1
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 PMCID: PMC12051134 DOI: 10.1016/j.neuropharm.2024.110084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Microglia play numerous important roles in brain development. From early embryonic stages through adolescence, these immune cells influence neuronal genesis and maturation, guide connectivity, and shape brain circuits. They also interact with other glial cells and structures, influencing the brain's supportive microenvironment. While this central role makes microglia essential, it means that early life perturbations to microglia can have widespread effects on brain development, potentially resulting in long-lasting behavioral impairments. Here, we will focus on the effects of early life psychosocial versus physiological stressors in rodent models. Psychosocial stress refers to perceived threats that lead to stress axes activation, including prenatal stress, or chronic postnatal stress, including maternal separation and resource scarcity. Physiological stress refers to physical threats, including maternal immune activation, postnatal infection, and traumatic brain injury. Differing sources of early life stress have varied impacts on microglia, and these effects are moderated by factors such as developmental age, brain region, and sex. Overall, these stressors appear to either 1) upregulate basal microglia numbers and activity throughout the lifespan, while possibly blunting their responsivity to subsequent stressors, or 2) shift the developmental curve of microglia, resulting in differential timing and function, impacting the critical periods they govern. Either could contribute to behavioral dysfunctions that occur after the resolution of early life stress. Exploring how different stressors impact microglia, as well as how multiple stressors interact to alter microglia's developmental functions, could deepen our understanding of how early life stress changes the brain's developmental trajectory. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Psychology, Ohio State University, Columbus, OH, USA.
| | - Kathryn M Lenz
- Department of Psychology, Ohio State University, Columbus, OH, USA; Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Ohio State University, Columbus, OH, USA
| |
Collapse
|
2
|
McPherson JI, Prakash Krishnan Muthaiah V, Kaliyappan K, Leddy JJ, Personius KE. Temporal expression of brainstem neurotrophic proteins following mild traumatic brain injury. Brain Res 2024; 1835:148908. [PMID: 38582416 DOI: 10.1016/j.brainres.2024.148908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
BDNF, a neurotrophic factor, and its receptors have been implicated in the pathophysiology of mild traumatic brain injury (mTBI). The brainstem houses many vital functions, that are also associated with signs and symptoms of mTBI, but has been understudied in mTBI animal models. We determined the extent to which neurotrophic protein and associated receptor expression is affected within the brainstem of adult rats following mTBI. Their behavioral function was assessed and temporal expression of the 'negative' regulators of neuronal function (p75, t-TrkB, and pro-BDNF) and 'positive' neuroprotective (FL-TrkB and m-BDNF) protein isoforms were determined via western blot and immunohistochemistry at 1, 3, 7, and 14 post-injury days (PID) following mTBI or sham (control) procedure. Within the brainstem, p75 expression increased at PID 1 vs. sham animals. t-TrkB and pro-BDNF expression increased at PID 7 and 14. The 'positive' protein isoforms of FL-TrkB and m-BDNF expression were increased only at PID 7. The ratio of t-TrkB:FL-TrkB (negative:positive) was substantial across groups and time points, suggesting a negative impact of neurotrophic signaling on neuronal function. Additional NeuN experiments revealed cell death occurring within a subset of neurons within the medulla. While behavioral measures improved by PID 7-14, negative neurotrophic biochemical responses persisted. Despite the assertion that mTBI produces "mild" injury, evidence of cell death was observed in the medulla. Ratios of TrkB and BDNF isoforms with conflicting functions suggest that future work should specifically measure each subtype since they induce opposing downstream effects on neuronal function.
Collapse
Affiliation(s)
- Jacob I McPherson
- Department of Rehabilitation Science, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, United States.
