1
|
Mousavi SR, Shadravanan M, Farrokhi MR, Karimi F, Karbalaei N, Zadeh MA, Naseh M. Exposure to Sunset Yellow FCF since post-weaning causes hippocampal structural changes and memory impairment in the adult rat: The neuroprotective effects of Coenzyme Q10. Int J Dev Neurosci 2024. [PMID: 39520069 DOI: 10.1002/jdn.10385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/28/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND This study aimed to investigate whether exposure to Sunset Yellow FCF (SY) since post-weaning can lead to hippocampal structural changes and memory impairment in adult rat and whether the Coenzyme Q10 (CoQ10) can protect against these adverse effects. METHODS The weanling rats were randomly divided into six groups and were treated daily by oral gavage for 6 weeks, as follows: (I) control group, administered distilled water (0.3 mL/100 g/day); (II) CoQ10 group, received 10 mg/kg/day CoQ10; (III) low SY group, received 2.5 mg/kg/day SY; (IV) high SY group, received 70 mg/kg/day SY; (V) low SY + CoQ10 group; and (VI) high SY + CoQ10 group. At the end of the sixth week, the novel object recognition (NOR) test was conducted to evaluate memory. Then, after sacrificing animals, the cerebral hemispheres were removed for stereological study and evaluation of MDA levels. RESULTS The low and high doses of SY led to significant neuronal loss and a decrease in the volume of the hippocampus (CA1 and DG subregions), as well as increased the MDA level, which was associated with short- and long-term memory impairment. Although, administration of CoQ10 prevented the hippocampal neural loss and volume, and caused a reduction in MDA and improved memory in the low and high SY groups. CONCLUSION It seems that CoQ10 could prevent the neuronal loss and hippocampal atrophy caused by post-weaning exposure to SY through preventing oxidative stress, ultimately improving memory impairment in rats.
Collapse
Affiliation(s)
- Seyed Reza Mousavi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurosurgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Shadravanan
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Reza Farrokhi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurosurgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Karimi
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Karbalaei
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Melika Arzhang Zadeh
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Naseh
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Sabetta Z, Krishna G, Curry-Koski T, Lopez M, Adelson PD, Thomas TC. Sex-dependent temporal changes in astrocyte-vessel interactions following diffuse traumatic brain injury in rats. Front Physiol 2024; 15:1469073. [PMID: 39387100 PMCID: PMC11461938 DOI: 10.3389/fphys.2024.1469073] [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: 07/23/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Traumatic brain injury (TBI) is associated with diffuse axonal injury (DAI), a primary pathology linked to progressive neurodegeneration and neuroinflammation, including chronic astrogliosis, which influences long-term post-TBI recovery and morbidity. Sex-based differences in blood-brain barrier (BBB) permeability increases the risk of accelerated brain aging and early-onset neurodegeneration. However, few studies have evaluated chronic time course of astrocytic responses around cerebrovascular in the context of aging after TBI and sex dependence. We observed increased glial fibrillary acidic protein (GFAP)-labeled accessory processes branching near and connecting with GFAP-ensheathed cortical vessels, suggesting a critical nuance in astrocyte-vessel interactions after TBI. To quantify this observation, male and female Sprague Dawley rats (∼3 months old, n = 5-6/group) underwent either sham surgery or midline fluid percussion injury. Using immunohistochemical analysis, we quantified GFAP-labeled astrocyte primary and accessory processes that contacted GFAP-ensheathed vessels in the somatosensory barrel cortex at 7, 56, and 168 days post-injury (DPI). TBI significantly increased GFAP-positive primary processes at 7 DPI (P < 0.01) in both sexes. At 56 DPI, these vessel-process interactions remained significantly increased exclusively in males (P < 0.05). At 168 DPI, both sexes showed a significant reduction in vessel-process interactions compared to 7 DPI (P < 0.05); however, a modest but significant injury effect reemerged in females (P < 0.05). A similar sex-dependent pattern in the number of accessory processes provides novel evidence of long-term temporal changes in astrocyte-vessel interactions. TBI-induced changes in astrocyte-vessel interactions may indicate chronic BBB vulnerability and processes responsible for early onset vascular and neurodegenerative pathology.
Collapse
Affiliation(s)
- Zackary Sabetta
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- A.T. Still University Kirksville College of Osteopathic Medicine, Kirksville, MO, United States
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Tala Curry-Koski
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Mackenzie Lopez
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - P. David Adelson
- West Virginia University School of Medicine, Rockefeller Neuroscience Institute, Morgantown, WV, United States
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
- Translational Neurotrauma and Neurochemistry, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
- Phoenix VA Healthcare System, Phoenix, AZ, United States
| |
Collapse
|
3
|
Huerta de la Cruz S, Santiago-Castañeda C, Rodríguez-Palma EJ, Rocha L, Sancho M. Lateral fluid percussion injury: A rat model of experimental traumatic brain injury. Methods Cell Biol 2024; 185:197-224. [PMID: 38556449 DOI: 10.1016/bs.mcb.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Traumatic brain injury (TBI) represents one of the leading causes of disability and death worldwide. The annual economic impact of TBI-including direct and indirect costs-is high, particularly impacting low- and middle-income countries. Despite extensive research, a comprehensive understanding of the primary and secondary TBI pathophysiology, followed by the development of promising therapeutic approaches, remains limited. These fundamental caveats in knowledge have motivated the development of various experimental models to explore the molecular mechanisms underpinning the pathogenesis of TBI. In this context, the Lateral Fluid Percussion Injury (LFPI) model produces a brain injury that mimics most of the neurological and systemic aspects observed in human TBI. Moreover, its high reproducibility makes the LFPI model one of the most widely used rodent-based TBI models. In this chapter, we provide a detailed surgical protocol of the LFPI model used to induce TBI in adult Wistar rats. We further highlight the neuroscore test as a valuable tool for the evaluation of TBI-induced sensorimotor consequences and their severity in rats. Lastly, we briefly summarize the current knowledge on the pathological aspects and functional outcomes observed in the LFPI-induced TBI model in rodents.
Collapse
Affiliation(s)
- Saúl Huerta de la Cruz
- Department of Pharmacology, University of Vermont, Burlington, VT, United States; Departamento de Farmacobiología, Cinvestav Sede Sur, Ciudad de México, México.
| | | | - Erick J Rodríguez-Palma
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Sede Sur, Mexico City, Mexico
| | - Luisa Rocha
- Departamento de Farmacobiología, Cinvestav Sede Sur, Ciudad de México, México
| | - Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT, United States; Department of Physiology, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
4
|
Wangler LM, Godbout JP. Microglia moonlighting after traumatic brain injury: aging and interferons influence chronic microglia reactivity. Trends Neurosci 2023; 46:926-940. [PMID: 37723009 PMCID: PMC10592045 DOI: 10.1016/j.tins.2023.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/11/2023] [Accepted: 08/24/2023] [Indexed: 09/20/2023]
Abstract
Most of the individuals who experience traumatic brain injury (TBI) develop neuropsychiatric and cognitive complications that negatively affect recovery and health span. Activation of multiple inflammatory pathways persists after TBI, but it is unclear how inflammation contributes to long-term behavioral and cognitive deficits. One outcome of TBI is microglial priming and subsequent hyper-reactivity to secondary stressors, injuries, or immune challenges that further augment complications. Additionally, microglia priming with aging contributes to exaggerated glial responses to TBI. One prominent inflammatory pathway, interferon (IFN) signaling, is increased after TBI and may contribute to microglial priming and subsequent reactivity. This review discusses the contributions of microglia to inflammatory processes after TBI, as well as the influence of aging and IFNs on microglia reactivity and chronic inflammation after TBI.
Collapse
Affiliation(s)
- Lynde M Wangler
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10th Ave, Columbus, OH, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10th Ave, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, 460 Medical Center Drive, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, 190 North Oval Mall, Columbus, OH, USA.
| |
Collapse
|
5
|
Doust YV, Bindoff A, Holloway OG, Wilson R, King AE, Ziebell JM. Temporal changes in the microglial proteome of male and female mice after a diffuse brain injury using label-free quantitative proteomics. Glia 2023; 71:880-903. [PMID: 36468604 PMCID: PMC10952308 DOI: 10.1002/glia.24313] [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: 05/27/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) triggers neuroinflammatory cascades mediated by microglia, which promotes tissue repair in the short-term. These cascades may exacerbate TBI-induced tissue damage and symptoms in the months to years post-injury. However, the progression of the microglial function across time post-injury and whether this differs between biological sexes is not well understood. In this study, we examined the microglial proteome at 3-, 7-, or 28-days after a midline fluid percussion injury (mFPI) in male and female mice using label-free quantitative proteomics. Data are available via ProteomeXchange with identifier PXD033628. We identified a reduction in microglial proteins involved with clearance of neuronal debris via phagocytosis at 3- and 7-days post-injury. At 28 days post-injury, pro-inflammatory proteins were decreased and anti-inflammatory proteins were increased in microglia. These results indicate a reduction in microglial clearance of neuronal debris in the days post-injury with a shift to anti-inflammatory function by 28 days following TBI. The changes in the microglial proteome that occurred across time post-injury did not differ between biological sexes. However, we did identify an increase in microglial proteins related to pro-inflammation and phagocytosis as well as insulin and estrogen signaling in males compared with female mice that occurred with or without a brain injury. Although the microglial response was similar between males and females up to 28 days following TBI, biological sex differences in the microglial proteome, regardless of TBI, has implications for the efficacy of treatment strategies targeting the microglial response post-injury.
Collapse
Affiliation(s)
- Yasmine V. Doust
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Olivia G. Holloway
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Richard Wilson
- Central Science Laboratory (CSL)University of TasmaniaHobartTasmaniaAustralia
| | - Anna E. King
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Jenna M. Ziebell
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
6
|
Sabetta Z, Krishna G, Curry T, Adelson PD, Thomas TC. Aging with TBI vs. Aging: 6-month temporal profiles for neuropathology and astrocyte activation converge in behaviorally relevant thalamocortical circuitry of male and female rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527058. [PMID: 36798182 PMCID: PMC9934568 DOI: 10.1101/2023.02.06.527058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Traumatic brain injury (TBI) manifests late-onset and persisting clinical symptoms with implications for sex differences and increased risk for the development of age-related neurodegenerative diseases. Few studies have evaluated chronic temporal profiles of neuronal and glial pathology that include sex as a biological variable. After experimental diffuse TBI, late-onset and persisting somatosensory hypersensitivity to whisker stimulation develops at one-month post-injury and persists to at least two months post-injury in male rats, providing an in vivo model to evaluate the temporal profile of pathology responsible for morbidity. Whisker somatosensation is dependent on signaling through the thalamocortical relays of the whisker barrel circuit made up of glutamatergic projections between the ventral posteromedial nucleus of the thalamus (VPM) and primary somatosensory barrel cortex (S1BF) with inhibitory (GABA) innervation from the thalamic reticular nucleus (TRN) to the VPM. To evaluate the temporal profiles of pathology, male and female Sprague Dawley rats ( n = 5-6/group) were subjected to sham surgery or midline fluid percussion injury (FPI). At 7-, 56-, and 168-days post-injury (DPI), brains were processed for amino-cupric silver stain and glial fibrillary acidic protein (GFAP) immunoreactivity, where pixel density of staining was quantified to determine the temporal profile of neuropathology and astrocyte activation in the VPM, S1BF, and TRN. FPI induced significant neuropathology in all brain regions at 7 DPI. At 168 DPI, neuropathology remained significantly elevated in the VPM and TRN, but returned to sham levels in the S1BF. GFAP immunoreactivity was increased as a function of FPI and DPI, with an FPI × DPI interaction in all regions and an FPI × Sex interaction in the S1BF. The interactions were driven by increased GFAP immunoreactivity in shams over time in the VPM and TRN. In the S1BF, GFAP immunoreactivity increased at 7 DPI and declined to age-matched sham levels by 168 DPI, while GFAP immunoreactivity in shams significantly increased between 7 and 168 days. The FPI × Sex interaction was driven by an overall greater level of GFAP immunoreactivity in FPI males compared to FPI females. Increased GFAP immunoreactivity was associated with an increased number of GFAP-positive soma, predominantly at 7 DPI. Overall, these findings indicate that FPI, time post-injury, sex, region, and aging with injury differentially contribute to chronic changes in neuronal pathology and astrocyte activation after diffuse brain injury. Thus, our results highlight distinct patterns of pathological alterations associated with the development and persistence of morbidity that supports chronic neuropathology, especially within the thalamus. Further, data indicate a convergence between TBI-induced and age-related pathology where further investigation may reveal a role for divergent astrocytic phenotypes associated with increased risk for neurodegenerative diseases.
