1
|
Cognacq G, Attwood JE, DeLuca GC. Traumatic Brain Injury and Alzheimer's Disease: A Shared Neurovascular Hypothesis. Neurosci Insights 2025; 20:26331055251323292. [PMID: 40124421 PMCID: PMC11926848 DOI: 10.1177/26331055251323292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Traumatic brain injury (TBI) is a modifiable risk factor for Alzheimer's disease (AD). TBI and AD share several histopathological hallmarks: namely, beta-amyloid aggregation, tau hyperphosphorylation, and plasma protein infiltration. The relative contributions of these proteinopathies and their interplay in the pathogenesis of both conditions remains unclear although important differences are emerging. This review synthesises emerging evidence for the critical role of the neurovascular unit in mediating protein accumulation and neurotoxicity in both TBI and AD. We propose a shared pathogenic cascade centred on a neurovascular unit, in which increased blood-brain barrier permeability induces a series of noxious mechanisms leading to neuronal loss, synaptic dysfunction and ultimately cognitive dysfunction in both conditions. We explore the application of this hypothesis to outstanding research questions and potential treatments for TBI and AD, as well as other neurodegenerative and neuroinflammatory conditions. Limitations of this hypothesis, including the challenges of establishing a causal relationship between neurovascular damage and proteinopathies, are also discussed.
Collapse
Affiliation(s)
- Gabrielle Cognacq
- John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, Oxfordshire, UK
| | - Jonathan E Attwood
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, Oxfordshire, UK
| |
Collapse
|
2
|
Astakhova O, Ivanova A, Komoltsev I, Gulyaeva N, Enikolopov G, Lazutkin A. Traumatic Brain Injury Promotes Neurogenesis and Oligodendrogenesis in Subcortical Brain Regions of Mice. Cells 2025; 14:92. [PMID: 39851520 PMCID: PMC11764027 DOI: 10.3390/cells14020092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Traumatic brain injury (TBI) is one of the major causes of severe neurological disorders and long-term dysfunction in the nervous system. Besides inducing neurodegeneration, TBI alters stem cell activity and neurogenesis within primary neurogenic niches. However, the fate of dividing cells in other brain regions remains unclear despite offering potential targets for therapeutic intervention. Here, we investigated cell division and differentiation in non-neurogenic brain regions during the acute and delayed phases of TBI-induced neurodegeneration. We subjected mice to lateral fluid percussion injury (LFPI) to model TBI and analyzed them 1 or 7 weeks later. To assess cellular proliferation and differentiation, we administered 5-ethinyl-2'-deoxyuridine (EdU) and determined the number and identity of dividing cells 2 h later using markers of neuronal precursors and astro-, micro-, and oligodendroglia. Our results demonstrated a significant proliferative response in several brain regions at one week post-injury that notably diminished by seven weeks, except in the optic tract. In addition to active astro- and microgliosis, we detected oligodendrogenesis in the striatum and optic tract. Furthermore, we observed trauma-induced neurogenesis in the striatum. These findings suggest that subcortical structures, particularly the striatum and optic tract, may possess a potential for self-repair through neuronal regeneration and axon remyelination.
Collapse
Affiliation(s)
- Olga Astakhova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (O.A.)
- Department of Human and Animal Physiology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - Anna Ivanova
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow 119991, Russia;
| | - Ilia Komoltsev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (O.A.)
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow 115419, Russia
| | - Natalia Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (O.A.)
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow 115419, Russia
| | - Grigori Enikolopov
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Alexander Lazutkin
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Moscow 117485, Russia; (O.A.)
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
3
|
Gober IG, Russell AL, Shick TJ, Vagni VA, Carlson JC, Kochanek PM, Wagner AK. Exploratory assessment of the effect of systemic administration of soluble glycoprotein 130 on cognitive performance and chemokine levels in a mouse model of experimental traumatic brain injury. J Neuroinflammation 2024; 21:149. [PMID: 38840141 PMCID: PMC11155101 DOI: 10.1186/s12974-024-03129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024] Open
Abstract
Uncontrolled neuroinflammation mediates traumatic brain injury (TBI) pathology and impairs recovery. Interleukin-6 (IL-6), a pleiotropic inflammatory regulator, is associated with poor clinical TBI outcomes. IL-6 operates via classical-signaling through membrane-bound IL-6 receptor (IL-6R) and trans-signaling through soluble IL-6 receptor (s)IL-6R. IL-6 trans-signaling specifically contributes to neuropathology, making it a potential precision therapeutic TBI target. Soluble glycoprotein 130 (sgp130) prevents IL-6 trans-signaling, sparing classical signaling, thus is a possible treatment. Mice received either controlled cortical impact (CCI) (6.0 ± 0.2 m/s; 2 mm; 50-60ms) or sham procedures. Vehicle (VEH) or sgp130-Fc was subcutaneously administered to sham (VEH or 1 µg) and CCI (VEH, 0.25 µg or 1 µg) mice on days 1, 4, 7, 10 and 13 post-surgery to assess effects on cognition [Morris Water Maze (MWM)] and ipsilateral hemisphere IL-6 related biomarkers (day 21 post-surgery). CCI + sgp130-Fc groups (0.25 µg and 1 µg) were combined for analysis given similar behavior/biomarker outcomes. CCI + VEH mice had longer latencies and path lengths to the platform and increased peripheral zone time versus Sham + VEH and Sham + sgp130-Fc mice, suggesting injury-induced impairments in learning and anxiety. CCI + sgp130-Fc mice had shorter platform latencies and path lengths and had decreased peripheral zone time, indicating a therapeutic benefit of sgp130-Fc after injury on learning and anxiety. Interestingly, Sham + sgp130-Fc mice had shorter platform latencies, path lengths and peripheral zone times than Sham + VEH mice, suggesting a beneficial effect of sgp130-Fc, independent of injury. CCI + VEH mice had increased brain IL-6 and decreased sgp130 levels versus Sham + VEH and Sham + sgp130-Fc mice. There was no treatment effect on IL-6, sIL6-R or sgp130 in Sham + VEH versus Sham + sgp130-Fc mice. There was also no treatment effect on IL-6 in CCI + VEH versus CCI + sgp130-Fc mice. However, CCI + sgp130-Fc mice had increased sIL-6R and sgp130 versus CCI + VEH mice, demonstrating sgp130-Fc treatment effects on brain biomarkers. Inflammatory chemokines (MIP-1β, IP-10, MIG) were increased in CCI + VEH mice versus Sham + VEH and Sham + sgp130-Fc mice. However, CCI + sgp130-Fc mice had decreased chemokine levels versus CCI + VEH mice. IL-6 positively correlated, while sgp130 negatively correlated, with chemokine levels. Overall, we found that systemic sgp130-Fc treatment after CCI improved learning, decreased anxiety and reduced CCI-induced brain chemokines. Future studies will explore sex-specific dosing and treatment mechanisms for sgp130-Fc therapy.
Collapse
Affiliation(s)
- Ian G Gober
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Ashley L Russell
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Tyler J Shick
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Vincent A Vagni
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenna C Carlson
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA.
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA.
- Center for Neuroscience, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
LeVine SM. Exploring Potential Mechanisms Accounting for Iron Accumulation in the Central Nervous System of Patients with Alzheimer's Disease. Cells 2024; 13:689. [PMID: 38667304 PMCID: PMC11049304 DOI: 10.3390/cells13080689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
Elevated levels of iron occur in both cortical and subcortical regions of the CNS in patients with Alzheimer's disease. This accumulation is present early in the disease process as well as in more advanced stages. The factors potentially accounting for this increase are numerous, including: (1) Cells increase their uptake of iron and reduce their export of iron, as iron becomes sequestered (trapped within the lysosome, bound to amyloid β or tau, etc.); (2) metabolic disturbances, such as insulin resistance and mitochondrial dysfunction, disrupt cellular iron homeostasis; (3) inflammation, glutamate excitotoxicity, or other pathological disturbances (loss of neuronal interconnections, soluble amyloid β, etc.) trigger cells to acquire iron; and (4) following neurodegeneration, iron becomes trapped within microglia. Some of these mechanisms are also present in other neurological disorders and can also begin early in the disease course, indicating that iron accumulation is a relatively common event in neurological conditions. In response to pathogenic processes, the directed cellular efforts that contribute to iron buildup reflect the importance of correcting a functional iron deficiency to support essential biochemical processes. In other words, cells prioritize correcting an insufficiency of available iron while tolerating deposited iron. An analysis of the mechanisms accounting for iron accumulation in Alzheimer's disease, and in other relevant neurological conditions, is put forward.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Cell Biology and Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Mail Stop 3043, Kansas City, KS 66160, USA
| |
Collapse
|
5
|
da Silva Fiorin F, do Espírito Santo CC, Da Silva JT, Chung MK. Inflammation, brain connectivity, and neuromodulation in post-traumatic headache. Brain Behav Immun Health 2024; 35:100723. [PMID: 38292321 PMCID: PMC10827408 DOI: 10.1016/j.bbih.2024.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Post-traumatic headache (PTH) is a debilitating condition that affects individuals with different levels of traumatic brain injury (TBI) severity. The difficulties in developing an effective treatment are related to a lack of understanding the complicated mechanisms and neurobiological changes in brain function after a brain injury. Preclinical studies have indicated that peripheral and central sensitization of the trigeminal nociceptive pathways contributes to PTH. While recent brain imaging studies have uncovered widespread changes in brain functional connectivity following trauma, understanding exactly how these networks contribute to PTH after injury remains unknown. Stimulation of peripheral (trigeminal or vagus) nerves show promising efficacies in PTH experimental animals, likely mediated by influencing TBI-induced pathological plasticity by decreasing neuroinflammation and neuronal apoptosis. Non-invasive brain stimulations, such as transcranial magnetic or direct current stimulations, show analgesia for multiple chronic pain conditions, including PTH. Better mechanistic understanding of analgesia achieved by neuromodulations can define peripheral and central mechanisms involved in the development, the resolution, and the management of PTH.
