidly induces inflammation. This inflammation is produced both by endogenous brain cells and circulating inflammatory cells that enter from the brain. Together they drive the inflammatory response through a wide variety of bioactive lipids, cytokines and chemokines. A large number of drugs with anti-inflammatory action have been tested in both preclinical studies and in clinical trials. These drugs either have known anti-inflammatory action or inhibit the inflammatory response through unknown mechanisms. The results of these preclinical studies and clinical trials are reviewed. Recommendations are suggested on how to improve preclinical testing of drugs to make them more relevant to evaluate for clinical trials.
Collapse
Affiliation(s)
- Peter J Bergold
- Robert F. Furchgott Center for Neural Science, Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, United States.
| |
Collapse
|
70
|
Iraji A, Benson RR, Welch RD, O'Neil BJ, Woodard JL, Ayaz SI, Kulek A, Mika V, Medado P, Soltanian-Zadeh H, Liu T, Haacke EM, Kou Z. Resting State Functional Connectivity in Mild Traumatic Brain Injury at the Acute Stage: Independent Component and Seed-Based Analyses. J Neurotrauma 2015; 32:1031-45. [PMID: 25285363 DOI: 10.1089/neu.2014.3610] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mild traumatic brain injury (mTBI) accounts for more than 1 million emergency visits each year. Most of the injured stay in the emergency department for a few hours and are discharged home without a specific follow-up plan because of their negative clinical structural imaging. Advanced magnetic resonance imaging (MRI), particularly functional MRI (fMRI), has been reported as being sensitive to functional disturbances after brain injury. In this study, a cohort of 12 patients with mTBI were prospectively recruited from the emergency department of our local Level-1 trauma center for an advanced MRI scan at the acute stage. Sixteen age- and sex-matched controls were also recruited for comparison. Both group-based and individual-based independent component analysis of resting-state fMRI (rsfMRI) demonstrated reduced functional connectivity in both posterior cingulate cortex (PCC) and precuneus regions in comparison with controls, which is part of the default mode network (DMN). Further seed-based analysis confirmed reduced functional connectivity in these two regions and also demonstrated increased connectivity between these regions and other regions of the brain in mTBI. Seed-based analysis using the thalamus, hippocampus, and amygdala regions further demonstrated increased functional connectivity between these regions and other regions of the brain, particularly in the frontal lobe, in mTBI. Our data demonstrate alterations of multiple brain networks at the resting state, particularly increased functional connectivity in the frontal lobe, in response to brain concussion at the acute stage. Resting-state functional connectivity of the DMN could serve as a potential biomarker for improved detection of mTBI in the acute setting.
Collapse
Affiliation(s)
- Armin Iraji
- 1 Department of Biomedical Engineering, Wayne State University , Detroit, Michigan
| | | | - Robert D Welch
- 3 Department of Emergency Medicine, Wayne State University , Detroit, Michigan
| | - Brian J O'Neil
- 3 Department of Emergency Medicine, Wayne State University , Detroit, Michigan
| | - John L Woodard
- 4 Department of Psychology, Wayne State University , Detroit, Michigan
| | - Syed Imran Ayaz
- 3 Department of Emergency Medicine, Wayne State University , Detroit, Michigan
| | - Andrew Kulek
- 3 Department of Emergency Medicine, Wayne State University , Detroit, Michigan
| | - Valerie Mika
- 1 Department of Biomedical Engineering, Wayne State University , Detroit, Michigan.,3 Department of Emergency Medicine, Wayne State University , Detroit, Michigan
| | - Patrick Medado
- 3 Department of Emergency Medicine, Wayne State University , Detroit, Michigan
| | | | - Tianming Liu
- 6 Department of Computer Science, University of Georgia , Athens, Georgia
| | - E Mark Haacke
- 1 Department of Biomedical Engineering, Wayne State University , Detroit, Michigan.,7 Department of Radiology, Wayne State University , Detroit, Michigan
| | - Zhifeng Kou
- 1 Department of Biomedical Engineering, Wayne State University , Detroit, Michigan.,7 Department of Radiology, Wayne State University , Detroit, Michigan
| |
Collapse
|
71
|
Alfonso Romero-Sandoval E, Sweitzer S. Nonneuronal central mechanisms of pain: glia and immune response. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:325-58. [PMID: 25744678 DOI: 10.1016/bs.pmbts.2014.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The role of central glial cells in the mechanisms underlying pain has been intensively studied in the last two decades. Most studies on glia and pain focused on the potential detrimental role of glial cells following noxious stimulus/insults manifested as an "activation" or a "reactive" state (increase in glial marker expression and production of proinflammatory/nociceptive molecules). Therefore, "activated" or "reactive" glial cells became a target for the future generation of drugs to treat chronic pain. Several glial modulators that reduce the activation of glial cells have shown great efficacy in multiple animal (rodents mostly) models of pain (acute, subacute, chronic, inflammatory, neuropathic, surgical, etc.). These encouraging findings inspired clinical trials that have been completed in the last 5 years. Unfortunately, all clinical trials with these glial modulators have failed to demonstrate efficacy for the treatment of pain. New lines of investigation and elegant experimental designs are shedding light on alternative glial functions, which demonstrate that "glial reactivity" is not necessarily deleterious in some pathological conditions. New strategies to validate findings through our current animal models are necessary to enhance the translational value of our preclinical studies. Also, more studies using human subjects would enhance our understanding of glial cells in the context of pain. This chapter explores the available literature to objectively ponder the potential role of glial cells in human pain conditions.