Collapse
|
7
|
Giordano KR, Law LM, Henderson J, Rowe RK, Lifshitz J. Time Course of Remote Neuropathology Following Diffuse Traumatic Brain Injury in the Male Rat. Exp Neurobiol 2022; 31:105-115. [PMID: 35673999 PMCID: PMC9194637 DOI: 10.5607/en21027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/15/2021] [Accepted: 04/12/2022] [Indexed: 11/19/2022] Open
Abstract
Traumatic brain injury (TBI) can affect different regions throughout the brain. Regions near the site of impact are the most vulnerable to injury. However, damage to distal regions occurs. We investigated progressive neuropathology in the dorsal hippocampus (near the impact) and cerebellum (distal to the impact) after diffuse TBI. Adult male rats were subjected to midline fluid percussion injury or sham injury. Brain tissue was stained by the amino cupric silver stain. Neuropathology was quantified in sub-regions of the dorsal hippocampus at 1, 7, and 28 days post-injury (DPI) and coronal cerebellar sections at 1, 2, and 7 DPI. The highest observed neuropathology in the dentate gyrus occurred at 7 DPI which attenuated by 28 DPI, whereas the highest observed neuropathology was at 1 DPI in the CA3 region. There was no significant neuropathology in the CA1 region at any time point. Neuropathology was increased at 7 DPI in the cerebellum compared to shams and stripes of pathology were observed in the molecular layer perpendicular to the cerebellar cortical surface. Together these data show that diffuse TBI can result in neuropathology across the brain. By describing the time course of pathology in response to TBI, it is possible to build the temporal profile of disease progression.
Collapse
Affiliation(s)
- Katherine R Giordano
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - L Matthew Law
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - Jordan Henderson
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA
| | - Rachel K Rowe
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80309, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ 85013, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| |
Collapse
|
8
|
Karimi F, Anari H, Yousefi Nejad A, Karbalay-Doust S, Naseh M. Post-weaning exposure to Sunset Yellow FCF induces behavioral impairment and structural changes in the adult rat medial prefrontal cortex: protective effects of Coenzyme Q10. Int J Dev Neurosci 2022; 82:303-313. [PMID: 35293019 DOI: 10.1002/jdn.10178] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 11/07/2022] Open
Abstract
Sunset Yellow FCF (E110) is a water soluble synthetic dye that has adverse neurobehavioral effects. Coenzyme Q10 (CoQ10) is known as a neuroprotective agent. The present study aimed to evaluate the effects of post-weaning exposure to Sunset Yellow FCF on behavioral and structural changes in the adult rat medial prefrontal cortex and the protective effects of CoQ10. The weanling rats were randomly divided into six groups: distilled water, CoQ10 (10 mg/kg/day), and low (2.5 mg/kg/day), and high (70 mg/kg/day) doses of Sunset Yellow FCF with or without CoQ10 consumption for six weeks. A battery of behavioral tests including open field and Morris water maze tests were done at the end of the 6th week, and then the animals' brains were removed for stereological methods. Our finding indicated that the high dose of Sunset Yellow FCF led to a reduced total volume of mPFC (15.16%), especially in the anterior cingulate cortex (ACC) region (21.96%), along with loss of neurons (32%) and glial cells (37%), which was associated with higher anxiety behavior and loss in spatial memory. However, CoQ10 prevented the neural loss and glial cells, improved anxiety like behaviors and memory impairment. On the other hand, the acceptable daily dose (low dose of Sunset Yellow FCF) did not show a discernible effect on the same parameters. This study showed that the CoQ10 can protect the alteration in mPFC structure and behavioral changes of the rats exposed to high dose of Sunset Yellow FCF.
Collapse
Affiliation(s)
- Fatemeh Karimi
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamideh Anari
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Yousefi Nejad
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saied Karbalay-Doust
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Naseh
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Doust YV, Rowe RK, Adelson PD, Lifshitz J, Ziebell JM. Age-at-Injury Determines the Extent of Long-Term Neuropathology and Microgliosis After a Diffuse Brain Injury in Male Rats. Front Neurol 2021; 12:722526. [PMID: 34566867 PMCID: PMC8455817 DOI: 10.3389/fneur.2021.722526] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/12/2021] [Indexed: 01/30/2023] Open
Abstract
Traumatic brain injury (TBI) can occur at any age, from youth to the elderly, and its contribution to age-related neuropathology remains unknown. Few studies have investigated the relationship between age-at-injury and pathophysiology at a discrete biological age. In this study, we report the immunohistochemical analysis of naïve rat brains compared to those subjected to diffuse TBI by midline fluid percussion injury (mFPI) at post-natal day (PND) 17, PND35, 2-, 4-, or 6-months of age. All brains were collected when rats were 10-months of age (n = 6–7/group). Generalized linear mixed models were fitted to analyze binomial proportion and count data with R Studio. Amyloid precursor protein (APP) and neurofilament (SMI34, SMI32) neuronal pathology were counted in the corpus callosum (CC) and primary sensory barrel field (S1BF). Phosphorylated TAR DNA-binding protein 43 (pTDP-43) neuropathology was counted in the S1BF and hippocampus. There was a significantly greater extent of APP and SMI34 axonal pathology and pTDP-43 neuropathology following a TBI compared with naïves regardless of brain region or age-at-injury. However, age-at-injury did determine the extent of dendritic neurofilament (SMI32) pathology in the CC and S1BF where all brain-injured rats exhibited a greater extent of pathology compared with naïve. No significant differences were detected in the extent of astrocyte activation between brain-injured and naïve rats. Microglia counts were conducted in the S1BF, hippocampus, ventral posteromedial (VPM) nucleus, zona incerta, and posterior hypothalamic nucleus. There was a significantly greater proportion of deramified microglia, regardless of whether the TBI was recent or remote, but this only occurred in the S1BF and hippocampus. The proportion of microglia with colocalized CD68 and TREM2 in the S1BF was greater in all brain-injured rats compared with naïve, regardless of whether the TBI was recent or remote. Only rats with recent TBI exhibited a greater proportion of CD68-positive microglia compared with naive in the hippocampus and posterior hypothalamic nucleus. Whilst, only rats with a remote brain-injury displayed a greater proportion of microglia colocalized with TREM2 in the hippocampus. Thus, chronic alterations in neuronal and microglial characteristics are evident in the injured brain despite the recency of a diffuse brain injury.
Collapse
Affiliation(s)
- Yasmine V Doust
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Rachel K Rowe
- Department of Integrative Physiology at University of Colorado, Boulder, CO, United States.,BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, United States
| | - P David Adelson
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, United States
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, United States.,Phoenix Veteran Affairs Health Care System, Phoenix, AZ, United States
| | - Jenna M Ziebell
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia.,BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, United States
| |
Collapse
|
10
|
Ndode-Ekane XE, Puigferrat Pérez MDM, Di Sapia R, Lapinlampi N, Pitkänen A. Reorganization of Thalamic Inputs to Lesioned Cortex Following Experimental Traumatic Brain Injury. Int J Mol Sci 2021; 22:6329. [PMID: 34199241 PMCID: PMC8231773 DOI: 10.3390/ijms22126329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/27/2021] [Accepted: 06/10/2021] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) disrupts thalamic and cortical integrity. The effect of post-injury reorganization and plasticity in thalamocortical pathways on the functional outcome remains unclear. We evaluated whether TBI causes structural changes in the thalamocortical axonal projection terminals in the primary somatosensory cortex (S1) that lead to hyperexcitability. TBI was induced in adult male Sprague Dawley rats with lateral fluid-percussion injury. A virus carrying the fluorescent-tagged opsin channel rhodopsin 2 transgene was injected into the ventroposterior thalamus. We then traced the thalamocortical pathways and analyzed the reorganization of their axonal terminals in S1. Next, we optogenetically stimulated the thalamocortical relays from the ventral posterior lateral and medial nuclei to assess the post-TBI functionality of the pathway. Immunohistochemical analysis revealed that TBI did not alter the spatial distribution or lamina-specific targeting of projection terminals in S1. TBI reduced the axon terminal density in the motor cortex by 44% and in S1 by 30%. A nematic tensor-based analysis revealed that in control rats, the axon terminals in layer V were orientated perpendicular to the pial surface (60.3°). In TBI rats their orientation was more parallel to the pial surface (5.43°, difference between the groups p < 0.05). Moreover, the level of anisotropy of the axon terminals was high in controls (0.063) compared with TBI rats (0.045, p < 0.05). Optical stimulation of the sensory thalamus increased alpha activity in electroencephalography by 312% in controls (p > 0.05) and 237% (p > 0.05) in TBI rats compared with the baseline. However, only TBI rats showed increased beta activity (33%) with harmonics at 5 Hz. Our findings indicate that TBI induces reorganization of thalamocortical axonal terminals in the perilesional cortex, which alters responses to thalamic stimulation.
Collapse
Affiliation(s)
- Xavier Ekolle Ndode-Ekane
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (M.d.M.P.P.); (R.D.S.); (N.L.); (A.P.)
| | | | | | | | | |
Collapse
|
11
|
Beitchman JA, Lifshitz J, Harris NG, Thomas TC, Lafrenaye AD, Hånell A, Dixon CE, Povlishock JT, Rowe RK. Spatial Distribution of Neuropathology and Neuroinflammation Elucidate the Biomechanics of Fluid Percussion Injury. Neurotrauma Rep 2021; 2:59-75. [PMID: 34223546 PMCID: PMC8240834 DOI: 10.1089/neur.2020.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Diffuse brain injury is better described as multi-focal, where pathology can be found adjacent to seemingly uninjured neural tissue. In experimental diffuse brain injury, pathology and pathophysiology have been reported far more lateral than predicted by the impact site. We hypothesized that local thickening of the rodent skull at the temporal ridges serves to focus the intracranial mechanical forces experienced during brain injury and generate predictable pathology. We demonstrated local thickening of the skull at the temporal ridges using contour analysis on magnetic resonance imaging. After diffuse brain injury induced by midline fluid percussion injury (mFPI), pathological foci along the anterior-posterior length of cortex under the temporal ridges were evident acutely (1, 2, and 7 days) and chronically (28 days) post-injury by deposition of argyophilic reaction product. Area CA3 of the hippocampus and lateral nuclei of the thalamus showed pathological change, suggesting that mechanical forces to or from the temporal ridges shear subcortical regions. A proposed model of mFPI biomechanics suggests that injury force vectors reflect off the skull base and radiate toward the temporal ridge, thereby injuring ventral thalamus, dorsolateral hippocampus, and sensorimotor cortex. Surgically thinning the temporal ridge before injury reduced injury-induced inflammation in the sensorimotor cortex. These data build evidence for temporal ridges of the rodent skull to contribute to the observed pathology, whether by focusing extracranial forces to enter the cranium or intracranial forces to escape the cranium. Pre-clinical investigations can take advantage of the predicted pathology to explore injury mechanisms and treatment efficacy.
Collapse
Affiliation(s)
- Joshua A Beitchman
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA.,Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA.,Midwestern University, Glendale, Arizona, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA.,Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA.,Arizona State University, Tempe, Arizona, USA.,Phoenix VA Health Care System, Phoenix, Arizona, USA
| | - Neil G Harris
- UCLA Brain Injury Research Center, Department of Neurosurgery, and Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Theresa Currier Thomas
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA.,Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA.,Arizona State University, Tempe, Arizona, USA.,Phoenix VA Health Care System, Phoenix, Arizona, USA
| | | | - Anders Hånell
- Virginia Commonwealth University, Richmond, Virginia, USA.,Uppsala University Hospital, Uppsala, Sweden
| | | | | | - Rachel K Rowe
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA.,Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA.,Phoenix VA Health Care System, Phoenix, Arizona, USA
| |
Collapse
|
12
|
Traumatic brain injury metabolome and mitochondrial impact after early stage Ru360 treatment. Mitochondrion 2021; 57:192-204. [PMID: 33484870 DOI: 10.1016/j.mito.2021.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/23/2020] [Accepted: 01/16/2021] [Indexed: 01/10/2023]
Abstract
Ru360, a mitochondrial Ca2+ uptake inhibitor, was tested in a unilateral fluid percussion TBI model in developing rats (P31). Vehicle and Ru360 treated TBI rats underwent sensorimotor behavioral monitoring between 24 and 72 h, thereafter which 185 brain metabolites were analyzed postmortem using LC/MS. Ru360 treatment after TBI improved sensorimotor behavioral recovery, upregulated glycolytic and pentose phosphate pathways, mitigated oxidative stress and prevented NAD+ depletion across both hemispheres. While neural viability improved ipsilaterally, it reduced contralaterally. Ru360 treatment, overall, had a global impact with most benefit near the strongest injury impact areas, while perturbing mitochondrial oxidative energetics in the milder TBI impact areas.