Collapse
Affiliation(s)
- Fernando da Silva Fiorin
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - Caroline Cunha do Espírito Santo
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Brazil
| | - Joyce T. Da Silva
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| |
Collapse
|
6
|
Szczygielski J, Hubertus V, Kruchten E, Müller A, Albrecht LF, Schwerdtfeger K, Oertel J. Prolonged course of brain edema and neurological recovery in a translational model of decompressive craniectomy after closed head injury in mice. Front Neurol 2023; 14:1308683. [PMID: 38053795 PMCID: PMC10694459 DOI: 10.3389/fneur.2023.1308683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
Background The use of decompressive craniectomy in traumatic brain injury (TBI) remains a matter of debate. According to the DECRA trial, craniectomy may have a negative impact on functional outcome, while the RescueICP trial revealed a positive effect of surgical decompression, which is evolving over time. This ambivalence of craniectomy has not been studied extensively in controlled laboratory experiments. Objective The goal of the current study was to investigate the prolonged effects of decompressive craniectomy (both positive and negative) in an animal model. Methods Male mice were assigned to the following groups: sham, decompressive craniectomy, TBI and TBI followed by craniectomy. The analysis of functional outcome was performed at time points 3d, 7d, 14d and 28d post trauma according to the Neurological Severity Score and Beam Balance Score. At the same time points, magnetic resonance imaging was performed, and brain edema was analyzed. Results Animals subjected to both trauma and craniectomy presented the exacerbation of the neurological impairment that was apparent mostly in the early course (up to 7d) after injury. Decompressive craniectomy also caused a significant increase in brain edema volume (initially cytotoxic with a secondary shift to vasogenic edema and gliosis). Notably, delayed edema plus gliosis appeared also after decompression even without preceding trauma. Conclusion In prolonged outcomes, craniectomy applied after closed head injury in mice aggravates posttraumatic brain edema, leading to additional functional impairment. This effect is, however, transient. Treatment options that reduce brain swelling after decompression may accelerate neurological recovery and should be explored in future experiments.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Instutute of Neuropathology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Institute of Medical Sciences, University of Rzeszów, Rzeszow, Poland
| | - Vanessa Hubertus
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Department of Neurosurgery, Charité University Medicine, Berlin, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| | - Eduard Kruchten
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Institute of Interventional and Diagnostic Radiology, Karlsruhe, Germany
| | - Andreas Müller
- Department of Radiology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Lisa Franziska Albrecht
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Karsten Schwerdtfeger
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| |
Collapse
|
7
|
Tang L, Liu S, Li S, Chen Y, Xie B, Zhou J. Induction Mechanism of Ferroptosis, Necroptosis, and Pyroptosis: A Novel Therapeutic Target in Nervous System Diseases. Int J Mol Sci 2023; 24:10127. [PMID: 37373274 DOI: 10.3390/ijms241210127] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
In recent years, three emerging cell deaths, ferroptosis, necroptosis and pyroptosis, have gradually attracted everyone's attention, and they also play an important role in the occurrence and development of various diseases. Ferroptosis is an idiographic iron-dependent form regulated cell death with the hallmark of accumulation of the intracellular reactive oxygen species (ROS). Necroptosis is a form of regulated necrotic cell death mediated by the receptor-interacting protein kinase 1(RIPK1) and receptor-interacting protein kinase 3RIPK3. Pyroptosis, also known as cell inflammatory necrosis, is a programmed cell necrosis mediated by Gasdermin D (GSDMD). It is manifested by the continuous swelling of the cells until the cell membrane ruptures, resulting in the release of the cell contents and the activation of a strong inflammatory response. Neurological disorders remain a clinical challenge and patients do not respond well to conventional treatments. Nerve cell death can aggravate the occurrence and development of neurological diseases. This article reviews the specific mechanisms of these three types of cell death and their relationship with neurological diseases and the evidence for the role of the three types of cell death in neurological diseases; understanding these pathways and their mechanisms is helpful for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Lu Tang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou 646000, China
| | - Sitong Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou 646000, China
| | - Shiwei Li
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou 646000, China
| | - Ye Chen
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou 646000, China
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
8
|
King H, Reiber M, Philippi V, Stirling H, Aulehner K, Bankstahl M, Bleich A, Buchecker V, Glasenapp A, Jirkof P, Miljanovic N, Schönhoff K, von Schumann L, Leenaars C, Potschka H. Anesthesia and analgesia for experimental craniotomy in mice and rats: a systematic scoping review comparing the years 2009 and 2019. Front Neurosci 2023; 17:1143109. [PMID: 37207181 PMCID: PMC10188949 DOI: 10.3389/fnins.2023.1143109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/27/2023] [Indexed: 05/21/2023] Open
Abstract
Experimental craniotomies are a common surgical procedure in neuroscience. Because inadequate analgesia appears to be a problem in animal-based research, we conducted this review and collected information on management of craniotomy-associated pain in laboratory mice and rats. A comprehensive search and screening resulted in the identification of 2235 studies, published in 2009 and 2019, describing craniotomy in mice and/or rats. While key features were extracted from all studies, detailed information was extracted from a random subset of 100 studies/year. Reporting of perioperative analgesia increased from 2009 to 2019. However, the majority of studies from both years did not report pharmacologic pain management. Moreover, reporting of multimodal treatments remained at a low level, and monotherapeutic approaches were more common. Among drug groups, reporting of pre- and postoperative administration of non-steroidal anti-inflammatory drugs, opioids, and local anesthetics in 2019 exceeded that of 2009. In summary, these results suggest that inadequate analgesia and oligoanalgesia are persistent issues associated with experimental intracranial surgery. This underscores the need for intensified training of those working with laboratory rodents subjected to craniotomies. Systematic review registration https://osf.io/7d4qe.
Collapse
Affiliation(s)
- Hannah King
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Maria Reiber
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Vanessa Philippi
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Helen Stirling
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Katharina Aulehner
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Marion Bankstahl
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - André Bleich
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Verena Buchecker
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Aylina Glasenapp
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Paulin Jirkof
- Office for Animal Welfare and 3Rs, University of Zurich, Zurich, Switzerland
| | - Nina Miljanovic
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Katharina Schönhoff
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Lara von Schumann
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Cathalijn Leenaars
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| |
Collapse
|
9
|
Newcombe VFJ, Ashton NJ, Posti JP, Glocker B, Manktelow A, Chatfield DA, Winzeck S, Needham E, Correia MM, Williams GB, Simrén J, Takala RSK, Katila AJ, Maanpää HR, Tallus J, Frantzén J, Blennow K, Tenovuo O, Zetterberg H, Menon DK. Post-acute blood biomarkers and disease progression in traumatic brain injury. Brain 2022; 145:2064-2076. [PMID: 35377407 PMCID: PMC9326940 DOI: 10.1093/brain/awac126] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/09/2022] [Accepted: 02/13/2022] [Indexed: 11/23/2022] Open
Abstract
There is substantial interest in the potential for traumatic brain injury to result in progressive neurological deterioration. While blood biomarkers such as glial fibrillary acid protein (GFAP) and neurofilament light have been widely explored in characterizing acute traumatic brain injury (TBI), their use in the chronic phase is limited. Given increasing evidence that these proteins may be markers of ongoing neurodegeneration in a range of diseases, we examined their relationship to imaging changes and functional outcome in the months to years following TBI. Two-hundred and three patients were recruited in two separate cohorts; 6 months post-injury (n = 165); and >5 years post-injury (n = 38; 12 of whom also provided data ∼8 months post-TBI). Subjects underwent blood biomarker sampling (n = 199) and MRI (n = 172; including diffusion tensor imaging). Data from patient cohorts were compared to 59 healthy volunteers and 21 non-brain injury trauma controls. Mean diffusivity and fractional anisotropy were calculated in cortical grey matter, deep grey matter and whole brain white matter. Accelerated brain ageing was calculated at a whole brain level as the predicted age difference defined using T1-weighted images, and at a voxel-based level as the annualized Jacobian determinants in white matter and grey matter, referenced to a population of 652 healthy control subjects. Serum neurofilament light concentrations were elevated in the early chronic phase. While GFAP values were within the normal range at ∼8 months, many patients showed a secondary and temporally distinct elevations up to >5 years after injury. Biomarker elevation at 6 months was significantly related to metrics of microstructural injury on diffusion tensor imaging. Biomarker levels at ∼8 months predicted white matter volume loss at >5 years, and annualized brain volume loss between ∼8 months and 5 years. Patients who worsened functionally between ∼8 months and >5 years showed higher than predicted brain age and elevated neurofilament light levels. GFAP and neurofilament light levels can remain elevated months to years after TBI, and show distinct temporal profiles. These elevations correlate closely with microstructural injury in both grey and white matter on contemporaneous quantitative diffusion tensor imaging. Neurofilament light elevations at ∼8 months may predict ongoing white matter and brain volume loss over >5 years of follow-up. If confirmed, these findings suggest that blood biomarker levels at late time points could be used to identify TBI survivors who are at high risk of progressive neurological damage.
Collapse
Affiliation(s)
- Virginia F J Newcombe
- University Division of Anaesthesia, Department of Medicine, University of
Cambridge, Cambridge, UK
| | - Nicholas J Ashton
- Wallenberg Centre for Molecular and Translational Medicine, University of
Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and
Physiology, The Sahlgrenska Academy at the University of Gothenburg,
Mölndal, Sweden
- King’s College London, Institute of Psychiatry, Psychology and
Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute,
London, UK
- Mental Health and Biomedical Research Unit for Dementia, Maudsley NIHR
Biomedical Research Centre, London, UK
| | - Jussi P Posti
- Neurocenter, Department of Neurosurgery, Turku University Hospital and
University of Turku, Turku, Finland
- Turku Brain Injury Center, Turku University Hospital and University of
Turku, Turku, Finland
| | - Ben Glocker
- Biomedical Image Analysis Group, Department of Computing, Imperial College
London, London, UK
| | - Anne Manktelow
- University Division of Anaesthesia, Department of Medicine, University of
Cambridge, Cambridge, UK
| | - Doris A Chatfield
- University Division of Anaesthesia, Department of Medicine, University of
Cambridge, Cambridge, UK
| | - Stefan Winzeck
- University Division of Anaesthesia, Department of Medicine, University of
Cambridge, Cambridge, UK
- Biomedical Image Analysis Group, Department of Computing, Imperial College
London, London, UK
| | - Edward Needham
- University Division of Anaesthesia, Department of Medicine, University of
Cambridge, Cambridge, UK
| | - Marta M Correia
- MRC (Medical Research Council) Cognition and Brain Sciences Unit,
University of Cambridge, Cambridge, UK
| | - Guy B Williams
- Wolfson Brain Imaging Centre, Department of Clinical
Neurosciences, Cambridge, UK
| | - Joel Simrén
- Institute of Neuroscience and Physiology, Department of Psychiatry and
Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg,
Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal, Sweden
| | - Riikka S K Takala
- Perioperative Services, Intensive Care Medicine and Pain Management,
Department of Anesthesiology and Intensive Care, Turku University Hospital, University
of Turku, Turku, Finland
| | - Ari J Katila
- Perioperative Services, Intensive Care Medicine and Pain Management,
Department of Anesthesiology and Intensive Care, Turku University Hospital, University
of Turku, Turku, Finland
| | - Henna Riikka Maanpää
- Neurocenter, Department of Neurosurgery, Turku University Hospital and
University of Turku, Turku, Finland
- Turku Brain Injury Center, Turku University Hospital and University of
Turku, Turku, Finland
| | - Jussi Tallus
- Turku Brain Injury Center, Turku University Hospital and University of
Turku, Turku, Finland
| | - Janek Frantzén
- Neurocenter, Department of Neurosurgery, Turku University Hospital and
University of Turku, Turku, Finland
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and
Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg,
Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal, Sweden
| | - Olli Tenovuo
- Turku Brain Injury Center, Turku University Hospital and University of
Turku, Turku, Finland
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and
Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg,
Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of
Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, University College
London, London, UK
- Hong Kong Center for Neurodegenerative Disease,
Hong Kong, China
| | - David K Menon
- University Division of Anaesthesia, Department of Medicine, University of
Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Traumatic Brain Injury: An Age-Dependent View of Post-Traumatic Neuroinflammation and Its Treatment. Pharmaceutics 2021; 13:pharmaceutics13101624. [PMID: 34683918 PMCID: PMC8537402 DOI: 10.3390/pharmaceutics13101624] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability all over the world. TBI leads to (1) an inflammatory response, (2) white matter injuries and (3) neurodegenerative pathologies in the long term. In humans, TBI occurs most often in children and adolescents or in the elderly, and it is well known that immune responses and the neuroregenerative capacities of the brain, among other factors, vary over a lifetime. Thus, age-at-injury can influence the consequences of TBI. Furthermore, age-at-injury also influences the pharmacological effects of drugs. However, the post-TBI inflammatory, neuronal and functional consequences have been mostly studied in experimental young adult animal models. The specificity and the mechanisms underlying the consequences of TBI and pharmacological responses are poorly understood in extreme ages. In this review, we detail the variations of these age-dependent inflammatory responses and consequences after TBI, from an experimental point of view. We investigate the evolution of microglial, astrocyte and other immune cells responses, and the consequences in terms of neuronal death and functional deficits in neonates, juvenile, adolescent and aged male animals, following a single TBI. We also describe the pharmacological responses to anti-inflammatory or neuroprotective agents, highlighting the need for an age-specific approach to the development of therapies of TBI.