Collapse
Affiliation(s)
- E Alfonso Romero-Sandoval
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, South Carolina, USA.
| | - Sarah Sweitzer
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, South Carolina, USA
| |
Collapse
|
72
|
Smith D, Brooks D, Wohlgehagen E, Rau T, Poulsen D. Temporal and Spatial Changes in the Pattern of Iba1 and CD68 Staining in the Rat Brain Following Severe Traumatic Brain Injury. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/mri.2015.42002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
73
|
Abstract
The brain is highly plastic after stroke or epilepsy; however, there is a paucity of brain plasticity investigation after traumatic brain injury (TBI). This mini review summarizes the most recent evidence of brain plasticity in human TBI patients from the perspective of advanced magnetic resonance imaging. Similar to other forms of acquired brain injury, TBI patients also demonstrated both structural reorganization as well as functional compensation by the recruitment of other brain regions. However, the large scale brain network alterations after TBI are still unknown, and the field is still short of proper means on how to guide the choice of TBI rehabilitation or treatment plan to promote brain plasticity. The authors also point out the new direction of brain plasticity investigation.
Collapse
Affiliation(s)
- Zhifeng Kou
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA ; Department of Radiology, Wayne State University, Detroit, MI, USA
| | - Armin Iraji
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA
| |
Collapse
|
74
|
Wagner AK. A Rehabilomics framework for personalized and translational rehabilitation research and care for individuals with disabilities: Perspectives and considerations for spinal cord injury. J Spinal Cord Med 2014; 37:493-502. [PMID: 25029659 PMCID: PMC4166184 DOI: 10.1179/2045772314y.0000000248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Despite many people having similar clinical presentation, demographic factors, and clinical care, outcome can differ for those sustaining significant injury such as spinal cord injury (SCI) and traumatic brain injury (TBI). In addition to traditional demographic, social, and clinical factors, variability also may be attributable to innate (including genetic, transcriptomic proteomic, epigenetic) biological variation that individuals bring to recovery and their unique response to their care and environment. Technologies collectively called "-omics" enable simultaneous measurement of an enormous number of biomolecules that can capture many potential biological contributors to heterogeneity of injury/disease course and outcome. Due to the nature of injury and complex disease, and its associations with impairment, disability, and recovery, rehabilitation does not lend itself to a singular "protocolized" plan of therapy. Yet, by nature and by necessity, rehabilitation medicine operates as a functional model of "Personalized Care". Thus, the challenge for successful programs of translational rehabilitation care and research is to identify viable approaches to examine broad populations, with varied impairments and functional limitations, and to identify effective treatment responses that incorporate personalized protocols to optimize functional recovery. The Rehabilomics framework is a translational model that provides an "-omics" overlay to the scientific study of rehabilitation processes and multidimensional outcomes. Rehabilomics research provides novel opportunities to evaluate the neurobiology of complex injury or chronic disease and can be used to examine methods and treatments for person-centered care among populations with disabilities. Exemplars for application in SCI and other neurorehabilitation populations are discussed.
Collapse
Affiliation(s)
- Amy K. Wagner
- Correspondence to: Amy K. Wagner, MD Department of Physical Medicine and Rehabilitation, Safar Center for Resuscitation Research, University of Pittsburgh, 3471 5th Avenue Suite 202, Pittsburgh, PA 15213, USA.
| |
Collapse
|
75
|
Butt AM, Fern RF, Matute C. Neurotransmitter signaling in white matter. Glia 2014; 62:1762-79. [PMID: 24753049 DOI: 10.1002/glia.22674] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 03/04/2014] [Accepted: 03/31/2014] [Indexed: 12/16/2022]
Abstract
White matter (WM) tracts are bundles of myelinated axons that provide for rapid communication throughout the CNS and integration in grey matter (GM). The main cells in myelinated tracts are oligodendrocytes and astrocytes, with small populations of microglia and oligodendrocyte precursor cells. The prominence of neurotransmitter signaling in WM, which largely exclude neuronal cell bodies, indicates it must have physiological functions other than neuron-to-neuron communication. A surprising aspect is the diversity of neurotransmitter signaling in WM, with evidence for glutamatergic, purinergic (ATP and adenosine), GABAergic, glycinergic, adrenergic, cholinergic, dopaminergic and serotonergic signaling, acting via a wide range of ionotropic and metabotropic receptors. Both axons and glia are potential sources of neurotransmitters and may express the respective receptors. The physiological functions of neurotransmitter signaling in WM are subject to debate, but glutamate and ATP-mediated signaling have been shown to evoke Ca(2+) signals in glia and modulate axonal conduction. Experimental findings support a model of neurotransmitters being released from axons during action potential propagation acting on glial receptors to regulate the homeostatic functions of astrocytes and myelination by oligodendrocytes. Astrocytes also release neurotransmitters, which act on axonal receptors to strengthen action potential propagation, maintaining signaling along potentially long axon tracts. The co-existence of multiple neurotransmitters in WM tracts suggests they may have diverse functions that are important for information processing. Furthermore, the neurotransmitter signaling phenomena described in WM most likely apply to myelinated axons of the cerebral cortex and GM areas, where they are doubtless important for higher cognitive function.
Collapse
Affiliation(s)
- Arthur M Butt
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, United Kingdom
| | | | | |
Collapse
|