Collapse
|
13
|
Krishna G, Bromberg C, Connell EC, Mian E, Hu C, Lifshitz J, Adelson PD, Thomas TC. Traumatic Brain Injury-Induced Sex-Dependent Changes in Late-Onset Sensory Hypersensitivity and Glutamate Neurotransmission. Front Neurol 2020; 11:749. [PMID: 32849211 PMCID: PMC7419702 DOI: 10.3389/fneur.2020.00749] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/17/2020] [Indexed: 01/15/2023] Open
Abstract
Women approximate one-third of the annual 2.8 million people in the United States who sustain traumatic brain injury (TBI). Several clinical reports support or refute that menstrual cycle-dependent fluctuations in sex hormones are associated with severity of persisting post-TBI symptoms. Previously, we reported late-onset sensory hypersensitivity to whisker stimulation that corresponded with changes in glutamate neurotransmission at 1-month following diffuse TBI in male rats. Here, we incorporated intact age-matched naturally cycling females into the experimental design while monitoring daily estrous cycle. We hypothesized that sex would not influence late-onset sensory hypersensitivity and associated in vivo amperometric extracellular recordings of glutamate neurotransmission within the behaviorally relevant thalamocortical circuit. At 28 days following midline fluid percussion injury (FPI) or sham surgery, young adult Sprague-Dawley rats were tested for hypersensitivity to whisker stimulation using the whisker nuisance task (WNT). As predicted, both male and female rats showed significantly increased sensory hypersensitivity to whisker stimulation after FPI, with females having an overall decrease in whisker nuisance scores (sex effect), but no injury and sex interaction. In males, FPI increased potassium chloride (KCl)-evoked glutamate overflow in primary somatosensory barrel cortex (S1BF) and ventral posteromedial nucleus of the thalamus (VPM), while in females the FPI effect was discernible only within the VPM. Similar to our previous report, we found the glutamate clearance parameters were not influenced by FPI, while a sex-specific effect was evident with female rats showing a lower uptake rate constant both in S1BF and VPM and longer clearance time (in S1BF) in comparison to male rats. Fluctuations in estrous cycle were evident among brain-injured females with longer diestrus (low circulating hormone) phase of the cycle over 28 days post-TBI. Together, these findings add to growing evidence indicating both similarities and differences between sexes in a chronic response to TBI. A better understanding of the influence of gonadal hormones on behavior, neurotransmission, secondary injury and repair processes after TBI is needed both clinically and translationally, with potential impact on acute treatment, rehabilitation, and symptom management.
Collapse
Affiliation(s)
- Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Caitlin Bromberg
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Emily Charlotte Connell
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Erum Mian
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Chengcheng Hu
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ, United States
| | - Jonathan Lifshitz
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Phoenix VA Health Care System, Phoenix, AZ, United States
| | - P. David Adelson
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Phoenix VA Health Care System, Phoenix, AZ, United States
| |
Collapse
|
14
|
A critical review of radiotracers in the positron emission tomography imaging of traumatic brain injury: FDG, tau, and amyloid imaging in mild traumatic brain injury and chronic traumatic encephalopathy. Eur J Nucl Med Mol Imaging 2020; 48:623-641. [DOI: 10.1007/s00259-020-04926-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
|
15
|
Beitchman JA, Griffiths DR, Hur Y, Ogle SB, Bromberg CE, Morrison HW, Lifshitz J, Adelson PD, Thomas TC. Experimental Traumatic Brain Injury Induces Chronic Glutamatergic Dysfunction in Amygdala Circuitry Known to Regulate Anxiety-Like Behavior. Front Neurosci 2020; 13:1434. [PMID: 32038140 PMCID: PMC6985437 DOI: 10.3389/fnins.2019.01434] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/18/2019] [Indexed: 01/01/2023] Open
Abstract
Up to 50% of traumatic brain injury (TBI) survivors demonstrate persisting and late-onset anxiety disorders indicative of limbic system dysregulation, yet the pathophysiology underlying the symptoms is unclear. We hypothesize that the development of TBI-induced anxiety-like behavior in an experimental model of TBI is mediated by changes in glutamate neurotransmission within the amygdala. Adult, male Sprague-Dawley rats underwent midline fluid percussion injury or sham surgery. Anxiety-like behavior was assessed at 7 and 28 days post-injury (DPI) followed by assessment of real-time glutamate neurotransmission in the basolateral amygdala (BLA) and central nucleus of the amygdala (CeA) using glutamate-selective microelectrode arrays. The expression of anxiety-like behavior at 28 DPI coincided with decreased evoked glutamate release and slower glutamate clearance in the CeA, not BLA. Numerous factors contribute to the changes in glutamate neurotransmission over time. In two additional animal cohorts, protein levels of glutamatergic transporters (Glt-1 and GLAST) and presynaptic modulators of glutamate release (mGluR2, TrkB, BDNF, and glucocorticoid receptors) were quantified using automated capillary western techniques at 28 DPI. Astrocytosis and microglial activation have been shown to drive maladaptive glutamate signaling and were histologically assessed over 28 DPI. Alterations in glutamate neurotransmission could not be explained by changes in protein levels for glutamate transporters, mGluR2 receptors, astrocytosis, and microglial activation. Presynaptic modulators, BDNF and TrkB, were significantly decreased at 28 DPI in the amygdala. Dysfunction in presynaptic regulation of glutamate neurotransmission may contribute to anxiety-related behavior and serve as a therapeutic target to improve circuit function.
Collapse
Affiliation(s)
- Joshua A Beitchman
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,College of Graduate Studies, Midwestern University, Glendale, AZ, United States
| | - Daniel R Griffiths
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Yerin Hur
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Sarah B Ogle
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Banner University Medical Center, Phoenix, AZ, United States
| | - Caitlin E Bromberg
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Helena W Morrison
- College of Nursing, University of Arizona, Tucson, AZ, United States
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix VA Health Care System, Phoenix, AZ, United States
| | - P David Adelson
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix VA Health Care System, Phoenix, AZ, United States
| |
Collapse
|
16
|
Krishna G, Beitchman JA, Bromberg CE, Currier Thomas T. Approaches to Monitor Circuit Disruption after Traumatic Brain Injury: Frontiers in Preclinical Research. Int J Mol Sci 2020; 21:ijms21020588. [PMID: 31963314 PMCID: PMC7014469 DOI: 10.3390/ijms21020588] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/03/2020] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
Mild traumatic brain injury (TBI) often results in pathophysiological damage that can manifest as both acute and chronic neurological deficits. In an attempt to repair and reconnect disrupted circuits to compensate for loss of afferent and efferent connections, maladaptive circuitry is created and contributes to neurological deficits, including post-concussive symptoms. The TBI-induced pathology physically and metabolically changes the structure and function of neurons associated with behaviorally relevant circuit function. Complex neurological processing is governed, in part, by circuitry mediated by primary and modulatory neurotransmitter systems, where signaling is disrupted acutely and chronically after injury, and therefore serves as a primary target for treatment. Monitoring of neurotransmitter signaling in experimental models with technology empowered with improved temporal and spatial resolution is capable of recording in vivo extracellular neurotransmitter signaling in behaviorally relevant circuits. Here, we review preclinical evidence in TBI literature that implicates the role of neurotransmitter changes mediating circuit function that contributes to neurological deficits in the post-acute and chronic phases and methods developed for in vivo neurochemical monitoring. Coupling TBI models demonstrating chronic behavioral deficits with in vivo technologies capable of real-time monitoring of neurotransmitters provides an innovative approach to directly quantify and characterize neurotransmitter signaling as a universal consequence of TBI and the direct influence of pharmacological approaches on both behavior and signaling.
Collapse
Affiliation(s)
- Gokul Krishna
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA; (G.K.); (J.A.B.); (C.E.B.)
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Joshua A. Beitchman
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA; (G.K.); (J.A.B.); (C.E.B.)
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Caitlin E. Bromberg
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA; (G.K.); (J.A.B.); (C.E.B.)
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA; (G.K.); (J.A.B.); (C.E.B.)
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Phoenix VA Healthcare System, Phoenix, AZ 85012, USA
- Correspondence: ; Tel.: +1-602-827-2348
| |
Collapse
|
17
|
Pernici CD, Kemp BS, Murray TA. Time course images of cellular injury and recovery in murine brain with high-resolution GRIN lens system. Sci Rep 2019; 9:7946. [PMID: 31138885 PMCID: PMC6538613 DOI: 10.1038/s41598-019-44174-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/10/2019] [Indexed: 12/20/2022] Open
Abstract
Time course, in vivo imaging of brain cells is crucial to fully understand the progression of secondary cellular damage and recovery in murine models of injury. We have combined high-resolution gradient index lens technology with a model of diffuse axonal injury in rodents to enable repeated visualization of fine features of individual cells in three-dimensional space over several weeks. For example, we recorded changes in morphology in the same axons in the external capsule numerous times over 30 to 60 days, before and after induced traumatic brain injury. We observed the expansion of secondary injury and limited recovery of individual axons in this subcortical white matter tract over time. In another application, changes in microglial activation state were visualized in the penumbra region of mice before and after ischemia induced by middle carotid artery occlusion. The ability to collect a series of high-resolution images of cellular features of the same cells pre- and post-injury enables a unique opportunity to study the progression of damage, spontaneous healing, and effects of therapeutics in mouse models of neurodegenerative disease and brain injury.
Collapse
Affiliation(s)
- Chelsea D Pernici
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Benjamin S Kemp
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Teresa A Murray
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, USA.
| |
Collapse
|
18
|
Chitturi J, Li Y, Santhakumar V, Kannurpatti SS. Consolidated Biochemical Profile of Subacute Stage Traumatic Brain Injury in Early Development. Front Neurosci 2019; 13:431. [PMID: 31130841 PMCID: PMC6509949 DOI: 10.3389/fnins.2019.00431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 04/15/2019] [Indexed: 11/30/2022] Open
Abstract
Traumatic brain injury (TBI) in general has varied neuropathological consequences depending upon the intensity and biomechanics of the injury. Furthermore, in pediatric TBI, intrinsic developmental changes add further complexity, necessitating a biochemical dimension for improved TBI characterization. In our earlier study investigating the subacute stage TBI metabolome (72 h post-injury) in a developmental rat model, significant ipsilateral brain biochemical changes occurred across 25 metabolite sets as determined by metabolite set enrichment analysis (MSEA). The broad metabolic perturbation was accompanied by behavioral deficits and neuronal loss across the ipsilateral hemisphere containing the injury epicenter. In order to obtain a consolidated biochemical profile of the TBI assessment, a subgrouping of the 190 identified brain metabolites was performed. Metabolites were divided into seven major subgroups: oxidative energy/mitochondrial, glycolysis/pentose phosphate pathway, fatty acid, amino acid, neurotransmitters/neuromodulators, one-carbon/folate and other metabolites. Subgroups were based on the chemical nature and association with critically altered biochemical pathways after TBI as obtained from our earlier untargeted analysis. Each metabolite subgroup extracted from the ipsilateral sham and TBI brains were modeled using multivariate partial least square discriminant analysis (PLS-DA) with the model accuracy used as a measurable index of TBI neurochemical impact. Volcano plots of each subgroup, corrected for multiple comparisons, determined the TBI neurochemical specificity. The results provide a ranked biochemical profile along with specificity of changes after developmental TBI, enabling a consolidated biochemical template for future classification of different TBI intensities and injury types in animal models.
Collapse
Affiliation(s)
- Jyothsna Chitturi
- Department of Radiology, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Ying Li
- Department of Pharmacology, Physiology & Neuroscience, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology & Neuroscience, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States.,Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Sridhar S Kannurpatti
- Department of Radiology, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
19
|
Rowe RK, Harrison JL, Morrison HW, Subbian V, Murphy SM, Lifshitz J. Acute Post-Traumatic Sleep May Define Vulnerability to a Second Traumatic Brain Injury in Mice. J Neurotrauma 2019; 36:1318-1334. [PMID: 30398389 PMCID: PMC6479254 DOI: 10.1089/neu.2018.5980] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Chronic neurological impairments can manifest from repetitive traumatic brain injury (rTBI), particularly when subsequent injuries occur before the initial injury completely heals. Herein, we apply post-traumatic sleep as a physiological biomarker of vulnerability, hypothesizing that a second TBI during post-traumatic sleep worsens neurological and histological outcomes compared to one TBI or a second TBI after post-traumatic sleep subsides. Mice received sham or diffuse TBI by midline fluid percussion injury; brain-injured mice received one TBI or rTBIs at 3- or 9-h intervals. Over 40 h post-injury, injured mice slept more than shams. Functional assessments indicated lower latencies on rotarod and increased Neurological Severity Scores for mice with rTBIs within 3 h. Anxiety-like behaviors in the open field task were increased for mice with rTBIs at 3 h. Based on pixel density of silver accumulation, neuropathology was greater at 28 days post-injury (DPI) in rTBI groups than sham and single TBI. Cortical microglia morphology was quantified and mice receiving rTBI were de-ramified at 14 DPI compared to shams and mice receiving a single TBI, suggesting robust microglial response in rTBI groups. Orexin-A-positive cells were sustained in the lateral hypothalamus with no loss detected, indicating that loss of wake-promoting neurons did not contribute to post-traumatic sleep. Thus, duration of post-traumatic sleep is a period of vulnerability that results in exacerbated injury from rTBI. Monitoring individual post-traumatic sleep is a potential clinical tool for personalized TBI management, where regular sleep patterns may inform rehabilitative strategies and return-to-activity guidelines.