Collapse
|
11
|
Snyder-Keller A, Bolivar VJ, Zink S, Kramer LD. Brain Iron Accumulation and the Formation of Calcifications After Developmental Zika Virus Infection. J Neuropathol Exp Neurol 2020; 79:767-776. [PMID: 32483612 DOI: 10.1093/jnen/nlaa043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/24/2020] [Indexed: 12/20/2022] Open
Abstract
Intracranial calcifications (ICC) are the most common neuropathological finding in the brains of children exposed in utero to the Zika virus (ZIKV). Using a mouse model of developmental ZIKV infection, we reported widespread calcifications in the brains of susceptible mice that correlated in multiple ways with the behavioral deficits observed. Here, we examined the time course of ICC development and the role of iron deposition in this process, in 3 genetically distinct inbred strains of mice. Brain iron deposits were evident by Perls' staining at 2 weeks post infection, becoming increasingly dense and coinciding with calcium buildup and the formation of ICCs. A regional analysis of the brains of susceptible mice (C57BL/6J and 129S1/SvImJ strains) revealed the presence of iron initially in regions containing many ZIKV-immunoreactive cells, but then spreading to regions containing few infected cells, most notably the thalamus and the fasciculus retroflexus. Microglial activation was widespread initially and later delineated the sites of ICC formation. Behavioral tests conducted at 5-6 weeks of age revealed greater deficits in mice with the most extensive iron deposition and calcification of subcortical regions, such as thalamus. These findings point to iron deposition as a key factor in the development of ICCs after developmental ZIKV infection.
Collapse
Affiliation(s)
- Abigail Snyder-Keller
- Wadsworth Center, New York State Department of Health.,Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York
| | - Valerie J Bolivar
- Wadsworth Center, New York State Department of Health.,Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York
| | - Steven Zink
- Wadsworth Center, New York State Department of Health
| | - Laura D Kramer
- Wadsworth Center, New York State Department of Health.,Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York
| |
Collapse
|
12
|
Wright DK, Brady RD, Kamnaksh A, Trezise J, Sun M, McDonald SJ, Mychasiuk R, Kolbe SC, Law M, Johnston LA, O'Brien TJ, Agoston DV, Shultz SR. Repeated mild traumatic brain injuries induce persistent changes in plasma protein and magnetic resonance imaging biomarkers in the rat. Sci Rep 2019; 9:14626. [PMID: 31602002 PMCID: PMC6787341 DOI: 10.1038/s41598-019-51267-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/28/2019] [Indexed: 01/05/2023] Open
Abstract
A single mild traumatic brain injury (mTBI) typically causes only transient symptoms, but repeated mTBI (RmTBI) is associated with cumulative and chronic neurological abnormalities. Clinical management of mTBI is challenging due to the heterogeneous, subjective and transient nature of symptoms, and thus would be aided by objective biomarkers. Promising biomarkers including advanced magnetic resonance imaging (MRI) and plasma levels of select proteins were examined here in a rat model of RmTBI. Rats received either two mild fluid percussion or sham injuries administered five days apart. Rats underwent MRI and behavioral testing 1, 3, 5, 7, and 30 days after the second injury and blood samples were collected on days 1, 7, and 30. Structural and diffusion-weighted MRI revealed that RmTBI rats had abnormalities in the cortex and corpus callosum. Proteomic analysis of plasma found that RmTBI rats had abnormalities in markers indicating axonal and vascular injury, metabolic and mitochondrial dysfunction, and glial reactivity. These changes occurred in the presence of ongoing cognitive and sensorimotor deficits in the RmTBI rats. Our findings demonstrate that RmTBI can result in chronic neurological abnormalities, provide insight into potential contributing pathophysiological mechanisms, and supports the use of MRI and plasma protein measures as RmTBI biomarkers.
Collapse
Affiliation(s)
- David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Alaa Kamnaksh
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Jack Trezise
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Scott C Kolbe
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Leigh A Johnston
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia.,Department of Biomedical Engineering, 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 Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia. .,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
13
|
Schweser F, Kyyriäinen J, Preda M, Pitkänen A, Toffolo K, Poulsen A, Donahue K, Levy B, Poulsen D. Visualization of thalamic calcium influx with quantitative susceptibility mapping as a potential imaging biomarker for repeated mild traumatic brain injury. Neuroimage 2019; 200:250-258. [PMID: 31201986 DOI: 10.1016/j.neuroimage.2019.06.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/03/2019] [Accepted: 06/11/2019] [Indexed: 11/17/2022] Open
Abstract
A key event in the pathophysiology of traumatic brain injury (TBI) is the influx of substantial amounts of Ca2+ into neurons, particularly in the thalamus. Detection of this calcium influx in vivo would provide a window into the biochemical mechanisms of TBI with potentially significant clinical implications. In the present work, our central hypothesis was that the Ca2+ influx could be imaged in vivo with the relatively recent MRI technique of quantitative susceptibility mapping (QSM). Wistar rats were divided into five groups: naive controls, sham-operated experimental controls, single mild TBI, repeated mild TBI, and single severe TBI. We employed the lateral fluid percussion injury (FPI) model, which replicates clinical TBI without skull fracture, performed 9.4 Tesla MRI with a 3D multi-echo gradient-echo sequence at weeks 1 and 4 post-injury, computed susceptibility maps using V-SHARP and the QUASAR-HEIDI technique, and performed histology. Sham, experimental controls animals, and injured animals did not demonstrate calcifications at 1 week after the injury. At week 4, calcifications were found in the ipsilateral thalamus of 25-50% of animals after a single TBI and 83% of animals after repeated mild TBI. The location and appearance of calcifications on stained sections was consistent with the appearance on the in vivo susceptibility maps (correlation of volumes: r = 0.7). Our findings suggest that persistent calcium deposits represent a primary pathology of repeated injury and that FPI-QSM has the potential to become a sensitive tool for studying pathophysiology related to mild TBI in vivo.
Collapse
Affiliation(s)
- Ferdinand Schweser
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, The State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging, Clinical and Translational Science Institute, University at Buffalo, The State University of New York, Buffalo, NY, USA.
| | - Jenni Kyyriäinen
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI, 70211, Kuopio, Finland
| | - Marilena Preda
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, The State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging, Clinical and Translational Science Institute, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Asla Pitkänen
- Epilepsy Research Laboratory, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI, 70211, Kuopio, Finland
| | - Kathryn Toffolo
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Austin Poulsen
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Kaitlynn Donahue
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Benett Levy
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - David Poulsen
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
14
|
Daglas M, Adlard PA. The Involvement of Iron in Traumatic Brain Injury and Neurodegenerative Disease. Front Neurosci 2018; 12:981. [PMID: 30618597 PMCID: PMC6306469 DOI: 10.3389/fnins.2018.00981] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/07/2018] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) consists of acute and long-term pathophysiological sequelae that ultimately lead to cognitive and motor function deficits, with age being a critical risk factor for poorer prognosis. TBI has been recently linked to the development of neurodegenerative diseases later in life including Alzheimer’s disease, Parkinson’s disease, chronic traumatic encephalopathy, and multiple sclerosis. The accumulation of iron in the brain has been documented in a number of neurodegenerative diseases, and also in normal aging, and can contribute to neurotoxicity through a variety of mechanisms including the production of free radicals leading to oxidative stress, excitotoxicity and by promoting inflammatory reactions. A growing body of evidence similarly supports a deleterious role of iron in the pathogenesis of TBI. Iron deposition in the injured brain can occur via hemorrhage/microhemorrhages (heme-bound iron) or independently as labile iron (non-heme bound), which is considered to be more damaging to the brain. This review focusses on the role of iron in potentiating neurodegeneration in TBI, with insight into the intersection with neurodegenerative conditions. An important implication of this work is the potential for therapeutic approaches that target iron to attenuate the neuropathology/phenotype related to TBI and to also reduce the associated risk of developing neurodegenerative disease.
Collapse
Affiliation(s)
- Maria Daglas
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul A Adlard
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
15
|
Imaging biomarkers of epileptogenecity after traumatic brain injury - Preclinical frontiers. Neurobiol Dis 2018; 123:75-85. [PMID: 30321600 DOI: 10.1016/j.nbd.2018.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/04/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
Posttraumatic epilepsy (PTE) is a major neurodegenerative disease accounting for 20% of symptomatic epilepsy cases. A long latent phase offers a potential window for prophylactic treatment strategies to prevent epilepsy onset, provided that the patients at risk can be identified. Some promising imaging biomarker candidates for posttraumatic epileptogenesis have been identified, but more are required to provide the specificity and sensitivity for accurate prediction. Experimental models and preclinical longitudinal, multimodal imaging studies allow follow-up of complex cascade of events initiated by traumatic brain injury, as well as monitoring of treatment effects. Preclinical imaging data from the posttraumatic brain are rich in information, yet examination of their specific relevance to epilepsy is lacking. Accumulating evidence from ongoing preclinical studies in TBI support insight into processes involved in epileptogenesis, e.g. inflammation and changes in functional and structural brain-wide connectivity. These efforts are likely to produce both new biomarkers and treatment targets for PTE.
Collapse
|
16
|
Elliott MB, Ward SJ, Abood ME, Tuma RF, Jallo JI. Understanding the endocannabinoid system as a modulator of the trigeminal pain response to concussion. Concussion 2018; 2:CNC49. [PMID: 30202590 PMCID: PMC6122691 DOI: 10.2217/cnc-2017-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/29/2017] [Indexed: 12/29/2022] Open
Abstract
Post-traumatic headache is the most common symptom of postconcussion syndrome and becomes a chronic neurological disorder in a substantial proportion of patients. This review provides a brief overview of the epidemiology of postconcussion headache, research models used to study this disorder, as well as the proposed mechanisms. An objective of this review is to enhance the understanding of how the endogenous cannabinoid system is essential for maintaining the balance of the CNS and regulating inflammation after injury, and in turn making the endocannabinoid system a potential modulator of the trigeminal response to concussion. The review describes the role of endocannabinoid modulation of pain and the potential for use of phytocannabinoids to treat pain, migraine and concussion.