Collapse
Affiliation(s)
- Rachel K. Rowe
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona
- Department of Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona
- Phoenix Veteran Affairs Health Care System, Phoenix, Arizona
| | - Jordan L. Harrison
- Department of Basic Medical Sciences, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona
| | | | - Vignesh Subbian
- University of Arizona College of Engineering, Tucson, Arizona
| | - Sean M. Murphy
- Department of Forestry and Natural Resources, University of Kentucky, Lexington, Kentucky
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona
- Department of Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona
- Phoenix Veteran Affairs Health Care System, Phoenix, Arizona
| |
Collapse
|
20
|
Chitturi J, Santhakumar V, Kannurpatti SS. Beneficial Effects of Kaempferol after Developmental Traumatic Brain Injury Is through Protection of Mitochondrial Function, Oxidative Metabolism, and Neural Viability. J Neurotrauma 2019; 36:1264-1278. [PMID: 30430900 PMCID: PMC6479259 DOI: 10.1089/neu.2018.6100] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Oxidative energy metabolism is depressed after mild/moderate traumatic brain injury (TBI) during early development, accompanied by behavioral debilitation and secondary neuronal death. A TBI metabolome analysis revealed broad effects with a striking impact on energy metabolism. Our studies on mitochondrial modulators and their effects on brain function have shown that kaempferol, a stimulator of the mitochondrial Ca2+ uniporter channel (mCU), enhanced neural and neurovascular activity in the normal brain and improved stimulus-induced brain activation and behavior after TBI during early development. Because kaempferol enhances mitochondrial Ca2+ uptake and cycling, with protective effects after TBI, we tested the hypothesis that kaempferol treatment during the acute/subacute stage after TBI (0-72 h) acted on mitochondria in improving TBI outcome. Developmental age rats (P31) underwent TBI and were treated with vehicle or kaempferol (1 mg/kg intraperitoneally) in three doses at 1, 24, and 48 h after TBI. Brains were harvested at 72 h and subjected to liquid chromatography mass spectrometric measurements. Decrease in pyruvate and tricarboxylic acid (TCA) cycle flux were observed in the untreated and vehicle-treated group, consistent with previously established energy metabolic decline after TBI. Kaempferol improved TCA cycle flux, maintained mitochondrial functional integrity as observed by decreased acyl carnitines, improved neural viability as evidenced by higher N-acetyl aspartate levels. The positive outcomes of kaempferol on metabolic profile corresponded with improved sensorimotor behavior.
Collapse
Affiliation(s)
- Jyothsna Chitturi
- Department of Radiology, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey
- Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California
| | | |
Collapse
|
21
|
Rod microglia and their role in neurological diseases. Semin Cell Dev Biol 2019; 94:96-103. [PMID: 30826549 DOI: 10.1016/j.semcdb.2019.02.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 12/21/2022]
Abstract
The striking morphology of microglia is one of their most prominent characteristics, with many studies categorising microglial function based on morphology e.g. ramified, hyper-ramified, activated, or amoeboid. Communications regarding rod microglia in neurological disease are scant, and where reported, these cells are rarely the focus of discussion. These factors make it difficult to determine how widespread these cells are not only through the brain but also across diseases. Studies in experimental diffuse brain injury are the first reports of not only significant numbers of rod microglia, but distinct arrangements of these cells, reminiscent of carriages of a train. This review summarises the available reports of rod microglia in vivo and rod-like microglia in vitro and eludes to possible functions and signalling cascades that may evoke this distinct morphology. More investigations are required to fully elucidate the function that rod microglia play in neurological diseases.
Collapse
|
22
|
Parent M, Li Y, Santhakumar V, Hyder F, Sanganahalli BG, Kannurpatti SS. Alterations of Parenchymal Microstructure, Neuronal Connectivity, and Cerebrovascular Resistance at Adolescence after Mild-to-Moderate Traumatic Brain Injury in Early Development. J Neurotrauma 2019; 36:601-608. [PMID: 29855211 PMCID: PMC6354598 DOI: 10.1089/neu.2018.5741] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity in children. To investigate outcome of early developmental TBI during adolescence, a rat model of fluid percussion injury was developed, where previous work reported deficits in sensorimotor behavior and cortical blood flow at adolescence.1 Based on the nonlocalized outcome, we hypothesized that multiple neurophysiological components of brain function, namely neuronal connectivity, synapse/axonal microstructural integrity, and neurovascular function, are altered and magnetic resonance imaging (MRI) methods could be used to determine regional alterations. Adolescent outcomes of developmental TBI were studied 2 months after injury, using functional MRI (fMRI) and diffusion tensor imaging (DTI). fMRI-based resting-state functional connectivity (RSFC), representing neural connectivity, was significantly altered between sham and TBI. RSFC strength decreased in the cortex, hippocampus, and thalamus, accompanied by decrease in spatial extent of their corresponding RSFC networks and interhemispheric asymmetry. Cerebrovascular reactivity to arterial CO2 changes diminished after TBI across both hemispheres, with a more pronounced decrease in the ipsilateral hippocampus, thalamus, and motor cortex. DTI measures of fractional anisotropy and apparent diffusion coefficient, reporting on axonal and microstructural integrity of the brain, indicated similar interhemispheric asymmetry, with highest change in the ipsilateral hippocampus and regions adjoining the ipsilateral thalamus, hypothalamus, and amygdala. TBI-induced corpus callosal microstructural alterations indicated measurable changes in interhemispheric structural connectivity. Hippocampus, thalamus, and select cortical regions were most consistently affected in multiple imaging markers. The multi-modal MRI results demonstrate cortical and subcortical alterations in neural connectivity, cerebrovascular resistance, and parenchymal microstructure in the adolescent brain, indicating the highly diffuse and persistent nature of the lateral fluid percussion TBI early in development.
Collapse
Affiliation(s)
- Maxime Parent
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Ying Li
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical and Health Sciences–New Jersey Medical School, Newark, New Jersey
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical and Health Sciences–New Jersey Medical School, Newark, New Jersey
- Department of Molecular, Cell and Systems Neuroscience, University of California at Riverside, Riverside, California
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Basavaraju G. Sanganahalli
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Sridhar S. Kannurpatti
- Department of Radiology, Rutgers Biomedical and Health Sciences–New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
23
|
Witcher KG, Bray CE, Dziabis JE, McKim DB, Benner BN, Rowe RK, Kokiko-Cochran ON, Popovich PG, Lifshitz J, Eiferman DS, Godbout JP. Traumatic brain injury-induced neuronal damage in the somatosensory cortex causes formation of rod-shaped microglia that promote astrogliosis and persistent neuroinflammation. Glia 2018; 66:2719-2736. [PMID: 30378170 DOI: 10.1002/glia.23523] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 12/21/2022]
Abstract
Microglia undergo dynamic structural and transcriptional changes during the immune response to traumatic brain injury (TBI). For example, TBI causes microglia to form rod-shaped trains in the cerebral cortex, but their contribution to inflammation and pathophysiology is unclear. The purpose of this study was to determine the origin and alignment of rod microglia and to determine the role of microglia in propagating persistent cortical inflammation. Here, diffuse TBI in mice was modeled by midline fluid percussion injury (FPI). Bone marrow chimerism and BrdU pulse-chase experiments revealed that rod microglia derived from resident microglia with limited proliferation. Novel data also show that TBI-induced rod microglia were proximal to axotomized neurons, spatially overlapped with dense astrogliosis, and aligned with apical pyramidal dendrites. Furthermore, rod microglia formed adjacent to hypertrophied microglia, which clustered among layer V pyramidal neurons. To better understand the contribution of microglia to cortical inflammation and injury, microglia were eliminated prior to TBI by CSF1R antagonism (PLX5622). Microglial elimination did not affect cortical neuron axotomy induced by TBI, but attenuated rod microglial formation and astrogliosis. Analysis of 262 immune genes revealed that TBI caused profound cortical inflammation acutely (8 hr) that progressed in nature and complexity by 7 dpi. For instance, gene expression related to complement, phagocytosis, toll-like receptor signaling, and interferon response were increased 7 dpi. Critically, these acute and chronic inflammatory responses were prevented by microglial elimination. Taken together, TBI-induced neuronal injury causes microglia to structurally associate with neurons, augment astrogliosis, and propagate diverse and persistent inflammatory/immune signaling pathways.
Collapse
Affiliation(s)
| | - Chelsea E Bray
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Julia E Dziabis
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Daniel B McKim
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Brooke N Benner
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Rachel K Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, Arizona
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, Arizona
| | | | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| |
Collapse
|
24
|
Chitturi J, Li Y, Santhakumar V, Kannurpatti SS. Early behavioral and metabolomic change after mild to moderate traumatic brain injury in the developing brain. Neurochem Int 2018; 120:75-86. [PMID: 30098378 DOI: 10.1016/j.neuint.2018.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/31/2018] [Accepted: 08/06/2018] [Indexed: 01/30/2023]
Abstract
Pathophysiology of developmental traumatic brain injury (TBI) is unique due to intrinsic differences in the developing brain. Energy metabolic studies of the brain during early development (P13 to P30) have indicated acute oxidative energy metabolic decreases below 24 h after TBI, which generally recovered by 48 h. However, marked neurodegeneration and altered neural functional connectivity have been observed at later stages into adolescence. As secondary neurodegeneration is most prominent during the first week after TBI in the rat model, we hypothesized that the subacute TBI-metabolome may contain predictive markers of neurodegeneration. Sham and TBI metabolomes were examined at 72 h after a mild to moderate intensity TBI in male Sprague-Dawley rats aged P31. Sensorimotor behavior was assessed at 24, 48 and 72 h after injury, followed by 72-hour postmortem brain removal for metabolomics using Liquid Chromatography/Mass Spectrometry (LC-MS) measurement. Broad TBI-induced metabolomic shifts occurred with relatively higher intensity in the injury-lateralized (ipsilateral) hemisphere. Intensity of metabolomic perturbation correlated with the extent of sensorimotor behavioral deficit. N-acetyl-aspartate (NAA) levels at 72 h after TBI, predicted the extent of neurodegeneration assessed histochemically 7-days post TBI. Results from the multivariate untargeted approach clearly distinguished metabolomic shifts induced by TBI. Several pathways including amino acid, fatty acid and energy metabolism continued to be affected at 72 h after TBI, whose collective effects may determine the overall pathological response after TBI in early development including neurodegeneration.
Collapse
Affiliation(s)
- Jyothsna Chitturi
- Department of Radiology, Rutgers New Jersey Medical School, Administrative Complex Building 5 (ADMC5), 30 Bergen Street Room 575, Newark, NJ, 07101, USA.
| | - Ying Li
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, MSB-H-512, 185 S. Orange Ave, Newark, NJ, 07103, USA.
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, MSB-H-512, 185 S. Orange Ave, Newark, NJ, 07103, USA; Molecular, Cell and Systems Biology, University of California Riverside, Spieth 1308, 3401 Watkins Drive, Riverside, CA, 92521, USA.
| | - Sridhar S Kannurpatti
- Department of Radiology, Rutgers New Jersey Medical School, Administrative Complex Building 5 (ADMC5), 30 Bergen Street Room 575, Newark, NJ, 07101, USA.
| |
Collapse
|
25
|
Noorafshan A, Hashemi M, Karbalay-Doust S, Karimi F. High dose Allura Red, rather than the ADI dose, induces structural and behavioral changes in the medial prefrontal cortex of rats and taurine can protect it. Acta Histochem 2018; 120:586-594. [PMID: 30031538 DOI: 10.1016/j.acthis.2018.07.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 07/11/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023]
Abstract
Allura Red is a food color that can lead to neurotoxicity. Taurine is an organic compound that can act as a neuroprotectant. This study aimed to assess the effects of Allura Red with or without taurine consumption on rats' medial Prefrontal Cortex (mPFC). The subjects were divided into six groups as follows: distilled water, taurine (200 mg/kg/day), and low (7 mg/kg/day = acceptable daily dose), and high (70 mg/kg/day) doses of Allura Red with or without taurine consumption for six weeks. The results of novel objects recognition and eight-arm radial maze tests indicated impairment of memory in the Allura Red groups. Subsequently, their brains were analyzed using stereological methods. Both doses of Allura Red caused an increase in working and reference memory errors during the acquisition and retention phases in comparison to the distilled water group (p < 0.01). Additionally, the high dose of Allura Red led to a reduction in the volume of mPFC (35%) and its subdivisions, number of neurons (59%) and glial cells (46%), length of dendrites, and number of spines (mushroom and thin) per dendritic length in comparison to the distilled water group (p < 0.05). The low dose group only showed a reduction in the number of glial cells. However, simultaneous treatment of rats with taurine plus Allura Red prevented the above-mentioned changes. The acceptable daily dose of Allura Red could bring about impairment in spatial learning and memory as well as in the number of glial cells. On the other hand, the high dose of Allura Red could impair learning, memory, and mPFC structure. Thus, taurine could act as a neuroprotectant.
Collapse
|
26
|
Carron SF, Yan EB, Allitt BJ, Rajan R. Immediate and Medium-term Changes in Cortical and Hippocampal Inhibitory Neuronal Populations after Diffuse TBI. Neuroscience 2018; 388:152-170. [PMID: 30036662 DOI: 10.1016/j.neuroscience.2018.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 01/09/2023]
Abstract
Changes in inhibition following traumatic brain injury (TBI) appear to be one of the major factors that contribute to excitation:inhibition imbalance. Neuron pathology, interneurons in particular evolves from minutes to weeks post injury and follows a complex time course. Previously, we showed that in the long-term in diffuse TBI (dTBI), there was select reduction of specific dendrite-targeting neurons in sensory cortex and hippocampus while in motor cortex there was up-regulation of specific dendrite-targeting neurons. We now investigated the time course of dTBI effects on interneurons in neocortex and hippocampus. Brains were labeled with antibodies against calbindin (CB), parvalbumin (PV), calretinin (CR) neuropeptide Y (NPY), and somatostatin (SOM) at 24 h and 2 weeks post dTBI. We found time-dependent, brain area-specific changes in inhibition at 24 h and 2 weeks. At 24 h post-injury, reduction of dendrite-targeting inhibitory neurons occurred in sensory cortex and hippocampus. At 2 weeks, we found compensatory changes in the somatosensory cortex and CA2/3 of hippocampus affected at 24 h, with affected interneuronal populations returning to sham levels. However, DG of hippocampus now showed reduction of dendrite-targeting inhibitory neurons. Finally, with respect to motor cortex, there was an upregulation of dendrite-targeting interneurons in the supragranular layers at 24 h returning to normal levels by 2 weeks. Overall, our findings reconfirm that dendritic inhibition is particularly susceptible to brain trauma, but also show that there are complex brain-area-specific changes in inhibitory neuronal numbers and in compensatory changes, rather than a simple monotonic progression of changes post-dTBI.
Collapse
Affiliation(s)
- Simone F Carron
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| | - Edwin B Yan
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| | - Benjamin J Allitt
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| | - Ramesh Rajan
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
27
|
Thomas TC, Stockhausen EM, Law LM, Khodadad A, Lifshitz J. Rehabilitation modality and onset differentially influence whisker sensory hypersensitivity after diffuse traumatic brain injury in the rat. Restor Neurol Neurosci 2018; 35:611-629. [PMID: 29036852 DOI: 10.3233/rnn-170753] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND As rehabilitation strategies advance as therapeutic interventions, the modality and onset of rehabilitation after traumatic brain injury (TBI) are critical to optimize treatment. Our laboratory has detected and characterized a late-onset, long-lasting sensory hypersensitivity to whisker stimulation in diffuse brain-injured rats; a deficit that is comparable to visual or auditory sensory hypersensitivity in humans with an acquired brain injury. OBJECTIVE We hypothesize that the modality and onset of rehabilitation therapies will differentially influence sensory hypersensitivity in response to the Whisker Nuisance Task (WNT) as well as WNT-induced corticosterone (CORT) stress response in diffuse brain-injured rats and shams. METHODS After midline fluid percussion brain injury (FPI) or sham surgery, rats were assigned to one of four rehabilitative interventions: (1) whisker sensory deprivation during week one or (2) week two or (3) whisker stimulation during week one or (4) week two. At 28 days following FPI and sham procedures, sensory hypersensitivity was assessed using the WNT. Plasma CORT was evaluated immediately following the WNT (aggravated levels) and prior to the pre-determined endpoint 24 hours later (non-aggravated levels). RESULTS Deprivation therapy during week two elicited significantly greater sensory hypersensitivity to the WNT compared to week one (p < 0.05), and aggravated CORT levels in FPI rats were significantly lower than sham levels. Stimulation therapy during week one resulted in low levels of sensory hypersensitivity to the WNT, similar to deprivation therapy and naïve controls, however, non-aggravated CORT levels in FPI rats were significantly higher than sham. CONCLUSION These data indicate that modality and onset of sensory rehabilitation can differentially influence FPI and sham rats, having a lasting impact on behavioral and stress responses to the WNT, emphasizing the necessity for continued evaluation of modality and onset of rehabilitation after TBI.
Collapse
Affiliation(s)
- Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA.,Phoenix VA Healthcare System - Phoenix, AZ, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| | - Ellen Magee Stockhausen
- Core Medical Group, Manchester, NH, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| | - L Matthew Law
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA
| | - Aida Khodadad
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA.,Phoenix VA Healthcare System - Phoenix, AZ, USA.,Neuroscience Program, Arizona State University - Tempe, AZ, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| |
Collapse
|
28
|
Rowe RK, Harrison JL, Zhang H, Bachstetter AD, Hesson DP, O'Hara BF, Greene MI, Lifshitz J. Novel TNF receptor-1 inhibitors identified as potential therapeutic candidates for traumatic brain injury. J Neuroinflammation 2018; 15:154. [PMID: 29789012 PMCID: PMC5964690 DOI: 10.1186/s12974-018-1200-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/13/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) begins with the application of mechanical force to the head or brain, which initiates systemic and cellular processes that are hallmarks of the disease. The pathological cascade of secondary injury processes, including inflammation, can exacerbate brain injury-induced morbidities and thus represents a plausible target for pharmaceutical therapies. We have pioneered research on post-traumatic sleep, identifying that injury-induced sleep lasting for 6 h in brain-injured mice coincides with increased cortical levels of inflammatory cytokines, including tumor necrosis factor (TNF). Here, we apply post-traumatic sleep as a physiological bio-indicator of inflammation. We hypothesized the efficacy of novel TNF receptor (TNF-R) inhibitors could be screened using post-traumatic sleep and that these novel compounds would improve functional recovery following diffuse TBI in the mouse. METHODS Three inhibitors of TNF-R activation were synthesized based on the structure of previously reported TNF CIAM inhibitor F002, which lodges into a defined TNFR1 cavity at the TNF-binding interface, and screened for in vitro efficacy of TNF pathway inhibition (IκB phosphorylation). Compounds were screened for in vivo efficacy in modulating post-traumatic sleep. Compounds were then tested for efficacy in improving functional recovery and verification of cellular mechanism. RESULTS Brain-injured mice treated with Compound 7 (C7) or SGT11 slept significantly less than those treated with vehicle, suggesting a therapeutic potential to target neuroinflammation. SGT11 restored cognitive, sensorimotor, and neurological function. C7 and SGT11 significantly decreased cortical inflammatory cytokines 3 h post-TBI. CONCLUSIONS Using sleep as a bio-indicator of TNF-R-dependent neuroinflammation, we identified C7 and SGT11 as potential therapeutic candidates for TBI.
Collapse
Affiliation(s)
- Rachel K Rowe
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA. .,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA. .,Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA.
| | - Jordan L Harrison
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Hongtao Zhang
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, Spinal Cord and Brain Injury Research Center, and Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - David P Hesson
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - Mark I Greene
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.,Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA.,Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
29
|
de la Tremblaye PB, O'Neil DA, LaPorte MJ, Cheng JP, Beitchman JA, Thomas TC, Bondi CO, Kline AE. Elucidating opportunities and pitfalls in the treatment of experimental traumatic brain injury to optimize and facilitate clinical translation. Neurosci Biobehav Rev 2018; 85:160-175. [PMID: 28576511 PMCID: PMC5709241 DOI: 10.1016/j.neubiorev.2017.05.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/12/2017] [Indexed: 12/19/2022]
Abstract
The aim of this review is to discuss the research presented in a symposium entitled "Current progress in characterizing therapeutic strategies and challenges in experimental CNS injury" which was presented at the 2016 International Behavioral Neuroscience Society annual meeting. Herein we discuss diffuse and focal traumatic brain injury (TBI) and ensuing chronic behavioral deficits as well as potential rehabilitative approaches. We also discuss the effects of stress on executive function after TBI as well as the response of the endocrine system and regulatory feedback mechanisms. The role of the endocannabinoids after CNS injury is also discussed. Finally, we conclude with a discussion of antipsychotic and antiepileptic drugs, which are provided to control TBI-induced agitation and seizures, respectively. The review consists predominantly of published data.
Collapse
Affiliation(s)
- Patricia B de la Tremblaye
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Darik A O'Neil
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Megan J LaPorte
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jeffrey P Cheng
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joshua A Beitchman
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, United States; Midwestern University, Glendale, AZ, United States
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, United States; Phoenix VA Healthcare System, Phoenix, AZ, United States
| | - Corina O Bondi
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Anthony E Kline
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
30
|
Quantitative microglia analyses reveal diverse morphologic responses in the rat cortex after diffuse brain injury. Sci Rep 2017; 7:13211. [PMID: 29038483 PMCID: PMC5643511 DOI: 10.1038/s41598-017-13581-z] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/25/2017] [Indexed: 01/04/2023] Open
Abstract
Determining regions of altered brain physiology after diffuse brain injury is challenging. Microglia, brain immune cells with ramified and dynamically moving processes, constantly surveil the parenchyma for dysfunction which, when present, results in a changed morphology. Our purpose was to define the spatiotemporal changes in microglia morphology over 28 days following rat midline fluid percussion injury (mFPI) as a first step in exploiting microglia morphology to reflect altered brain physiology. Microglia morphology was quantified from histological sections using Image J skeleton and fractal analysis procedures at three time points and in three regions post-mFPI: impact site, primary somatosensory cortex barrel field (S1BF), and a remote region. Microglia ramification (process length/cell and endpoints/cell) decreased in the impact and S1BF but not the remote region (p < 0.05). Microglia complexity was decreased in the S1BF (p = 0.003) and increased in the remote region (p < 0.02). Rod-shaped microglia were present in the S1BF and had a 1.8:1.0 length:width ratio. An in-depth quantitative morphologic analysis revealed diverse and widespread changes to microglia morphology in the cortex post-mFPI. Due to their close link to neuronal function, changes in microglia morphology, summarized in this study, likely reflect altered physiology with diverse and widespread impact on neuronal and circuit function.
Collapse
|
31
|
Ziebell JM, Ray-Jones H, Lifshitz J. Nogo presence is inversely associated with shifts in cortical microglial morphology following experimental diffuse brain injury. Neuroscience 2017; 359:209-223. [PMID: 28736137 DOI: 10.1016/j.neuroscience.2017.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/28/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023]
Abstract
Diffuse traumatic brain injury (TBI) initiates secondary pathology, including inflammation and reduced myelination. Considering these injury-related pathologies, the many states of activated microglia as demonstrated by differing morphologies would form, migrate, and function in and through fields of growth-inhibitory myelin byproduct, specifically Nogo. Here we evaluate the relationship between inflammation and reduced myelin antigenicity in the wake of diffuse TBI and present the hypothesis that the Nogo-66 receptor antagonist peptide NEP(1-40) would reverse the injury-induced shift in distribution of microglia morphologies by limiting myelin-based inhibition. Adult male rats were subjected to midline fluid percussion sham or brain injury. At 2h, 6h, 1d, 2d, 7d, and 21d post-injury, immunohistochemical staining was analyzed in sensory cortex (S1BF) for myelin antigens (myelin basic protein; MBP and CNPase), microglia morphology (ionized calcium-binding adapter protein; Iba1), Nogo receptor and Nogo. Pronounced reduction in myelin antigenicity was evident transiently at 1d post-injury, as evidenced by decreased MBP and CNPase staining, as well as loss of white matter organization, compared to sham and later injury time points. Concomitant with reduced myelin antigenicity, injury shifted microglia morphology from the predominantly ramified morphology observed in sham-injured cortex to hyper-ramified, activated, fully activated, or rod. Changes in microglial morphology were evident as early as 2h post-injury, and remained at least until day 21. Additional cohorts of uninjured and brain-injured animals received vehicle or drug (NEP(1-40), i.p., 15min and 19h post-injury) and brains were collected at 2h, 6h, 1d, 2d, or 7d post-injury. NEP(1-40) administration further shifted distributions of microglia away from an injury-induced activated morphology toward greater proportions of rod and macrophage-like morphologies compared to vehicle-treated. By 7d post-injury, no differences in the distributions of microglia were noted between vehicle and NEP(1-40). This study begins to link secondary pathologies of white matter damage and inflammation after diffuse TBI. In the injured brain, secondary pathologies co-occur and likely interact, with consequences for neuronal circuit disruption leading to neurological symptoms.
Collapse
Affiliation(s)
- Jenna M Ziebell
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.
| | - Helen Ray-Jones
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Department of Biology and Biochemistry, University of Bath, Bath, England, UK
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; VA Healthcare System, Phoenix, AZ, USA; Psychology, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
32
|
Using vitamin E to prevent the impairment in behavioral test, cell loss and dendrite changes in medial prefrontal cortex induced by tartrazine in rats. Acta Histochem 2017; 119:172-180. [PMID: 28126192 DOI: 10.1016/j.acthis.2017.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 01/14/2017] [Accepted: 01/18/2017] [Indexed: 02/07/2023]
Abstract
Tartrazine is a food color that may adversely affect the nervous system. Vitamin E is a neuro-protective agent. This study aimed to evaluate the effects of tartrazine and vitamin E on the performance of rats in memory and learning tests as well as the structure of medial Prefrontal Cortex (mPFC). The rats were first divided into seven groups which received the followings for a period of seven weeks: distilled water, corn oil, vitamin E (100mg/kg/day), a low dose (50mg/kg/day) and a high dose (50mg/kg/day) of tartrazine with and without vitamin E. Behavioral tests were conducted and the brain was extracted for stereological methods The high dose of tartrazine decreased the exploration time of novel objects (P<0.01). The low and high doses of tartrazine led into an increase in working and reference memory errors in acquisition and retention phases (eight-arm radial maze) compared to distilled water group (P<0.01). Additionally, the high dose of tartrazine induced a reduction in the volume of mPFC (∼13%) and its subdivision. Not only that, but the number of neurons and glial cells (∼14%) as well as the mushroom and thin spines per dendrite length declined. The length of dendrites per neuron also reduced in comparison to the distilled water group (P<0.01). Nonetheless, concomitant treatment of the rats with vitamin E plus tartrazine prevented the above-mentioned changes. An acceptable daily dose of tartrazine could induce impairment in spatial memory and dendrite structure. Moreover, a high dose of tartrazine may defect the visual memory, mPFC structure, the spatial memory and also cause dendrite changes. Vitamin E could prevent the behavioral and structural changes.