Collapse
Affiliation(s)
- Melanie B Elliott
- Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA.,Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA
| | - Sara J Ward
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Mary E Abood
- Department of Anatomy & Cell Biology, Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Anatomy & Cell Biology, Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Ronald F Tuma
- Department of Physiology Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Physiology Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Jack I Jallo
- Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA.,Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA
| |
Collapse
|
17
|
Dating of Traumatic Brain Injury in Forensic Cases Using Immunohistochemical Markers (I). ACTA ACUST UNITED AC 2018; 39:201-207. [DOI: 10.1097/paf.0000000000000412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
18
|
Prasad GL, Anmol N, Menon GR. Outcome of Traumatic Brain Injury in the Elderly Population: A Tertiary Center Experience in a Developing Country. World Neurosurg 2018; 111:e228-e234. [DOI: 10.1016/j.wneu.2017.12.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 12/04/2017] [Accepted: 12/08/2017] [Indexed: 12/11/2022]
|
19
|
Pischiutta F, Micotti E, Hay JR, Marongiu I, Sammali E, Tolomeo D, Vegliante G, Stocchetti N, Forloni G, De Simoni MG, Stewart W, Zanier ER. Single severe traumatic brain injury produces progressive pathology with ongoing contralateral white matter damage one year after injury. Exp Neurol 2017; 300:167-178. [PMID: 29126888 DOI: 10.1016/j.expneurol.2017.11.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/27/2017] [Accepted: 11/06/2017] [Indexed: 01/29/2023]
Abstract
There is increasing recognition that traumatic brain injury (TBI) may initiate long-term neurodegenerative processes, particularly chronic traumatic encephalopathy. However, insight into the mechanisms transforming an initial biomechanical injury into a neurodegenerative process remain elusive, partly as a consequence of the paucity of informative pre-clinical models. This study shows the functional, whole brain imaging and neuropathological consequences at up to one year survival from single severe TBI by controlled cortical impact in mice. TBI mice displayed persistent sensorimotor and cognitive deficits. Longitudinal T2 weighted magnetic resonance imaging (MRI) showed progressive ipsilateral (il) cortical, hippocampal and striatal volume loss, with diffusion tensor imaging demonstrating decreased fractional anisotropy (FA) at up to one year in the il-corpus callosum (CC: -30%) and external capsule (EC: -21%). Parallel neuropathological studies indicated reduction in neuronal density, with evidence of microgliosis and astrogliosis in the il-cortex, with further evidence of microgliosis and astrogliosis in the il-thalamus. One year after TBI there was also a decrease in FA in the contralateral (cl) CC (-17%) and EC (-13%), corresponding to histopathological evidence of white matter loss (cl-CC: -68%; cl-EC: -30%) associated with ongoing microgliosis and astrogliosis. These findings indicate that a single severe TBI induces bilateral, long-term and progressive neuropathology at up to one year after injury. These observations support this model as a suitable platform for exploring the mechanistic link between acute brain injury and late and persistent neurodegeneration.
Collapse
Affiliation(s)
- Francesca Pischiutta
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Jennifer R Hay
- Institute of Neuroscience and Psychology, University of Glasgow, UK; Department of Laboratory Medicine, Queen Elizabeth University Hospital, Glasgow, UK
| | - Ines Marongiu
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Eliana Sammali
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy; Department of Cerebrovascular Diseases, Fondazione IRCCS - Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniele Tolomeo
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Gloria Vegliante
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Nino Stocchetti
- Department of Physiopathology and Transplantation, Milan University, Milan, Italy; ICU Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - William Stewart
- Institute of Neuroscience and Psychology, University of Glasgow, UK; Department of Laboratory Medicine, Queen Elizabeth University Hospital, Glasgow, UK
| | - Elisa R Zanier
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| |
Collapse
|
20
|
Portbury SD, Hare DJ, Finkelstein DI, Adlard PA. Trehalose improves traumatic brain injury-induced cognitive impairment. PLoS One 2017; 12:e0183683. [PMID: 28837626 PMCID: PMC5570321 DOI: 10.1371/journal.pone.0183683] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/09/2017] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain Injury (TBI) is a significant cause of death and long-term disability for which there are currently no effective pharmacological treatment options. In this study then, we utilized a mouse model of TBI to assess the therapeutic potential of the stable disaccharide trehalose, which is known to protect against oxidative stress, increase levels of chaperone molecules and enhance autophagy. Furthermore, trehalose has demonstrated neuroprotective properties in numerous animal models and has been proposed as a potential treatment for neurodegeneration. As TBI (and associated neurodegenerative disorders) is complicated by a sudden and dramatic change in brain metal concentrations, including iron (Fe) and zinc (Zn), the collective accumulation and translocation of which has been hypothesized to contribute to the pathogenesis of TBI, then we also sought to determine whether trehalose modulated the metal dyshomeostasis associated with TBI. In this study three-month-old C57Bl/6 wildtype mice received a controlled cortical impact TBI, and were subsequently treated for one month with trehalose. During this time animals were assessed on multiple behavioral tasks prior to tissue collection. Results showed an overall significant improvement in the Morris water maze, Y-maze and open field behavioral tests in trehalose-treated mice when compared to controls. These functional benefits occurred in the absence of any change in lesion volume or any significant modulation of biometals, as assessed by laser ablation inductively coupled plasma mass spectrometry. Western blot analysis, however, revealed an upregulation of synaptophysin, doublecortin and brain derived neurotrophic factor protein in trehalose treated mice in the contralateral cortex. These results indicate that trehalose may be efficacious in improving functional outcomes following TBI by a previously undescribed mechanism of action that has relevance to multiple disorders of the central nervous system.
Collapse
Affiliation(s)
- Stuart D. Portbury
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Dominic J. Hare
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- University of Technology Sydney, Elemental Bio-imaging, Sydney, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul A. Adlard
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
21
|
Portbury SD, Hare DJ, Sgambelloni CJ, Bishop DP, Finkelstein DI, Doble PA, Adlard PA. Age modulates the injury-induced metallomic profile in the brain. Metallomics 2017; 9:402-410. [DOI: 10.1039/c6mt00260a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
22
|
Garton T, Keep RF, Hua Y, Xi G. Brain iron overload following intracranial haemorrhage. Stroke Vasc Neurol 2016; 1:172-184. [PMID: 28959481 PMCID: PMC5435218 DOI: 10.1136/svn-2016-000042] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 12/15/2022] Open
Abstract
Intracranial haemorrhages, including intracerebral haemorrhage (ICH), intraventricular haemorrhage (IVH) and subarachnoid haemorrhage (SAH), are leading causes of morbidity and mortality worldwide. In addition, haemorrhage contributes to tissue damage in traumatic brain injury (TBI). To date, efforts to treat the long-term consequences of cerebral haemorrhage have been unsatisfactory. Incident rates and mortality have not showed significant improvement in recent years. In terms of secondary damage following haemorrhage, it is becoming increasingly apparent that blood components are of integral importance, with haemoglobin-derived iron playing a major role. However, the damage caused by iron is complex and varied, and therefore, increased investigation into the mechanisms by which iron causes brain injury is required. As ICH, IVH, SAH and TBI are related, this review will discuss the role of iron in each, so that similarities in injury pathologies can be more easily identified. It summarises important components of normal brain iron homeostasis and analyses the existing evidence on iron-related brain injury mechanisms. It further discusses treatment options of particular promise.
Collapse
Affiliation(s)
- Thomas Garton
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
23
|
Wu X, Kirov II, Gonen O, Ge Y, Grossman RI, Lui YW. MR Imaging Applications in Mild Traumatic Brain Injury: An Imaging Update. Radiology 2016; 279:693-707. [PMID: 27183405 DOI: 10.1148/radiol.16142535] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mild traumatic brain injury (mTBI), also commonly referred to as concussion, affects millions of Americans annually. Although computed tomography is the first-line imaging technique for all traumatic brain injury, it is incapable of providing long-term prognostic information in mTBI. In the past decade, the amount of research related to magnetic resonance (MR) imaging of mTBI has grown exponentially, partly due to development of novel analytical methods, which are applied to a variety of MR techniques. Here, evidence of subtle brain changes in mTBI as revealed by these techniques, which are not demonstrable by conventional imaging, will be reviewed. These changes can be considered in three main categories of brain structure, function, and metabolism. Macrostructural and microstructural changes have been revealed with three-dimensional MR imaging, susceptibility-weighted imaging, diffusion-weighted imaging, and higher order diffusion imaging. Functional abnormalities have been described with both task-mediated and resting-state blood oxygen level-dependent functional MR imaging. Metabolic changes suggesting neuronal injury have been demonstrated with MR spectroscopy. These findings improve understanding of the true impact of mTBI and its pathogenesis. Further investigation may eventually lead to improved diagnosis, prognosis, and management of this common and costly condition. (©) RSNA, 2016.
Collapse
Affiliation(s)
- Xin Wu
- From the Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, 660 First Ave, 4th Floor, New York, NY 10016
| | - Ivan I Kirov
- From the Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, 660 First Ave, 4th Floor, New York, NY 10016
| | - Oded Gonen
- From the Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, 660 First Ave, 4th Floor, New York, NY 10016
| | - Yulin Ge
- From the Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, 660 First Ave, 4th Floor, New York, NY 10016
| | - Robert I Grossman
- From the Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, 660 First Ave, 4th Floor, New York, NY 10016
| | - Yvonne W Lui
- From the Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, 660 First Ave, 4th Floor, New York, NY 10016
| |
Collapse
|
24
|
Portbury SD, Hare DJ, Sgambelloni C, Finkelstein DI, Adlard PA. A time-course analysis of changes in cerebral metal levels following a controlled cortical impact. Metallomics 2016; 8:193-200. [PMID: 26689359 DOI: 10.1039/c5mt00234f] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI) is complicated by a sudden and dramatic change in brain metal levels, including iron (Fe), copper (Cu) and zinc (Zn). Specific 'metallo-pathological' features of TBI include increased non-heme bound Fe and the liberation of free Zn ions, both of which may contribute to the pathogenesis of TBI. To further characterise the metal dyshomeostasis that occurs following brain trauma, we performed a quantitative time-course survey of spatial Fe, Cu and Zn distribution in mice receiving a controlled cortical impact TBI. Images of brain metal levels produced using laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) in the upper quadrant of the ipsilateral hemisphere were compared to the corresponding contralateral hemisphere, together with regional areas radiating toward the center of the brain from the site of lesion. Significant regional and time point specific elevations in Fe, Zn and Cu were detected immediately and up to 28 days after TBI. The magnitude and timeframe of many of these changes suggest that TBI results in a pronounced and sustained alteration in normal metal levels within the brain. Such alterations are likely to play a role in both the short- and long-term consequences of head trauma and suggest that pharmacological modulation to normalize these metal levels may be efficacious in improving functional outcome.