Collapse
|
33
|
Clausen F, Hansson HA, Raud J, Marklund N. Intranasal Administration of the Antisecretory Peptide AF-16 Reduces Edema and Improves Cognitive Function Following Diffuse Traumatic Brain Injury in the Rat. Front Neurol 2017; 8:39. [PMID: 28261150 PMCID: PMC5306199 DOI: 10.3389/fneur.2017.00039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/27/2017] [Indexed: 12/19/2022] Open
Abstract
A synthetic peptide with antisecretory activity, antisecretory factor (AF)-16, improves injury-related deficits in water and ion transport and decreases intracranial pressure after experimental cold lesion injury and encephalitis although its role in traumatic brain injury (TBI) is unknown. AF-16 or an inactive reference peptide was administrated intranasally 30 min following midline fluid percussion injury (mFPI; n = 52), a model of diffuse mild-moderate TBI in rats. Sham-injured (n = 14) or naïve (n = 24) animals were used as controls. The rats survived for either 48 h or 15 days post-injury. At 48 h, the animals were tested in the Morris water maze (MWM) for memory function and their brains analyzed for cerebral edema. Here, mFPI-induced brain edema compared to sham or naïve controls that was significantly reduced by AF-16 treatment (p < 0.05) although MWM performance was not altered. In the 15-day survival groups, the MWM learning and memory abilities as well as histological changes were analyzed. AF-16-treated brain-injured animals shortened both MWM latency and swim path in the learning trials (p < 0.05) and improved probe trial performance compared to brain-injured controls treated with the inactive reference peptide. A modest decrease by AF-16 on TBI-induced changes in hippocampal glial acidic fibrillary protein (GFAP) staining (p = 0.11) was observed. AF-16 treatment did not alter any other immunohistochemical analyses (degenerating neurons, beta-amyloid precursor protein (β-APP), and Olig2). In conclusion, intranasal AF-16-attenuated brain edema and enhanced visuospatial learning and memory following diffuse TBI in the rat. Intranasal administration early post-injury of a promising neuroprotective substance offers a novel treatment approach for TBI.
Collapse
Affiliation(s)
- Fredrik Clausen
- Unit for Neurosurgery, Department of Neuroscience, Uppsala University , Uppsala , Sweden
| | - Hans-Arne Hansson
- Institute of Biomedicine, University of Gothenburg , Göteborg , Sweden
| | - Johan Raud
- Lantmännen AS Faktor AB , Stockholm , Sweden
| | - Niklas Marklund
- Unit for Neurosurgery, Department of Neuroscience, Uppsala University , Uppsala , Sweden
| |
Collapse
|
34
|
Ziebell JM, Rowe RK, Muccigrosso MM, Reddaway JT, Adelson PD, Godbout JP, Lifshitz J. Aging with a traumatic brain injury: Could behavioral morbidities and endocrine symptoms be influenced by microglial priming? Brain Behav Immun 2017; 59:1-7. [PMID: 26975888 DOI: 10.1016/j.bbi.2016.03.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/01/2016] [Accepted: 03/11/2016] [Indexed: 12/20/2022] Open
Abstract
A myriad of factors influence the developmental and aging process and impact health and life span. Mounting evidence indicates that brain injury, even moderate injury, can lead to lifetime of physical and mental health symptoms. Therefore, the purpose of this mini-review is to discuss how recovery from traumatic brain injury (TBI) depends on age-at-injury and how aging with a TBI affects long-term recovery. TBI initiates pathophysiological processes that dismantle circuits in the brain. In response, reparative and restorative processes reorganize circuits to overcome the injury-induced damage. The extent of circuit dismantling and subsequent reorganization depends as much on the initial injury parameters as other contributing factors, such as genetics and age. Age-at-injury influences the way the brain is able to repair itself, as a result of developmental status, extent of cellular senescence, and injury-induced inflammation. Moreover, endocrine dysfunction can occur with TBI. Depending on the age of the individual at the time of injury, endocrine dysfunction may disrupt growth, puberty, influence social behaviors, and possibly alter the inflammatory response. In turn, activation of microglia, the brain's immune cells, after injury may continue to fuel endocrine dysfunction. With age, the immune system develops and microglia become primed to subsequent challenges. Sustained inflammation and microglial activation can continue for weeks to months post-injury. This prolonged inflammation can influence developmental processes, behavioral performance and age-related decline. Overall, brain injury may influence the aging process and expedite glial and neuronal alterations that impact mental health.
Collapse
Affiliation(s)
- Jenna M Ziebell
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.
| | - Rachel K Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Neuroscience Graduate Program, Arizona State University, Tempe, AZ, USA
| | | | - Jack T Reddaway
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; University of Bath, Department of Biology and Biochemistry, Bath, United Kingdom
| | - P David Adelson
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; University of Bath, Department of Biology and Biochemistry, Bath, United Kingdom
| | - Jonathan P Godbout
- Department of Neuroscience, Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Neuroscience Graduate Program, Arizona State University, Tempe, AZ, USA; VA Healthcare System, Phoenix, AZ, USA
| |
Collapse
|
35
|
Does time heal all wounds? Experimental diffuse traumatic brain injury results in persisting histopathology in the thalamus. Behav Brain Res 2016; 340:137-146. [PMID: 28042008 DOI: 10.1016/j.bbr.2016.12.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/30/2016] [Accepted: 12/28/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND Thalamic dysfunction has been implicated in overall chronic neurological dysfunction after traumatic brain injury (TBI), however little is known about the underlying histopathology. In experimental diffuse TBI (dTBI), we hypothesize that persisting histopathological changes in the ventral posteromedial (VPM) nucleus of the thalamus is indicative of progressive circuit reorganization. Since circuit reorganization in the VPM impacts the whisker sensory system, the histopathology could explain the development of hypersensitivity to whisker stimulation by 28days post-injury; similar to light and sound hypersensitivity in human TBI survivors. METHODS Adult, male Sprague-Dawley rats underwent craniotomy and midline fluid percussion injury (FPI) (moderate severity; 1.8-2.0atm) or sham surgery. At 1d, 7d, and 28days post-FPI (d FPI) separate experiments confirmed the cytoarchitecture (Giemsa stain) and evaluated neuropathology (silver stain), activated astrocytes (GFAP), neuron morphology (Golgi stain) and microglial morphology (Iba-1) in the VPM. RESULTS Cytoarchitecture was unchanged throughout the time course, similar to previously published data; however, neuropathology and astrocyte activation were significantly increased at 7d and 28d and activated microglia were present at all time points. Neuron morphology was dynamic over the time course with decreased dendritic complexity (fewer branch points; decreased length of processes) at 7d FPI and return to sham values by 28d FPI. CONCLUSIONS These data indicate that dTBI results in persisting thalamic histopathology out to a chronic time point. While these changes can be indicative of either adaptive (recovery) or maladaptive (neurological dysfunction) circuit reorganization, they also provide a potential mechanism by which maladaptive circuit reorganization could contribute to the development of chronic neurological dysfunction. Understanding the processes that mediate circuit reorganization is critical to the development of future therapies for TBI patients.
Collapse
|
36
|
Allitt BJ, Iva P, Yan EB, Rajan R. Hypo-excitation across all cortical laminae defines intermediate stages of cortical neuronal dysfunction in diffuse traumatic brain injury. Neuroscience 2016; 334:290-308. [PMID: 27530700 DOI: 10.1016/j.neuroscience.2016.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/06/2016] [Accepted: 08/08/2016] [Indexed: 01/18/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of morbidity and mortality world-wide and can result in persistent cognitive, sensory and behavioral dysfunction. Understanding the time course of TBI-induced pathology is essential to effective treatment outcomes. We induced TBI in rats using an impact acceleration method and tested for sensorimotor skill and sensory sensitivity behaviors for two weeks to find persistently poor outcomes post-injury. At two weeks post-injury we made high resolution extracellular recordings from barrel cortex neurons, to simple and complex whisker deflections. We found that the supragranular suppression of neural firing (compared to normal) previously seen in the immediate post-TBI aftermath had spread to include suppression of input and infragranular layers at two weeks post-injury; thus, there was suppression of whisker-driven firing rates in all cortical layers to both stimulus types. Further, there were abnormalities in temporal response patterns such that in layers 3-5 there was a temporal broadening of response patterns in response to both whisker deflection stimulus types and in L2 a narrowing of temporal patterns in response to the complex stimulus. Thus, at two weeks post-TBI, supragranular hypo-excitation has evolved to include deep cortical layers likely as a function of progressive atrophy and neurodegeneration. These results are consistent with the hypothesis that TBI alters the delicate excitatory/inhibitory balance in cortex and likely contributes to temporal broadening of responses and restricts the ability to code for complex sensory stimuli.
Collapse
Affiliation(s)
- Benjamin J Allitt
- Department of Physiology, Monash University, Clayton, VIC, Australia.
| | - Pippa Iva
- Department of Physiology, Monash University, Clayton, VIC, Australia.
| | - Edwin B Yan
- Department of Physiology, Monash University, Clayton, VIC, Australia.
| | - Ramesh Rajan
- Department of Physiology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
37
|
Hoffman AN, Paode PR, May HG, Ortiz JB, Kemmou S, Lifshitz J, Conrad CD, Currier Thomas T. Early and Persistent Dendritic Hypertrophy in the Basolateral Amygdala following Experimental Diffuse Traumatic Brain Injury. J Neurotrauma 2016; 34:213-219. [PMID: 27306143 DOI: 10.1089/neu.2015.4339] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In the pathophysiology of traumatic brain injury (TBI), the amygdala remains understudied, despite involvement in processing emotional and stressful stimuli associated with anxiety disorders, such as post-traumatic stress disorder (PTSD). Because the basolateral amygdala (BLA) integrates inputs from sensory and other limbic structures coordinating emotional learning and memory, injury-induced changes in circuitry may contribute to psychiatric sequelae of TBI. This study quantified temporal changes in dendritic complexity of BLA neurons after experimental diffuse TBI, modeled by midline fluid percussion injury. At post-injury days (PIDs) 1, 7, and 28, brain tissue from sham and brain-injured adult, male rats was processed for Golgi, glial fibrillary acidic protein (GFAP), or silver stain and analyzed to quantify BLA dendritic branch intersections, activated astrocytes, and regional neuropathology, respectively. Compared to sham, brain-injured rats at all PIDs showed enhanced dendritic branch intersections in both pyramidal and stellate BLA neuronal types, as evidenced by Sholl analysis. GFAP staining in the BLA was significantly increased at PID1 and 7 in comparison to sham. However, the BLA was relatively spared from neuropathology, demonstrated by an absence of argyrophilic accumulation over time, in contrast to other brain regions. These data suggest an early and persistent enhancement of dendritic complexity within the BLA after a single diffuse TBI. Increased dendritic complexity would alter information processing into and through the amygdala, contributing to emotional symptoms post-TBI, including PTSD.