Collapse
Affiliation(s)
- Stuart D Portbury
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria 3052, Australia.
| | - Dominic J Hare
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria 3052, Australia. and Elemental Bio-imaging Facility, University of Technology Sydney, Thomas Street, Broadway, New South Wales 2007, Australia
| | - Charlotte Sgambelloni
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria 3052, Australia.
| | - David I Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria 3052, Australia.
| | - Paul A Adlard
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria 3052, Australia.
| |
Collapse
|
25
|
Straub S, Schneider TM, Emmerich J, Freitag MT, Ziener CH, Schlemmer HP, Ladd ME, Laun FB. Suitable reference tissues for quantitative susceptibility mapping of the brain. Magn Reson Med 2016; 78:204-214. [DOI: 10.1002/mrm.26369] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 06/24/2016] [Accepted: 07/13/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Sina Straub
- Department of Medical Physics in Radiology; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Till M. Schneider
- Department of Neuroradiology; University of Heidelberg; Heidelberg Germany
- Department of Radiology; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Julian Emmerich
- Department of Medical Physics in Radiology; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Martin T. Freitag
- Department of Radiology; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Christian H. Ziener
- Department of Radiology; German Cancer Research Center (DKFZ); Heidelberg Germany
| | | | - Mark E. Ladd
- Department of Medical Physics in Radiology; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Frederik B. Laun
- Department of Medical Physics in Radiology; German Cancer Research Center (DKFZ); Heidelberg Germany
| |
Collapse
|
26
|
Uddin MN, Lebel RM, Seres P, Blevins G, Wilman AH. Spin echo transverse relaxation and atrophy in multiple sclerosis deep gray matter: A two-year longitudinal study. Mult Scler 2016; 22:1133-43. [DOI: 10.1177/1352458515614091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/03/2015] [Indexed: 02/04/2023]
Abstract
Background: Deep gray matter (DGM) is affected in relapsing–remitting multiple sclerosis (RRMS) and may be studied using short-term longitudinal MRI. Objective: To investigate two-year changes in spin-echo transverse relaxation rate (R2) and atrophy in DGM, and its relationship with disease severity in RRMS patients. Methods: Twenty six RRMS patients and 26 matched controls were imaged at 4.7 T. Multiecho spin-echo R2 maps and atrophy measurements were obtained in DGM at baseline and two-year follow-up. Differences between MRI measures and correlations to disease severity were examined. Results: After two years, mean R2 values in the globus pallidus and pulvinar increased by ~4% ( p<0.001) in patients and <1.7% in controls. Two-year changes in R2 showed significant correlation to disease severity in the globus pallidus, pulvinar, substantia nigra, and thalamus. Multiple regression of the two-year R2 difference using these four DGM structures as variables, yielded high correlation with disease severity ( r=0.83, p<0.001). Two-year changes in volume and R2 showed significant correlation only for the globus pallidus in multiple sclerosis (MS) ( p<0.05). Conclusions: Two-year difference R2 measurements in DGM correlate to disease severity in MS. R2 mapping and atrophy measurements over two years can be used to identify changes in DGM in MS.
Collapse
Affiliation(s)
- Md Nasir Uddin
- Department of Biomedical Engineering, University of Alberta, Canada
| | - R Marc Lebel
- Department of Biomedical Engineering, University of Alberta, Canada
| | - Peter Seres
- Department of Biomedical Engineering, University of Alberta, Canada
| | - Gregg Blevins
- Division of Neurology, Department of Medicine, University of Alberta, Canada
| | - Alan H Wilman
- Department of Biomedical Engineering, University of Alberta, Canada
| |
Collapse
|
27
|
Harris NG, Verley DR, Gutman BA, Sutton RL. Bi-directional changes in fractional anisotropy after experiment TBI: Disorganization and reorganization? Neuroimage 2016; 133:129-143. [PMID: 26975556 PMCID: PMC4889542 DOI: 10.1016/j.neuroimage.2016.03.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 11/26/2022] Open
Abstract
The current dogma to explain the extent of injury-related changes following rodent controlled cortical impact (CCI) injury is a focal injury with limited axonal pathology. However, there is in fact good, published histologic evidence to suggest that axonal injury is far more widespread in this model than generally thought. One possibility that might help to explain this is the often-used region-of-interest data analysis approach taken by experimental traumatic brain injury (TBI) diffusion tensor imaging (DTI) or histologic studies that might miss more widespread damage, when compared to the whole brain, statistically robust method of tract-based analysis used more routinely in clinical research. To determine the extent of DTI changes in this model, we acquired in vivo DTI data before and at 1 and 4weeks after CCI injury in 17 adult male rats and analyzed parametric maps of fractional anisotropy (FA), axial, radial, and mean diffusivity (AD, RD, MD), tensor mode (MO), and fiber tract density (FTD) using tract-based spatial statistics. Contusion volume was used as a surrogate marker of injury severity and as a covariate for investigating severity dependence of the data. Mean fiber tract length was also computed from seeds in the cortical spinal tract regions. In parallel experiments (n=3-5/group), we investigated corpus callosum neurofilaments and demyelination using immunohistochemistry (IHC) at 3days and 6weeks, callosal tract patency using dual-label retrograde tract tracing at 5weeks, and the contribution of gliosis to DTI parameter maps using GFAP IHC at 4weeks post-injury. The data show widespread ipsilateral regions of significantly reduced FA at 1week post-injury, driven by temporally changing values of AD, RD, and MD that persist to 4weeks. Demyelination, retrograde label tract loss, and reductions in MO (tract degeneration) and FTD were shown to underpin these data. Significant FA increases occurred in subcortical and corticospinal tract regions that were spatially distinct from regions of FA decrease, grossly affected gliotic areas, and MO changes. However, there was good spatial correspondence between regions of increased FA and areas of increased FTD and mean fiber length. We discuss these widespread changes in DTI parameters in terms of axonal degeneration and potential reorganization, with reference to a resting state fMRI companion paper (Harris et al., 2016, Exp. Neurol. 227:124-138) that demonstrated altered functional connectivity data acquired from the same rats used in this study.
Collapse
Affiliation(s)
- N G Harris
- UCLA Brain injury Research Center, Department of Neurosurgery, University of California, Los Angeles, Los Angeles, USA.
| | - D R Verley
- UCLA Brain injury Research Center, Department of Neurosurgery, University of California, Los Angeles, Los Angeles, USA
| | - B A Gutman
- Department of Neurology, Imaging Genetics Center, Keck/USC School of Medicine, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
| | - R L Sutton
- UCLA Brain injury Research Center, Department of Neurosurgery, University of California, Los Angeles, Los Angeles, USA
| |
Collapse
|
28
|
Iron Deposition Is Positively Related to Cognitive Impairment in Patients with Chronic Mild Traumatic Brain Injury: Assessment with Susceptibility Weighted Imaging. BIOMED RESEARCH INTERNATIONAL 2015; 2015:470676. [PMID: 26798636 PMCID: PMC4698517 DOI: 10.1155/2015/470676] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/21/2015] [Indexed: 01/17/2023]
Abstract
Background. This study aimed to evaluate the usability of SWI in assessment of brain iron to detect cognitive dysfunction in patients with chronic mild traumatic brain injury (mTBI). Methods. 39 patients with mTBI and 37 normal controls were given the Mini-Mental State Examination (MMSE) and underwent SWI scanning at least 6 months after injury. Angle radian values were calculated with phase images. The angle radian values were compared between groups using analysis of covariance, and their association with MMSE scores was analyzed using Spearman correlations. Results. Significantly higher angle radian values (p < 0.05) were found in the head of the caudate nucleus, the lenticular nucleus, the hippocampus, the thalamus, the right substantia nigra, the red nucleus, and the splenium of the corpus callosum (SCC) in the mTBI group, compared to the control group. MMSE scores were negatively correlated with angle radian values in the right substantia nigra (r = −0.685, p < 0.001). Conclusions. Patients with chronic mTBI might have abnormally high accumulations of iron, and their MMSE scores are negatively associated with angle radian values in the right substantia nigra, suggesting a role of SWI in the assessment of cognitive impairments of these patients.
Collapse
|
29
|
Daiutolo BV, Tyburski A, Clark SW, Elliott MB. Trigeminal Pain Molecules, Allodynia, and Photosensitivity Are Pharmacologically and Genetically Modulated in a Model of Traumatic Brain Injury. J Neurotrauma 2015; 33:748-60. [PMID: 26472135 DOI: 10.1089/neu.2015.4087] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pain-signaling molecules, nitric oxide synthase (NOS) and calcitonin gene-related peptide (CGRP), are implicated in the pathophysiology of post-traumatic headache (PTH) as they are for migraine. This study assessed the changes of inducible NOS (iNOS) and its cellular source in the trigeminal pain circuit, as well as the relationship between iNOS and CGRP after controlled cortical impact (CCI) injury in mice. The effects of a CGRP antagonist (MK8825) and sumatriptan on iNOS messenger RNA (mRNA) and protein were compared to vehicle at 2 weeks postinjury. Changes in CGRP levels in the trigeminal nucleus caudalis (TNC) in iNOS knockouts with CCI were compared to wild-type (WT) mice at 3 days and 2 weeks post injury. Trigeminal allodynia and photosensitivity were measured. MK8825 and sumatriptan increased allodynic thresholds in CCI groups compared to vehicle (p < 0.01), whereas iNOS knockouts were not different from WT. Photosensitivity was attenuated in MK8825 mice and iNOS knockouts compared to WT (p < 0.05). MK8825 and sumatriptan reduced levels of iNOS mRNA and iNOS immunoreactivity in the TNC and ganglia (p < 0.01). Differences in iNOS cellular localization were found between the trigeminal ganglia and TNC. Although the knockout of iNOS attenuated CGRP at 3 days (p < 0.05), it did not reduce CGRP at 2 weeks. CGRP immunoreactivity was found in the meningeal layers post-CCI, while negligible in controls. Findings support the importance of interactions between CGRP and iNOS in mediating allodynia, as well as the individual roles in photosensitivity. Mitigating prolonged increases in CGRP may be a promising intervention for treating acute PTH.
Collapse
Affiliation(s)
- Brittany V Daiutolo
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ashley Tyburski
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Shannon W Clark
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Melanie B Elliott
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Hazra A, Macolino C, Elliott MB, Chin J. Delayed thalamic astrocytosis and disrupted sleep-wake patterns in a preclinical model of traumatic brain injury. J Neurosci Res 2014; 92:1434-45. [PMID: 24964253 DOI: 10.1002/jnr.23430] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 05/01/2014] [Accepted: 05/04/2014] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) involves diffuse axonal injury and induces subtle but persistent changes in brain tissue and function and poses challenges for early detection of neurological injury. The present study uses an automated behavioral analysis system to assess alterations in rodent behavior in the subacute phase in a preclinical mouse model of TBI, controlled cortical impact (CCI) injury. In the first few weeks following CCI, mice demonstrated normal exploratory behaviors and other typical home-cage behaviors. However, beginning 4 weeks post-injury, CCI mice developed disruptions in sleep-wake patterns, including an increased number of awakenings from sleep. Such impaired sleep maintenance was accompanied by an increased latency to reach peak sleep in CCI mice. These sleep disruptions implicate involvement of the thalamocortical network, the activity of which must be tightly regulated to control sleep maintenance. After injury, there was an increase in reactive microglia in thalamic regions as well as delayed reactive astrocytosis that was evident in the thalamic reticular nucleus, which preceded the development of sleep disruptions. These data suggest that cortical injury may trigger inflammatory responses in deeper neuroanatomical structures, including the thalamic reticular nucleus. Such engagement of the thalamus may perturb the thalamocortical network that regulates sleep/awake patterns and contribute to sleep disruptions observed in this model as well as those documented in patients with TBI.
Collapse
Affiliation(s)
- Anupam Hazra
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania; Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | | |
Collapse
|
31
|
Zhao J, Chen Z, Xi G, Keep RF, Hua Y. Deferoxamine attenuates acute hydrocephalus after traumatic brain injury in rats. Transl Stroke Res 2014; 5:586-94. [PMID: 24935175 DOI: 10.1007/s12975-014-0353-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 06/05/2014] [Accepted: 06/05/2014] [Indexed: 11/26/2022]
Abstract
Acute post-traumatic ventricular dilation and hydrocephalus are relatively frequent consequences of traumatic brain injury (TBI). Several recent studies have indicated that high iron levels in brain may relate to hydrocephalus development after intracranial hemorrhage. However, the role of iron in the development of post-traumatic hydrocephalus is still unclear. This study was to determine whether or not iron has a role in hydrocephalus development after TBI. TBI was induced by lateral fluid-percussion in male Sprague-Dawley rats. Some rats had intraventricular injection of iron. Acute hydrocephalus was measured by magnetic resonance T2-weighted imaging and brain hemorrhage was determined by T2* gradient-echo sequence imaging and brain hemoglobin levels. The effect of deferoxamine on TBI-induced hydrocephalus was examined. TBI resulted in acute hydrocephalus at 24 h (lateral ventricle volume: 24.1 ± 3.0 vs. 9.9 ± 0.2 mm(3) in sham group). Intraventricular injection of iron also caused hydrocephalus (25.7 ± 3.4 vs. 9.0 ± 0.6 mm(3) in saline group). Deferoxamine treatment attenuated TBI-induced hydrocephalus and heme oxygenase-1 upregulation. In conclusion, iron may contribute to acute hydrocephalus after TBI.
Collapse
Affiliation(s)
- Jinbing Zhao
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | | | | | | | | |
Collapse
|
32
|
Sands SA, Williams R, Marshall S, LeVine SM. Perivascular iron deposits are associated with protein nitration in cerebral experimental autoimmune encephalomyelitis. Neurosci Lett 2014; 582:133-8. [PMID: 24846416 DOI: 10.1016/j.neulet.2014.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/01/2014] [Accepted: 05/02/2014] [Indexed: 12/12/2022]
Abstract
Nitration of proteins, which is thought to be mediated by peroxynitrite, is a mechanism of tissue damage in multiple sclerosis (MS). However, protein nitration can also be catalyzed by iron, heme or heme-associated molecules independent of peroxynitrite. Since microhemorrhages and perivascular iron deposits are present in the CNS of MS patients, we sought to determine if iron is associated with protein nitration. A cerebral model of experimental autoimmune encephalomyelitis (cEAE) was utilized since this model has been shown to have perivascular iron deposits similar to those present in MS. Histochemical staining for iron was used together with immunohistochemistry for nitrotyrosine, eNOS, or iNOS on cerebral sections. Leakage of the blood-brain barrier (BBB) was studied by albumin immunohistochemistry. Iron deposits were colocalized with nitrotyrosine staining around vessels in cEAE mice while control animals revealed minimal staining. This finding supports the likelihood that nitrotyrosine formation was catalyzed by iron or iron containing molecules. Examples of iron deposits were also observed in association with eNOS and iNOS, which could be one source of substrates for this reaction. Extravasation of albumin was present in cEAE mice, but not in control animals. Extravasated albumin may act to limit tissue injury by binding iron and/or heme as well as being a target of nitration, but the protection is incomplete. In summary, iron-catalyzed nitration of proteins is a likely mechanism of tissue damage in MS.
Collapse
Affiliation(s)
- Scott A Sands
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Rachel Williams
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Sylvester Marshall
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Steven M LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA.
| |
Collapse
|
33
|
Macolino CM, Daiutolo BV, Albertson BK, Elliott MB. Mechanical allodynia induced by traumatic brain injury is independent of restraint stress. J Neurosci Methods 2014; 226:139-146. [PMID: 24486873 DOI: 10.1016/j.jneumeth.2014.01.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/20/2013] [Accepted: 01/13/2014] [Indexed: 01/28/2023]
Abstract
BACKGROUND This study identifies the relationship between a test for post-traumatic headache and a marker for acute stress in rodent models of traumatic brain injury. NEW METHOD C57BL/6 mice and Sprague Dawley rats were divided into Controlled Cortical Impact (CCI) injury, craniotomy (CR), and incision groups. Periorbital and paw allodynia were evaluated using the von Frey test prior to injury and up to four weeks post-operatively. Serum corticosterone was evaluated in groups with and without mild restraint. RESULTS Periorbital and forepaw thresholds, but not hindpaw thresholds, were reduced in CCI and CR mice compared to incision (p<0.0001 and p<0.01). In contrast to mice, reduced periorbital and forepaw periorbital thresholds were found in CCI rats but not CR rats compared to incision (p<0.0001). Right periorbital thresholds were reduced compared to left thresholds for both rat and mouse at one week (p<0.01), but there were no side differences for forepaw thresholds. Hindpaw thresholds did not change from baseline values for any groups of mice or rats. In mice serum corticosterone levels were increased at one, two and four weeks post-CCI and CR, while the levels for rats were not different from incision (p<0.0001). Corticosterone levels were not different in mice subjected to restraint compared to no restraint. COMPARISON WITH EXISTING METHODS This study presents novel data for allodynia in a rat model of TBI, and differences among mouse and rat species. CONCLUSIONS Mechanical allodynia occurs independent of evoked restraint stress, while hypothalamic pituitary adrenal axis activity is dependent on head trauma and species.
Collapse
Affiliation(s)
- Christine M Macolino
- Department of Neurological Surgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | - Brittany V Daiutolo
- Department of Neurological Surgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | - Brad K Albertson
- Jefferson Medical College, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | - Melanie B Elliott
- Department of Neuroscience, Jefferson Hospital for Neuroscience Chairs Office 900 Walnut Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
34
|
Nisenbaum EJ, Novikov DS, Lui YW. The presence and role of iron in mild traumatic brain injury: an imaging perspective. J Neurotrauma 2014; 31:301-7. [PMID: 24295521 DOI: 10.1089/neu.2013.3102] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mild traumatic brain injury (mTBI), although often presenting without the gross structural abnormalities seen in more severe forms of brain trauma, can nonetheless result in lingering cognitive and behavioral problems along with subtle alterations in brain structure and function. Repeated injuries are associated with brain atrophy and dementia in the form of chronic traumatic encephalopathy (CTE). The mechanisms underlying these dysfunctions are poorly understood. There is a growing body of evidence that brain iron is abnormal after TBI, and brain iron has also been implicated in a host of neurodegenerative disorders. The purpose of this article is to review evidence about the function of iron in the pathophysiology of mTBI and the role that advanced imaging modalities can play in further elucidating said function. MRI techniques sensitive to field inhomogeneities provide supporting evidence for both deep gray matter non-heme iron accumulation as well as focal microhemorrhage resulting from mTBI. In addition, there is evidence that iron may contribute to pathology after mTBI through a number of mechanisms, including generation of reactive oxygen species (ROS), exacerbation of oxidative stress from other sources, and encouragement of tau phosphorylation and the formation of neurofibrillary tangles. Finally, recent animal studies suggest that iron may serve as a therapeutic target in mitigating the effects of mTBI. However, research on the presence and role of iron in mTBI and CTE is still relatively sparse, and further work is necessary to elucidate issues such as the sources of increased iron and the chain of secondary injury.
Collapse
Affiliation(s)
- Eric J Nisenbaum
- Department of Radiology, NYU Langone Medical Center , New York, New York
| | | | | |
Collapse
|
35
|
LeVine SM, Bilgen M, Lynch SG. Iron accumulation in multiple sclerosis: an early pathogenic event. Expert Rev Neurother 2013; 13:247-50. [PMID: 23448214 DOI: 10.1586/ern.13.14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Iron has been shown to accumulate in deep gray matter structures in many forms of multiple sclerosis (MS), but detecting its presence early in the disease course (e.g., clinically isolated syndrome [CIS]) has been less clear. Here, we review a recent study where MRI scanning at 7 T together with susceptibility mapping was performed to assess iron deposition in CIS and control subjects. Susceptibility indicative of iron deposition was found to be increased in the globus pallidus, caudate, putamen and pulvinar of CIS patients compared with controls. The findings suggest that iron deposition is a pathological change that occurs early in the development of MS. Identifying the mechanisms of iron accumulation and determining whether iron promotes pathogenesis in MS are important areas of future research.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| | | | | |
Collapse
|
36
|
Boulet T, Kelso ML, Othman SF. Long-term in vivo imaging of viscoelastic properties of the mouse brain after controlled cortical impact. J Neurotrauma 2013; 30:1512-20. [PMID: 23534701 DOI: 10.1089/neu.2012.2788] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) presents a variety of causes and symptoms, thus making the development of reliable diagnostic methods and therapeutic treatments challenging. Magnetic resonance elastography (MRE) is a technique that allows for a noninvasive assessment of the mechanical properties of soft biological tissue, such as tissue stiffness, storage modulus, and loss modulus. Importantly, by quantifying the changes in the stiffness of tissue that is often associated with disease, MRE is able to detect tissue pathologies at early stages. Recent improvements in instrumentation have allowed for the investigation of small samples with microscopic resolution (μMRE). We hypothesize that μMRE can sensitively detect variations in micromechanical properties in the brain caused by the compressive and shearing forces sustained during TBI. To test this hypothesis, we randomized 13 C57BL mice to receive a controlled cortical impact at a 0.5 mm or 0.75 mm depth, with both sham and naïve mice as controls. Our objective was to propagate mechanical shear waves throughout the brain for in vivo TBI μMRE imaging. The mechanical properties of the injured brain tissue were determined at days 0, 1, 7, and 28 post-injury. For both groups, we observed a significant drop in the stiffness of the impacted region immediately following the injury; the 0.75 mm animals experienced increased tissue softness that lasted longer than that for the 0.5 mm group. Although the shear stiffness, storage modulus, and loss modulus parameters all followed the same trend, the tissue stiffness yielded the most statistically significant results. Overall, this article introduces a transformative technique for mechanically mapping the brain and detecting brain diseases and injury.
Collapse
Affiliation(s)
- Thomas Boulet
- Department of Engineering Mechanics, University of Nebraska-Lincoln, Lincoln, Nebraksa 43353, USA
| | | | | |
Collapse
|
37
|
Shultz SR, Cardamone L, Liu YR, Hogan RE, Maccotta L, Wright DK, Zheng P, Koe A, Gregoire MC, Williams JP, Hicks RJ, Jones NC, Myers DE, O'Brien TJ, Bouilleret V. Can structural or functional changes following traumatic brain injury in the rat predict epileptic outcome? Epilepsia 2013; 54:1240-50. [PMID: 23718645 DOI: 10.1111/epi.12223] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2013] [Indexed: 02/06/2023]
Abstract
PURPOSE Posttraumatic epilepsy (PTE) occurs in a proportion of traumatic brain injury (TBI) cases, significantly compounding the disability, and risk of injury and death for sufferers. To date, predictive biomarkers for PTE have not been identified. This study used the lateral fluid percussion injury (LFPI) rat model of TBI to investigate whether structural, functional, and behavioral changes post-TBI relate to the later development of PTE. METHODS Adult male Wistar rats underwent LFPI or sham injury. Serial magnetic resonance (MR) and positron emission tomography (PET) imaging, and behavioral analyses were performed over 6 months postinjury. Rats were then implanted with recording electrodes and monitored for two consecutive weeks using video-electroencephalography (EEG) to assess for PTE. Of the LFPI rats, 52% (n = 12) displayed spontaneous recurring seizures and/or epileptic discharges on the video-EEG recordings. KEY FINDINGS MRI volumetric and signal analysis of changes in cortex, hippocampus, thalamus, and amygdala, (18) F-fluorodeoxyglucose (FDG)-PET analysis of metabolic function, and behavioral analysis of cognitive and emotional changes, at 1 week, and 1, 3, and 6 months post-LFPI, all failed to identify significant differences on univariate analysis between the epileptic and nonepileptic groups. However, hippocampal surface shape analysis using large-deformation high-dimensional mapping identified significant changes in the ipsilateral hippocampus at 1 week postinjury relative to baseline that differed between rats that would go onto become epileptic versus those who did not. Furthermore, a multivariate logistic regression model that incorporated the 1 week, and 1 and 3 month (18) F-FDG PET parameters from the ipsilateral hippocampus was able to correctly predict the epileptic outcome in all of the LFPI cases. As such, these subtle changes in the ipsilateral hippocampus at acute phases after LFPI may be related to PTE and require further examination. SIGNIFICANCE These findings suggest that PTE may be independent of major structural, functional, and behavioral changes induced by TBI, and suggest that more subtle abnormalities are likely involved. However, there are limitations associated with studying acquired epilepsies in animal models that must be considered when interpreting these results, in particular the failure to detect differences between the groups may be related to the limitations of properly identifying/separating the epileptic and nonepileptic animals into the correct group.