Collapse
Affiliation(s)
- Ann N Hoffman
- 1 Department of Psychology, Arizona State University , Tempe, Arizona.,5 Department of Psychology, UCLA , Los Angeles, California.,6 Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA , Los Angeles, California
| | - Pooja R Paode
- 1 Department of Psychology, Arizona State University , Tempe, Arizona
| | - Hazel G May
- 2 Department of Child Health, University of Arizona College of Medicine-Phoenix , Phoenix, Arizona.,3 Barrow Neurological Institute at Phoenix Children's Hospital , Phoenix, Arizona.,7 Department of Biology and Biochemistry, University of Bath , Bath, United Kingdom
| | - J Bryce Ortiz
- 1 Department of Psychology, Arizona State University , Tempe, Arizona
| | - Salma Kemmou
- 1 Department of Psychology, Arizona State University , Tempe, Arizona
| | - Jonathan Lifshitz
- 1 Department of Psychology, Arizona State University , Tempe, Arizona.,2 Department of Child Health, University of Arizona College of Medicine-Phoenix , Phoenix, Arizona.,3 Barrow Neurological Institute at Phoenix Children's Hospital , Phoenix, Arizona.,4 Phoenix VA Healthcare System , Phoenix, Arizona
| | - Cheryl D Conrad
- 1 Department of Psychology, Arizona State University , Tempe, Arizona
| | - Theresa Currier Thomas
- 2 Department of Child Health, University of Arizona College of Medicine-Phoenix , Phoenix, Arizona.,3 Barrow Neurological Institute at Phoenix Children's Hospital , Phoenix, Arizona.,4 Phoenix VA Healthcare System , Phoenix, Arizona
| |
Collapse
|
38
|
Rowe RK, Rumney BM, May HG, Permana P, Adelson PD, Harman SM, Lifshitz J, Thomas TC. Diffuse traumatic brain injury affects chronic corticosterone function in the rat. Endocr Connect 2016; 5:152-66. [PMID: 27317610 PMCID: PMC5002959 DOI: 10.1530/ec-16-0031] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/17/2016] [Indexed: 01/02/2023]
Abstract
As many as 20-55% of patients with a history of traumatic brain injury (TBI) experience chronic endocrine dysfunction, leading to impaired quality of life, impaired rehabilitation efforts and lowered life expectancy. Endocrine dysfunction after TBI is thought to result from acceleration-deceleration forces to the brain within the skull, creating enduring hypothalamic and pituitary neuropathology, and subsequent hypothalamic-pituitary endocrine (HPE) dysfunction. These experiments were designed to test the hypothesis that a single diffuse TBI results in chronic dysfunction of corticosterone (CORT), a glucocorticoid released in response to stress and testosterone. We used a rodent model of diffuse TBI induced by midline fluid percussion injury (mFPI). At 2months postinjury compared with uninjured control animals, circulating levels of CORT were evaluated at rest, under restraint stress and in response to dexamethasone, a synthetic glucocorticoid commonly used to test HPE axis regulation. Testosterone was evaluated at rest. Further, we assessed changes in injury-induced neuron morphology (Golgi stain), neuropathology (silver stain) and activated astrocytes (GFAP) in the paraventricular nucleus (PVN) of the hypothalamus. Resting plasma CORT levels were decreased at 2months postinjury and there was a blunted CORT increase in response to restraint induced stress. No changes in testosterone were measured. These changes in CORT were observed concomitantly with altered complexity of neuron processes in the PVN over time, devoid of neuropathology or astrocytosis. Results provide evidence that a single moderate diffuse TBI leads to changes in CORT function, which can contribute to the persistence of symptoms related to endocrine dysfunction. Future experiments aim to evaluate additional HP-related hormones and endocrine circuit pathology following diffuse TBI.
Collapse
Affiliation(s)
- Rachel K Rowe
- Phoenix Veterans Affairs Health Care SystemPhoenix, Arizona, USA BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA
| | - Benjamin M Rumney
- BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA Department of Biology and BiochemistryUniversity of Bath, UK
| | - Hazel G May
- BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA Department of Biology and BiochemistryUniversity of Bath, UK
| | - Paska Permana
- Phoenix Veterans Affairs Health Care SystemPhoenix, Arizona, USA
| | - P David Adelson
- BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA School of Biological and Health Systems EngineeringArizona State University, Tempe, Arizona, USA
| | | | - Jonathan Lifshitz
- Phoenix Veterans Affairs Health Care SystemPhoenix, Arizona, USA BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA
| | - Theresa C Thomas
- Phoenix Veterans Affairs Health Care SystemPhoenix, Arizona, USA BARROW Neurological Institute at Phoenix Children's HospitalPhoenix, Arizona, USA Department of Child HealthUniversity of Arizona College of Medicine - Phoenix, Phoenix, Arizona, USA
| |
Collapse
|
39
|
Alwis DS, Yan EB, Johnstone V, Carron S, Hellewell S, Morganti-Kossmann MC, Rajan R. Environmental Enrichment Attenuates Traumatic Brain Injury: Induced Neuronal Hyperexcitability in Supragranular Layers of Sensory Cortex. J Neurotrauma 2016; 33:1084-101. [DOI: 10.1089/neu.2014.3774] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Dasuni Sathsara Alwis
- Department of Physiology, Monash University, Clayton, VIC, Australia
- National Trauma Research Institute, Alfred Hospital, Prahran, VIC, Australia
| | - Edwin Bingbing Yan
- National Trauma Research Institute, Alfred Hospital, Prahran, VIC, Australia
| | | | - Simone Carron
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Sarah Hellewell
- National Trauma Research Institute, Alfred Hospital, Prahran, VIC, Australia
| | | | - Ramesh Rajan
- Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
40
|
Carron SF, Yan EB, Alwis DS, Rajan R. Differential susceptibility of cortical and subcortical inhibitory neurons and astrocytes in the long term following diffuse traumatic brain injury. J Comp Neurol 2016; 524:3530-3560. [PMID: 27072754 DOI: 10.1002/cne.24014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 02/02/2023]
Abstract
Long-term diffuse traumatic brain injury (dTBI) causes neuronal hyperexcitation in supragranular layers in sensory cortex, likely through reduced inhibition. Other forms of TBI affect inhibitory interneurons in subcortical areas but it is unknown if this occurs in cortex, or in any brain area in dTBI. We investigated dTBI effects on inhibitory neurons and astrocytes in somatosensory and motor cortex, and hippocampus, 8 weeks post-TBI. Brains were labeled with antibodies against calbindin (CB), parvalbumin (PV), calretinin (CR) and neuropeptide Y (NPY), and somatostatin (SOM) and glial fibrillary acidic protein (GFAP), a marker for astrogliosis during neurodegeneration. Despite persistent behavioral deficits in rotarod performance up to the time of brain extraction (TBI = 73.13 ± 5.23% mean ± SEM, Sham = 92.29 ± 5.56%, P < 0.01), motor cortex showed only a significant increase, in NPY neurons in supragranular layers (mean cells/mm2 ± SEM, Sham = 16 ± 0.971, TBI = 25 ± 1.51, P = 0.001). In somatosensory cortex, only CR+ neurons showed changes, being decreased in supragranular (TBI = 19 ± 1.18, Sham = 25 ± 1.10, P < 0.01) and increased in infragranular (TBI = 28 ± 1.35, Sham = 24 ± 1.07, P < 0.05) layers. Heterogeneous changes were seen in hippocampal staining: CB+ decreased in dentate gyrus (TBI = 2 ± 0.382, Sham = 4 ± 0.383, P < 0.01), PV+ increased in CA1 (TBI = 39 ± 1.26, Sham = 33 ± 1.69, P < 0.05) and CA2/3 (TBI = 26 ± 2.10, Sham = 20 ± 1.49, P < 0.05), and CR+ decreased in CA1 (TBI = 10 ± 1.02, Sham = 14 ± 1.14, P < 0.05). Astrogliosis significantly increased in corpus callosum (TBI = 6.7 ± 0.69, Sham = 2.5 ± 0.38; P = 0.007). While dTBI effects on inhibitory neurons appear region- and type-specific, a common feature in all cases of decrease was that changes occurred in dendrite targeting interneurons involved in neuronal integration. J. Comp. Neurol. 524:3530-3560, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Simone F Carron
- Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Edwin B Yan
- Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Dasuni S Alwis
- Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Ramesh Rajan
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
41
|
Murugan M, Santhakumar V, Kannurpatti SS. Facilitating Mitochondrial Calcium Uptake Improves Activation-Induced Cerebral Blood Flow and Behavior after mTBI. Front Syst Neurosci 2016; 10:19. [PMID: 27013987 PMCID: PMC4782040 DOI: 10.3389/fnsys.2016.00019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 02/19/2016] [Indexed: 11/13/2022] Open
Abstract
Mild to moderate traumatic brain injury (mTBI) leads to secondary neuronal loss via excitotoxic mechanisms, including mitochondrial Ca(2+) overload. However, in the surviving cellular population, mitochondrial Ca(2+) influx, and oxidative metabolism are diminished leading to suboptimal neuronal circuit activity and poor prognosis. Hence we tested the impact of boosting neuronal electrical activity and oxidative metabolism by facilitating mitochondrial Ca(2+) uptake in a rat model of mTBI. In developing rats (P25-P26) sustaining an mTBI, we demonstrate post-traumatic changes in cerebral blood flow (CBF) in the sensorimotor cortex in response to whisker stimulation compared to sham using functional Laser Doppler Imaging (fLDI) at adulthood (P67-P73). Compared to sham, whisker stimulation-evoked positive CBF responses decreased while negative CBF responses increased in the mTBI animals. The spatiotemporal CBF changes representing underlying neuronal activity suggested profound changes to neurovascular activity after mTBI. Behavioral assessment of the same cohort of animals prior to fLDI showed that mTBI resulted in persistent contralateral sensorimotor behavioral deficit along with ipsilateral neuronal loss compared to sham. Treating mTBI rats with Kaempferol, a dietary flavonol compound that enhanced mitochondrial Ca(2+) uptake, eliminated the inter-hemispheric asymmetry in the whisker stimulation-induced positive CBF responses and the ipsilateral negative CBF responses otherwise observed in the untreated and vehicle-treated mTBI animals in adulthood. Kaempferol also improved somatosensory behavioral measures compared to untreated and vehicle treated mTBI animals without augmenting post-injury neuronal loss. The results indicate that reduced mitochondrial Ca(2+) uptake in the surviving populations affect post-traumatic neural activation leading to persistent behavioral deficits. Improvement in sensorimotor behavior and spatiotemporal neurovascular activity following kaempferol treatment suggests that facilitation of mitochondrial Ca(2+) uptake in the early window after injury may sustain optimal neural activity and metabolism and contribute to improved function of the surviving cellular populations after mTBI.
Collapse
Affiliation(s)
- Madhuvika Murugan
- Department of Radiology, Rutgers New Jersey Medical School Newark, NJ, USA
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School Newark, NJ, USA
| | | |
Collapse
|
42
|
Ziebell JM, Rowe RK, Harrison JL, Eakin KC, Colburn T, Willyerd FA, Lifshitz J. Experimental diffuse brain injury results in regional alteration of gross vascular morphology independent of neuropathology. Brain Inj 2015; 30:217-24. [PMID: 26646974 DOI: 10.3109/02699052.2015.1090012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PRIMARY OBJECTIVE A dynamic relationship exists between diffuse traumatic brain injury and changes to the neurovascular unit. The purpose of this study was to evaluate vascular changes during the first week following diffuse TBI. It was hypothesized that pathology is associated with modification of the vasculature. METHODS Male Sprague-Dawley rats underwent either midline fluid percussion injury or sham-injury. Brain tissue was collected 1, 2 or 7 days post-injury or sham-injury (n = 3/time point). Tissue was collected and stained by de Olmos amino-cupric silver technique to visualize neuropathology or animals were perfused with AltaBlue casting resin before high-resolution vascular imaging. The average volume, surface area, radius, branching and tortuosity of the vessels were evaluated across three regions of interest. RESULTS In M2, average vessel volume (p < 0.01) and surface area (p < 0.05) were significantly larger at 1 day relative to 2 days, 7 days and sham. In S1BF and VPM, no significant differences in the average vessel volume or surface area at any of the post-injury time points were observed. No significant changes in average radius, branching or tortuosity were observed. CONCLUSIONS Preliminary findings suggest gross morphological changes within the vascular network likely represent an acute response to mechanical forces of injury, rather than delayed or chronic pathological processes.
Collapse
Affiliation(s)
- Jenna M Ziebell
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA
| | - Rachel K Rowe
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA.,c Phoenix VA Healthcare System , Phoenix , AZ , USA
| | - Jordan L Harrison
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA
| | - Katharine C Eakin
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA
| | - Taylor Colburn
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA
| | - F Anthony Willyerd
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA.,d Critical Care, Phoenix Children's Hospital , Phoenix , AZ , USA
| | - Jonathan Lifshitz
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA.,c Phoenix VA Healthcare System , Phoenix , AZ , USA.,e Psychology , Arizona State University , Tempe , AZ , USA
| |
Collapse
|
43
|
Harrison JL, Rowe RK, Ellis TW, Yee NS, O’Hara BF, Adelson PD, Lifshitz J. Resolvins AT-D1 and E1 differentially impact functional outcome, post-traumatic sleep, and microglial activation following diffuse brain injury in the mouse. Brain Behav Immun 2015; 47:131-40. [PMID: 25585137 PMCID: PMC4468045 DOI: 10.1016/j.bbi.2015.01.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 12/22/2014] [Accepted: 01/01/2015] [Indexed: 02/09/2023] Open
Abstract
Traumatic brain injury (TBI) is induced by mechanical forces which initiate a cascade of secondary injury processes, including inflammation. Therapies which resolve the inflammatory response may promote neural repair without exacerbating the primary injury. Specific derivatives of omega-3 fatty acids loosely grouped as specialized pro-resolving lipid mediators (SPMs) and termed resolvins promote the active resolution of inflammation. In the current study, we investigate the effect of two resolvin molecules, RvE1 and AT-RvD1, on post-traumatic sleep and functional outcome following diffuse TBI through modulation of the inflammatory response. Adult, male C57BL/6 mice were injured using a midline fluid percussion injury (mFPI) model (6-10min righting reflex time for brain-injured mice). Experimental groups included mFPI administered RvE1 (100ng daily), AT-RvD1 (100ng daily), or vehicle (sterile saline) and counterbalanced with uninjured sham mice. Resolvins or saline were administered daily for seven consecutive days beginning 3days prior to TBI to evaluate proof-of-principle to improve outcome. Immediately following diffuse TBI, post-traumatic sleep was recorded for 24h post-injury. For days 1-7 post-injury, motor outcome was assessed by rotarod. Cognitive function was measured at 6days post-injury using novel object recognition (NOR). At 7days post-injury, microglial activation was quantified using immunohistochemistry for Iba-1. In the diffuse brain-injured mouse, AT-RvD1 treatment, but not RvE1, mitigated motor and cognitive deficits. RvE1 treatment significantly increased post-traumatic sleep in brain-injured mice compared to all other groups. RvE1 treated mice displayed a higher proportion of ramified microglia and lower proportion of activated rod microglia in the cortex compared to saline or AT-RvD1 treated brain-injured mice. Thus, RvE1 treatment modulated post-traumatic sleep and the inflammatory response to TBI, albeit independently of improvement in motor and cognitive outcome as seen in AT-RvD1-treated mice. This suggests AT-RvD1 may impart functional benefit through mechanisms other than resolution of inflammation alone.