Collapse
Affiliation(s)
- Sandy R Shultz
- Department of Medicine, RMH, University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Turtzo LC, Budde MD, Gold EM, Lewis BK, Janes L, Yarnell A, Grunberg NE, Watson W, Frank JA. The evolution of traumatic brain injury in a rat focal contusion model. NMR IN BIOMEDICINE 2013; 26:468-479. [PMID: 23225324 PMCID: PMC3596464 DOI: 10.1002/nbm.2886] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 08/28/2012] [Accepted: 10/05/2012] [Indexed: 06/01/2023]
Abstract
Serial MRI facilitates the in vivo analysis of the intra- and intersubject evolution of traumatic brain injury lesions. Despite the availability of MRI, the natural history of experimental focal contusion lesions in the controlled cortical impact (CCI) rat model has not been well described. We performed CCI on rats and MRI during the acute to chronic stages of cerebral injury to investigate the time course of changes in the brain. Female Wistar rats underwent CCI of their left motor cortex with a flat impact tip driven by an electromagnetic piston. In vivo MRI was performed at 7 T serially over 6 weeks post-CCI. The appearances of CCI-induced lesions and lesion-associated cortical volumes were variable on MRI, with the percentage change in cortical volume of the CCI ipsilateral side relative to the contralateral side ranging from 18% within 2 h of injury on day 0 to a peak of 35% on day 1, and a trough of -28% by week 5/6, with an average standard deviation of ± 14% at any given time point. In contrast, the percentage change in cortical volume of the ipsilateral side relative to the contralateral side in control rats was not significant (1 ± 2%). Hemorrhagic conversion within and surrounding the CCI lesion occurred between days 2 and 9 in 45% of rats, with no hemorrhage noted on the initial scan. Furthermore, hemorrhage and hemosiderin within the lesion were positive for Prussian blue and highly autofluorescent on histological examination. Although some variation in injuries may be technique related, the divergence of similar lesions between initial and final scans demonstrates the inherent biological variability of the CCI rat model.
Collapse
Affiliation(s)
- L. Christine Turtzo
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Matthew D. Budde
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Eric M. Gold
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Bobbi K. Lewis
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay Janes
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Angela Yarnell
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Neil E. Grunberg
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - William Watson
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Joseph A. Frank
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
39
|
Acosta SA, Tajiri N, Shinozuka K, Ishikawa H, Grimmig B, Diamond D, Sanberg PR, Bickford PC, Kaneko Y, Borlongan CV. Long-term upregulation of inflammation and suppression of cell proliferation in the brain of adult rats exposed to traumatic brain injury using the controlled cortical impact model. PLoS One 2013; 8:e53376. [PMID: 23301065 PMCID: PMC3536766 DOI: 10.1371/journal.pone.0053376] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 11/27/2012] [Indexed: 11/19/2022] Open
Abstract
The long-term consequences of traumatic brain injury (TBI), specifically the detrimental effects of inflammation on the neurogenic niches, are not very well understood. In the present in vivo study, we examined the prolonged pathological outcomes of experimental TBI in different parts of the rat brain with special emphasis on inflammation and neurogenesis. Sixty days after moderate controlled cortical impact injury, adult Sprague-Dawley male rats were euthanized and brain tissues harvested. Antibodies against the activated microglial marker, OX6, the cell cycle-regulating protein marker, Ki67, and the immature neuronal marker, doublecortin, DCX, were used to estimate microglial activation, cell proliferation, and neuronal differentiation, respectively, in the subventricular zone (SVZ), subgranular zone (SGZ), striatum, thalamus, and cerebral peduncle. Stereology-based analyses revealed significant exacerbation of OX6-positive activated microglial cells in the striatum, thalamus, and cerebral peduncle. In parallel, significant decrements in Ki67-positive proliferating cells in SVZ and SGZ, but only trends of reduced DCX-positive immature neuronal cells in SVZ and SGZ were detected relative to sham control group. These results indicate a progressive deterioration of the TBI brain over time characterized by elevated inflammation and suppressed neurogenesis. Therapeutic intervention at the chronic stage of TBI may confer abrogation of these deleterious cell death processes.
Collapse
Affiliation(s)
- Sandra A. Acosta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Naoki Tajiri
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Kazutaka Shinozuka
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Hiroto Ishikawa
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Bethany Grimmig
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
- James A. Haley Veterans Affairs Hospital, Tampa, Florida, United States of America
| | - David Diamond
- Department of Psychology, University of South Florida, Tampa, Florida, United States of America
| | - Paul R. Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
- Office of Research and Innovation, University of South Florida, Tampa, Florida, United States of America
| | - Paula C. Bickford
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
- James A. Haley Veterans Affairs Hospital, Tampa, Florida, United States of America
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
40
|
Al-Radaideh AM, Wharton SJ, Lim SY, Tench CR, Morgan PS, Bowtell RW, Constantinescu CS, Gowland PA. Increased iron accumulation occurs in the earliest stages of demyelinating disease: an ultra-high field susceptibility mapping study in Clinically Isolated Syndrome. Mult Scler 2012; 19:896-903. [PMID: 23139386 DOI: 10.1177/1352458512465135] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine, using ultra-high field magnetic resonance imaging (MRI), whether changes in iron content occur in the earliest phases of demyelinating disease, by quantifying the magnetic susceptibility of deep grey matter structures in patients with Clinically Isolated Syndrome (CIS) that is suggestive of multiple sclerosis (MS), as compared with age-matched healthy subjects. METHODS We compared 19 CIS patients to 20 age-matched, healthy controls. Scanning of the study subjects was performed on a 7T Philips Achieva system, using a 3-dimensional, T2*-weighted gradient echo acquisition. Phase data were first high-pass filtered, using a dipole fitting method, and then inverted to produce magnetic susceptibility maps. Region of interest (ROI) analysis was used to estimate magnetic susceptibility values for deep grey matter structures (caudate nucleus, putamen, globus pallidus, the thalamus and its pulvinar). RESULTS Significantly increased relative susceptibilities were found in the CIS group, compared with controls, for the caudate nucleus (p = < 0.01), putamen (p < 0.01), globus pallidus (p < 0.01) and pulvinar (p < 0.05). We found no significant nor consistent trends in the relationship between susceptibility and age for either the study controls or CIS patients, in any ROI (r(2) < 0.5; p > 0.05). In CIS patients, the time elapsed since the clinical event and the Expanded Disability Status Scale (EDSS) scores were not correlated with iron levels in any ROI (r(2) < 0.5; p > 0.05); however, a moderate correlation (r(2) = 0.3; p < 0.01) was found between the T1 lesion load and the mean susceptibility of the caudate nucleus. CONCLUSION CIS patients showed an increased iron accumulation, as measured using susceptibility mapping of the deep grey matter, suggesting that iron changes did occur at the earlier stages of CIS disease.
Collapse
Affiliation(s)
- Ali M Al-Radaideh
- Sir Peter Mansfield Magnetic Resonance Centre, University of Nottingham, UK
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Influence of age on brain edema formation, secondary brain damage and inflammatory response after brain trauma in mice. PLoS One 2012; 7:e43829. [PMID: 22952778 PMCID: PMC3431406 DOI: 10.1371/journal.pone.0043829] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 07/30/2012] [Indexed: 01/13/2023] Open
Abstract
After traumatic brain injury (TBI) elderly patients suffer from higher mortality rate and worse functional outcome compared to young patients. However, experimental TBI research is primarily performed in young animals. Aim of the present study was to clarify whether age affects functional outcome, neuroinflammation and secondary brain damage after brain trauma in mice. Young (2 months) and old (21 months) male C57Bl6N mice were anesthetized and subjected to a controlled cortical impact injury (CCI) on the right parietal cortex. Animals of both ages were randomly assigned to 15 min, 24 h, and 72 h survival. At the end of the observation periods, contusion volume, brain water content, neurologic function, cerebral and systemic inflammation (CD3+ T cell migration, inflammatory cytokine expression in brain and lung, blood differential cell count) were determined. Old animals showed worse neurological function 72 h after CCI and a high mortality rate (19.2%) compared to young (0%). This did not correlate with histopathological damage, as contusion volumes were equal in both age groups. Although a more pronounced brain edema formation was detected in old mice 24 hours after TBI, lack of correlation between brain water content and neurological deficit indicated that brain edema formation is not solely responsible for age-dependent differences in neurological outcome. Brains of old naïve mice were about 8% smaller compared to young naïve brains, suggesting age-related brain atrophy with possible decline in plasticity. Onset of cerebral inflammation started earlier and primarily ipsilateral to damage in old mice, whereas in young mice inflammation was delayed and present in both hemispheres with a characteristic T cell migration pattern. Pulmonary interleukin 1β expression was up-regulated after cerebral injury only in young, not aged mice. The results therefore indicate that old animals are prone to functional deficits and strong ipsilateral cerebral inflammation without major differences in morphological brain damage compared to young.
Collapse
|
42
|
Mak CHK, Wong SKH, Wong GK, Ng S, Wang KKW, Lam PK, Poon WS. Traumatic Brain Injury in the Elderly: Is it as Bad as we Think? CURRENT TRANSLATIONAL GERIATRICS AND EXPERIMENTAL GERONTOLOGY REPORTS 2012; 1:171-178. [PMID: 24014175 PMCID: PMC3758513 DOI: 10.1007/s13670-012-0017-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury in elderly patients is a neglected global disease burden. The main cause is fall, followed by motor vehicle accidents. This review article summarizes different aspects of geriatric traumatic brain injury, including epidemiology, pathology, and effects of comorbidities and pre-injury medications such as antiplatelets and anticoagulants. Functional outcome with or without surgical intervention, cognitive outcome, and psychiatric complications are discussed. Animal models are also reviewed in attempt to explain the relationship of aging and outcome, together with advances in stem cell research. Though elderly people in general did fare worse after traumatic brain injury, certain "younger elderly" people, aged 65-75 years, could have a comparable outcome to younger adults after minor to moderate head injury.