Collapse
Affiliation(s)
- Jordan L. Harrison
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
,Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ
| | - Rachel K. Rowe
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
,Phoenix Veteran Affairs Healthcare System, Phoenix, AZ
| | - Timothy W. Ellis
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
,College of Osteopathic Medicine, Midwestern University, Glendale, AZ
| | - Nicole S. Yee
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
| | - Bruce F. O’Hara
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY
,Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - P. David Adelson
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ
,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ
,Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ, USA; Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA.
| |
Collapse
|
44
|
Ziebell JM, Adelson PD, Lifshitz J. Microglia: dismantling and rebuilding circuits after acute neurological injury. Metab Brain Dis 2015; 30:393-400. [PMID: 24733573 PMCID: PMC4198517 DOI: 10.1007/s11011-014-9539-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/26/2014] [Indexed: 01/06/2023]
Abstract
The brain is comprised of neurons and its support system including astrocytes, glial cells and microglia, thereby forming neurovascular units. Neurons require support from glial cells to establish and maintain functional circuits, but microglia are often overlooked. Microglia function as the immune cell of the central nervous system, acting to monitor the microenvironment for changes in signaling, pathogens and injury. More recently, other functional roles for microglia within the healthy brain have been identified, including regulating synapse formation, elimination and function. This review aims to highlight and discuss these alternate microglial roles in the healthy and in contrast, diseased brain with a focus on two acute neurological diseases, traumatic brain injury and epilepsy. In these conditions, microglial roles in synaptic stripping and stabilization as part of neuronal:glial interactions may position them as mediators of the transition between injury-induced circuit dismantling and subsequent reorganization. Increased understanding of microglia roles could identify therapeutic targets to mitigate the consequences of neurological disease.
Collapse
Affiliation(s)
- Jenna M Ziebell
- Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA,
| | | | | |
Collapse
|
45
|
Wang JY, Liou A, Ren ZH, Zhang L, Brown BN, Cui XT, Badylak SF, Cai YN, Guan YQ, Leak RK, Chen J, Ji X, Chen L. Neurorestorative effect of urinary bladder matrix-mediated neural stem cell transplantation following traumatic brain injury in rats. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2014; 12:413-425. [PMID: 23469853 DOI: 10.2174/1871527311312030014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/01/2012] [Accepted: 11/11/2012] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of cell death and disability among young adults and lacks a successful therapeutic strategy. The multiphasic injuries of TBI severely limit the success of conventional pharmacological approaches. Recent successes with transplantation of stem cells in bioactive scaffolds in other injury paradigms provide new hope for the treatment of TBI. In this study, we transplanted neural stem cells (0.5x10(5) cells/µl) cultured in a bioactive scaffold derived from porcine urinary bladder matrix (UBM; 4 injection sites, 2.5µl each) into the rat brain following controlled cortical impact (CCI, velocity, 4.0 m/sec; duration, 0.5 sec; depth, 3.2mm). We evaluated the effectiveness of this strategy to combat the loss of motor, memory and cognitive faculties. Before transplantation, compatibility experiments showed that UBM was able to support extended proliferation and differentiation of neural stem cells. Together with its reported anti-inflammatory properties and rapid degradation characteristics in vivo, UBM emerged to be an ideal scaffold. The transplants reduced neuron/tissue loss and white matter injury, and also significantly ameliorated motor, memory, and cognitive impairments. Furthermore, exposure to UBM alone was sufficient to decrease the loss of sensorimotor skills from TBI (examined 3-28 days post-CCI). However, only UBMs that contained proliferating neural stem cells helped attenuate memory and cognitive impairments (examined 26-28 days post-CCI). In summary, these results demonstrate the therapeutic efficacy of stem cells in bioactive scaffolds against TBI and show promise for translation into future clinical use.
Collapse
Affiliation(s)
- J Y Wang
- Department of Neurosurgery and China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Akf Liou
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Z H Ren
- Department of Neurosurgery and China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - L Zhang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - B N Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA 15261, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - X T Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA 15261, USA.,Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - S F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA 15261, USA.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Y N Cai
- Department of Neurosurgery and China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Y Q Guan
- Department of Neurosurgery and China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, U.S.A
| | - J Chen
- Department of Neurosurgery and China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - X Ji
- Department of Neurosurgery and China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - L Chen
- Department of Neurosurgery and China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
46
|
Taylor SE, Morganti-Kossmann C, Lifshitz J, Ziebell JM. Rod microglia: a morphological definition. PLoS One 2014; 9:e97096. [PMID: 24830807 PMCID: PMC4022629 DOI: 10.1371/journal.pone.0097096] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 04/15/2014] [Indexed: 11/30/2022] Open
Abstract
Brain microglial morphology relates to function, with ramified microglia surveying the micro-environment and amoeboid microglia engulfing debris. One subgroup of microglia, rod microglia, have been observed in a number of pathological conditions, however neither a function nor specific morphology has been defined. Historically, rod microglia have been described intermittently as cells with a sausage-shaped soma and long, thin processes, which align adjacent to neurons. More recently, our group has described rod microglia aligning end-to-end with one another to form trains adjacent to neuronal processes. Confusion in the literature regarding rod microglia arises from some reports referring to the sausage-shaped cell body, while ignoring the spatial distribution of processes. Here, we systematically define the morphological characteristics of rod microglia that form after diffuse brain injury in the rat, which differ morphologically from the spurious rod microglia found in uninjured sham. Rod microglia in the diffuse-injured rat brain show a ratio of 1.79 ± 0.03 cell length:cell width at day 1 post-injury, which increases to 3.35 ± 0.05 at day 7, compared to sham (1.17 ± 0.02). The soma length:width differs only at day 7 post-injury (2.92 ± 0.07 length:width), compared to sham (2.49 ± 0.05). Further analysis indicated that rod microglia may not elongate in cell length but rather narrow in cell width, and retract planar (side) processes. These morphological characteristics serve as a tool for distinguishing rod microglia from other morphologies. The function of rod microglia remains enigmatic; based on morphology we propose origins and functions for rod microglia after acute neurological insult, which may provide biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Samuel E. Taylor
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, United States of America
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, Arizona, United States of America
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Cristina Morganti-Kossmann
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, United States of America
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, Arizona, United States of America
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, United States of America
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, Arizona, United States of America
- Phoenix VA Healthcare System, Phoenix, Arizona, United States of America
- Neuroscience Program, Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
| | - Jenna M. Ziebell
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, United States of America
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, Arizona, United States of America
| |
Collapse
|
47
|
Alwis DS, Johnstone V, Yan E, Rajan R. Diffuse traumatic brain injury and the sensory brain. Clin Exp Pharmacol Physiol 2014; 40:473-83. [PMID: 23611812 DOI: 10.1111/1440-1681.12100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 04/17/2013] [Indexed: 01/06/2023]
Abstract
In this review we discuss the consequences to the brain's cortex, specifically to the sensory cortex, of traumatic brain injury. The thesis underlying this approach is that long-term deficits in cognition seen after brain damage in humans are likely underpinned by an impaired cortical processing of the sensory information needed to drive cognition or to be used by cognitive processes to produce a response. We take it here that the impairment to sensory processing does not arise from damage to peripheral sensory systems, but from disordered brain processing of sensory input.
Collapse
Affiliation(s)
- Dasuni S Alwis
- Department of Physiology, Monash University, Melbourne, Vic., Australia
| | | | | | | |
Collapse
|
48
|
Miremami JD, Talauliker PM, Harrison JL, Lifshitz J. Neuropathology in sensory, but not motor, brainstem nuclei of the rat whisker circuit after diffuse brain injury. Somatosens Mot Res 2014; 31:127-35. [DOI: 10.3109/08990220.2014.897602] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
49
|
Chen Z, Shin D, Chen S, Mikhail K, Hadass O, Tomlison BN, Korkin D, Shyu CR, Cui J, Anthony DC, Gu Z. Histological quantitation of brain injury using whole slide imaging: a pilot validation study in mice. PLoS One 2014; 9:e92133. [PMID: 24637518 PMCID: PMC3956884 DOI: 10.1371/journal.pone.0092133] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/18/2014] [Indexed: 12/28/2022] Open
Abstract
Quantitative assessment of serial brain sections provides an objective measure of neurological events at cellular and molecular levels but is difficult to implement in experimental neuroscience laboratories because of variation from person-to-person and the time required for analysis. Whole slide imaging (WSI) technology, recently introduced for pathological diagnoses, offers an electronic environment and a variety of computational tools for performing high-throughput histological analysis and managing the associated information. In our study, we applied various algorithms to quantify histologic changes associated with brain injury and compared the results to manual assessment. WSI showed a high degree of concordance with manual quantitation by Pearson correlation and strong agreement using Bland-Altman plots in: (i) cortical necrosis in cresyl-violet-stained brain sections of mice after focal cerebral ischemia; (ii) intracerebral hemorrhage in ischemic mouse brains for automated annotation of the small regions, rather than whole hemisphere of the tissue sections; (iii) Iba1-immunoreactive cell density in the adjacent and remote brain regions of mice subject to controlled cortical impact (CCI); and (iv) neuronal degeneration by silver staining after CCI. These results show that WSI, when appropriately applied and carefully validated, is a highly efficient and unbiased tool to locate and identify neuropathological features, delineate affected regions and histologically quantify these events.
Collapse
Affiliation(s)
- Zhenzhou Chen
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dmitriy Shin
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| | - Shanyan Chen
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Interdisciplinary Neuroscience Program, University of Missouri, Columbia, Missouri, United States of America
| | - Kovalenko Mikhail
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| | - Orr Hadass
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| | - Brittany N Tomlison
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| | - Dmitry Korkin
- Informatics Institute and Department of Computer Science, University of Missouri, Columbia, Missouri, United States of America
| | - Chi-Ren Shyu
- Informatics Institute and Department of Computer Science, University of Missouri, Columbia, Missouri, United States of America
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| | - Douglas C Anthony
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America; Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| |
Collapse
|
50
|
Rowe RK, Striz M, Bachstetter AD, Van Eldik LJ, Donohue KD, O'Hara BF, Lifshitz J. Diffuse brain injury induces acute post-traumatic sleep. PLoS One 2014; 9:e82507. [PMID: 24416145 PMCID: PMC3885381 DOI: 10.1371/journal.pone.0082507] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 10/24/2013] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Clinical observations report excessive sleepiness immediately following traumatic brain injury (TBI); however, there is a lack of experimental evidence to support or refute the benefit of sleep following a brain injury. The aim of this study is to investigate acute post-traumatic sleep. METHODS Sham, mild or moderate diffuse TBI was induced by midline fluid percussion injury (mFPI) in male C57BL/6J mice at 9:00 or 21:00 to evaluate injury-induced sleep behavior at sleep and wake onset, respectively. Sleep profiles were measured post-injury using a non-invasive, piezoelectric cage system. In separate cohorts of mice, inflammatory cytokines in the neocortex were quantified by immunoassay, and microglial activation was visualized by immunohistochemistry. RESULTS Immediately after diffuse TBI, quantitative measures of sleep were characterized by a significant increase in sleep (>50%) for the first 6 hours post-injury, resulting from increases in sleep bout length, compared to sham. Acute post-traumatic sleep increased significantly independent of injury severity and time of injury (9:00 vs 21:00). The pro-inflammatory cytokine IL-1β increased in brain-injured mice compared to sham over the first 9 hours post-injury. Iba-1 positive microglia were evident in brain-injured cortex at 6 hours post-injury. CONCLUSION Post-traumatic sleep occurs for up to 6 hours after diffuse brain injury in the mouse regardless of injury severity or time of day. The temporal profile of secondary injury cascades may be driving the significant increase in post-traumatic sleep and contribute to the natural course of recovery through cellular repair.
Collapse
Affiliation(s)
- Rachel K. Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, United States of America
- Department of Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona, United States of America
- Department of Anatomy and Neurobiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Martin Striz
- Department of Biology, College of Arts and Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| | - Adam D. Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Linda J. Van Eldik
- Department of Anatomy and Neurobiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Kevin D. Donohue
- Department of Electrical and Computer Engineering, College of Engineering, University of Kentucky, Lexington, Kentucky, United States of America
| | - Bruce F. O'Hara
- Department of Biology, College of Arts and Sciences, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, United States of America
- Department of Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona, United States of America
- Phoenix Veteran Affairs Healthcare System, Phoenix, Arizona, United States of America
- Department of Anatomy and Neurobiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Physical Medicine & Rehabilitation, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|