Collapse
Affiliation(s)
- Calvin H. K. Mak
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - Stephen K. H. Wong
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - George K. Wong
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - Stephanie Ng
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - Kevin K. W. Wang
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
- Center for Neuroproteomics and Biomarkers Research, The Department of Psychiatry and Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611 USA
| | - Ping Kuen Lam
- Chow Tai Fook-Cheung Yu Tung Surgical Stem Cell Research Center, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| |
Collapse
|
43
|
Valdés Hernández MDC, Maconick LC, Tan EMJ, Wardlaw JM. Identification of mineral deposits in the brain on radiological images: a systematic review. Eur Radiol 2012; 22:2371-81. [PMID: 22688125 DOI: 10.1007/s00330-012-2494-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 03/29/2012] [Accepted: 04/04/2012] [Indexed: 01/11/2023]
Abstract
OBJECTIVES MRI has allowed the study of mineral deposition in the brain throughout life and in disease. However, studies differ in their reporting of minerals on MRI for reasons that are unclear. METHODS We conducted a systematic review from 1985 to July 2011 to determine the appearance of iron, calcium, copper and manganese on MRI and CT and their reliability. We assessed which imaging investigations provided the most consistent results compared with histology. RESULTS Of 325 papers on minerals imaging, we included 46 studies that confirmed findings either directly or indirectly using a non-imaging method such as histology. Within this group, there was inconsistency in the identification of iron probably because of changes in its paramagnetic properties during its degradation. Iron appeared consistently hypointense only on T2*-weighted MRI, and along with calcified areas, hyperattenuated on CT. Appearance of copper, calcium and manganese, although consistently reported as hyperintense on T1-weighted MRI, was confirmed histologically in few studies. On T2-weighted imaging, calcified areas were always reported as hypointense, while the appearance of iron depended on the concentration, location and degradation stage. CONCLUSIONS More work is required to improve the reliability of imaging methods to detect and differentiate brain mineral deposition accurately. KEY POINTS There is inconsistency in reporting the appearance of minerals on radiological images. • Only 46 studies confirmed mineral appearance using a non-imaging method. • Iron is the mineral more widely studied, consistently hypointense on T2*-weighted MRI. • T1-weighted MRI consistently reported copper, calcium and manganese hyperintense. • Calcium is consistently reported hypointense on T2-weighted MRI and hyperattenuating on CT.
Collapse
|
44
|
Williams R, Buchheit CL, Berman NEJ, LeVine SM. Pathogenic implications of iron accumulation in multiple sclerosis. J Neurochem 2011; 120:7-25. [PMID: 22004421 DOI: 10.1111/j.1471-4159.2011.07536.x] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Iron, an essential element used for a multitude of biochemical reactions, abnormally accumulates in the CNS of patients with multiple sclerosis (MS). The mechanisms of abnormal iron deposition in MS are not fully understood, nor do we know whether these deposits have adverse consequences, that is, contribute to pathogenesis. With some exceptions, excess levels of iron are represented concomitantly in multiple deep gray matter structures often with bilateral representation, whereas in white matter, pathological iron deposits are usually located at sites of inflammation that are associated with veins. These distinct spatial patterns suggest disparate mechanisms of iron accumulation between these regions. Iron has been postulated to promote disease activity in MS by various means: (i) iron can amplify the activated state of microglia resulting in the increased production of proinflammatory mediators; (ii) excess intracellular iron deposits could promote mitochondria dysfunction; and (iii) improperly managed iron could catalyze the production of damaging reactive oxygen species (ROS). The pathological consequences of abnormal iron deposits may be dependent on the affected brain region and/or accumulation process. Here, we review putative mechanisms of enhanced iron uptake in MS and address the likely roles of iron in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Rachel Williams
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | |
Collapse
|
45
|
Xu S, Zhuo J, Racz J, Shi D, Roys S, Fiskum G, Gullapalli R. Early microstructural and metabolic changes following controlled cortical impact injury in rat: a magnetic resonance imaging and spectroscopy study. J Neurotrauma 2011; 28:2091-102. [PMID: 21761962 DOI: 10.1089/neu.2010.1739] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Understanding tissue alterations at an early stage following traumatic brain injury (TBI) is critical for injury management and limiting severe consequences from secondary injury. We investigated the early microstructural and metabolic profiles using in vivo diffusion tensor imaging (DTI) and proton magnetic resonance spectroscopy ((1)H MRS) at 2 and 4 h following a controlled cortical impact injury in the rat brain using a 7.0 Tesla animal MRI system and compared profiles to baseline. Significant decrease in mean diffusivity (MD) and increased fractional anisotropy (FA) was found near the impact site (hippocampus and bilateral thalamus; p<0.05) immediately following TBI, suggesting cytotoxic edema. Although the DTI parameters largely normalized on the contralateral side by 4 h, a large inter-individual variation was observed with a trend towards recovery of MD and FA in the ipsilateral hippocampus and a sustained elevation of FA in the ipsilateral thalamus (p<0.05). Significant reduction in metabolite to total creatine ratios of N-acetylaspartate (NAA, p=0.0002), glutamate (p=0.0006), myo-inositol (Ins, p=0.04), phosphocholine and glycerophosphocholine (PCh+GPC, p=0.03), and taurine (Tau, p=0.009) were observed ipsilateral to the injury as early as 2 h, while glutamine concentration increased marginally (p=0.07). These metabolic alterations remained sustained over 4 h after TBI. Significant reductions of Ins (p=0.024) and Tau (p=0.013) and marginal reduction of NAA (p=0.06) were also observed on the contralateral side at 4 h after TBI. Overall our findings suggest significant microstructural and metabolic alterations as early as 2 h following injury. The tendency towards normalization at 4 h from the DTI data and no further metabolic changes at 4 h from MRS suggest an optimal temporal window of about 3 h for interventions that might limit secondary damage to the brain. Results indicate that early assessment of TBI patients using DTI and MRS may provide valuable information on the available treatment window to limit secondary brain damage.
Collapse
Affiliation(s)
- Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Raz E, Jensen JH, Ge Y, Babb JS, Miles L, Reaume J, Grossman RI, Inglese M. Brain iron quantification in mild traumatic brain injury: a magnetic field correlation study. AJNR Am J Neuroradiol 2011; 32:1851-6. [PMID: 21885717 DOI: 10.3174/ajnr.a2637] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Experimental studies have suggested a role for iron accumulation in the pathology of TBI. Magnetic field correlation MR imaging is sensitive to the presence of non-heme iron. The aims of this study are to 1) assess the presence, if any, and the extent of iron deposition in the deep gray matter and regional white matter of patients with mTBI by using MFC MR imaging; and 2) investigate the association of regional brain iron deposition with cognitive and behavioral performance of patients with mTBI. MATERIALS AND METHODS We prospectively enrolled 28 patients with mTBI. Eighteen healthy subjects served as controls. The subjects were administered the Stroop color word test, the Verbal Fluency Task, and the Post-Concussion Symptoms Scale. The MR imaging protocol (on a 3T imager) consisted of conventional brain imaging and MFC sequences. After the calculation of parametric maps, MFC was measured by using a region of interest approach. MFC values across groups were compared by using analysis of covariance, and the relationship of MFC values and neuropsychological tests were evaluated by using Spearman correlations. RESULTS Compared with controls, patients with mTBI demonstrated significant higher MFC values in the globus pallidus (P = .002) and in the thalamus (P = .036). In patients with mTBI, Stroop test scores were associated with the MFC value in frontal white matter (r = -0.38, P = .043). CONCLUSIONS MFC values were significantly elevated in the thalamus and globus pallidus of patients with mTBI, suggesting increased accumulation of iron. This supports the hypothesis that deep gray matter is a site of injury in mTBI and suggests a possible role for iron accumulation in the pathophysiological events after mTBI.
Collapse
Affiliation(s)
- E Raz
- Department of Radiology, New York University School of Medicine, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Williams R, Rohr AM, Wang WT, Choi IY, Lee P, Berman NEJ, Lynch SG, LeVine SM. Iron deposition is independent of cellular inflammation in a cerebral model of multiple sclerosis. BMC Neurosci 2011; 12:59. [PMID: 21699685 PMCID: PMC3128570 DOI: 10.1186/1471-2202-12-59] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 06/23/2011] [Indexed: 11/16/2022] Open
Abstract
Background Perivenular inflammation is a common early pathological feature in multiple sclerosis (MS). A recent hypothesis stated that CNS inflammation is induced by perivenular iron deposits that occur in response to altered blood flow in MS subjects. In order to evaluate this hypothesis, an animal model was developed, called cerebral experimental autoimmune encephalomyelitis (cEAE), which presents with CNS perivascular iron deposits. This model was used to investigate the relationship of iron deposition to inflammation. Methods In order to generate cEAE, mice were given an encephalitogen injection followed by a stereotactic intracerebral injection of TNF-α and IFN-γ. Control animals received encephalitogen followed by an intracerebral injection of saline, or no encephalitogen plus an intracerebral injection of saline or cytokines. Laser Doppler was used to measure cerebral blood flow. MRI and iron histochemistry were used to localize iron deposits. Additional histological procedures were used to localize inflammatory cell infiltrates, microgliosis and astrogliosis. Results Doppler analysis revealed that cEAE mice had a reduction in cerebral blood flow compared to controls. MRI revealed T2 hypointense areas in cEAE animals that spatially correlated with iron deposition around vessels and at some sites of inflammation as detected by iron histochemistry. Vessels with associated iron deposits were distributed across both hemispheres. Mice with cEAE had more iron-labeled vessels compared to controls, but these vessels were not commonly associated with inflammatory cell infiltrates. Some iron-laden vessels had associated microgliosis that was above the background microglial response, and iron deposits were observed within reactive microglia. Vessels with associated astrogliosis were more commonly observed without colocalization of iron deposits. Conclusion The findings indicate that iron deposition around vessels can occur independently of inflammation providing evidence against the hypothesis that iron deposits account for inflammatory cell infiltrates observed in MS.
Collapse
Affiliation(s)
- Rachel Williams
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Swan AA, Chandrashekar R, Beare J, Hoane MR. Preclinical efficacy testing in middle-aged rats: nicotinamide, a novel neuroprotectant, demonstrates diminished preclinical efficacy after controlled cortical impact. J Neurotrauma 2011; 28:431-40. [PMID: 21083416 PMCID: PMC3057203 DOI: 10.1089/neu.2010.1519] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Age is a consistent predictor of poor outcome following traumatic brain injury (TBI). Although the elderly population has one of the highest rates of TBI-related hospitalization and death, few preclinical studies have attempted to model and treat TBI in the aged population. Recent studies have indicated that nicotinamide (NAM), a soluble B-group vitamin, improved functional recovery in experimental models of TBI in young animals. The purpose of the present study was to examine the preclinical efficacy of NAM in middle-aged rats. Groups of middle-aged (14-month-old) rats were assigned to NAM (500 mg/kg or 50 mg/kg) or saline alone (1 mL/kg) treatment conditions, and received unilateral cortical contusion injuries (CCI) and injections at 1 h and 24 h following injury. The animals were tested on a variety of tasks to assess vestibulomotor (tapered beam) and cognitive performance (reference and working memory in the Morris water maze), and were evaluated for lesion size, blood-brain barrier compromise, astrocytic activation, and edema formation. In summary, the preclinical efficacy of NAM as a treatment following CCI in middle-aged rats differs from that previously documented in younger rats; while treatment with 50 mg/kg NAM appeared to have no effect, the 500-mg/kg dose worsened performance in middle-aged animals. Histological indicators demonstrated more nuanced group differences, indicating that NAM may positively impact some of the cellular cascades following injury, but were not substantial enough to improve functional recovery. These findings emphasize the need to examine potential treatments for TBI utilizing non-standard populations, and may explain why so many treatments have failed in clinical trials.
Collapse
Affiliation(s)
- Alicia A Swan
- Restorative Neuroscience Laboratory, Center for Integrative Research for Cognitive and Neural Sciences, Department of Psychology, Southern Illinois University , Carbondale, Illinois 62901, USA
| | | | | | | |
Collapse
|