| | - Vijaya Prakash Krishnan Muthaiah
- Department of Rehabilitation Science, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, United States
| | - Kathiravan Kaliyappan
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - John J Leddy
- Department of Orthopaedics and Sports Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - Kirkwood E Personius
- Department of Rehabilitation Science, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
3
|
Smith AM, Grayson BE. A strike to the head: Parallels between the pediatric and adult human and the rodent in traumatic brain injury. J Neurosci Res 2024; 102:e25364. [PMID: 38953607 DOI: 10.1002/jnr.25364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024]
Abstract
Traumatic brain injury (TBI) is a condition that occurs commonly in children from infancy through adolescence and is a global health concern. Pediatric TBI presents with a bimodal age distribution, with very young children (0-4 years) and adolescents (15-19 years) more commonly injured. Because children's brains are still developing, there is increased vulnerability to the effects of head trauma, which results in entirely different patterns of injury than in adults. Pediatric TBI has a profound and lasting impact on a child's development and quality of life, resulting in long-lasting consequences to physical, cognitive, and emotional development. Chronic issues like learning disabilities, behavioral problems, and emotional disturbances can develop. Early intervention and ongoing support are critical for minimizing these long-term deficits. Many animal models of TBI exist, and each varies significantly, displaying different characteristics of clinical TBI. The neurodevelopment differs in the rodent from the human in timing and effect, so TBI outcomes in the juvenile rodent can thus vary from the human child. The current review compares findings from preclinical TBI work in juvenile and adult rodents to clinical TBI research in pediatric and adult humans. We focus on the four brain regions most affected by TBI: the prefrontal cortex, corpus callosum, hippocampus, and hypothalamus. Each has its unique developmental projections and thus is impacted by TBI differently. This review aims to compare the healthy neurodevelopment of these four brain regions in humans to the developmental processes in rodents.
Collapse
Affiliation(s)
- Allie M Smith
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Bernadette E Grayson
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Population Health Science, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
4
|
Sgro M, Kodila ZN, Li C, Carmichael I, Warren S, Reichelt AC, Yamakawa GR, Mychasiuk R. Microbiome depletion prior to repeat mild TBI differentially alters social deficits and prefrontal cortex plasticity in adolescent and adult rats. iScience 2024; 27:109395. [PMID: 38510122 PMCID: PMC10952042 DOI: 10.1016/j.isci.2024.109395] [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: 10/02/2023] [Revised: 11/21/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Although aging, repeat mild traumatic brain injury (RmTBI), and microbiome modifications independently change social behavior, there has been no investigation into their cumulative effects on social behavior and neuroplasticity within the prefrontal cortex. Therefore, we examined how microbiome depletion prior to RmTBI affected social behavior and neuroplasticity in adolescent and adult rats. Play, temperament analysis, elevated plus maze, and the hot/cold plate assessed socio-emotional function. Analyses of perineuronal nets (PNNs) and parvalbumin (PV) interneurons was completed. Social-emotional deficits were more pronounced in adults, with microbiome depletion attenuating social behavior deficits associated with RmTBI in both age groups. Microbiome depletion increased branch length and PNN arborization within the PFC but decreased the overall number of PNNs. Adults and males were more vulnerable to RmTBI. Interestingly, microbiome depletion may have attenuated the changes to neuroplasticity and subsequent social deficits, suggesting that the microbiome is a viable, but age-specific, target for RmTBI therapeutics.
Collapse
Affiliation(s)
- Marissa Sgro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Zoe N. Kodila
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Crystal Li
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Irena Carmichael
- Monash Micro Imaging, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Samantha Warren
- Monash Micro Imaging, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Amy C. Reichelt
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Glenn R. Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
5
|
Semple BD, Mychasiuk R. Sex and Age-at-Injury as Determinants of Social Behavior Outcomes After TBI. ADVANCES IN NEUROBIOLOGY 2024; 42:205-218. [PMID: 39432044 DOI: 10.1007/978-3-031-69832-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
While our understanding of long-term disability after traumatic brain injury (TBI) has habitually focused on cognitive and sensorimotor functioning, it is increasingly appreciated that changes in social function for survivors of a brain injury are common and have a profound impact on one's quality of life. In this chapter, we highlight the consequences of TBI on social behavior, taking into account evidence from studies of patient populations as well as from preclinical animal models. After first considering the protracted nature of the development of social behavior across the lifespan, including the neurobiological networks that underlie social functioning, we discuss how TBI results in social behavior impairments and how these manifest. We focus particularly on how age-at-injury influences TBI-induced social impairments, with most of the evidence suggesting age-dependent vulnerability after injury at a younger age. In addition, we explore how biological sex is a key determinant of social behavior impairments after TBI, while gender in humans may also influence the nature and extent of social outcomes. Finally, we identify key knowledge gaps and emphasize the need for further research in the field.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia.
| | - Richelle Mychasiuk
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
6
|
Dill LK, Teymornejad S, Sharma R, Bozkurt S, Christensen J, Chu E, Rewell SS, Shad A, Mychasiuk R, Semple BD. Modulating chronic outcomes after pediatric traumatic brain injury: Distinct effects of social and environmental enrichment. Exp Neurol 2023; 364:114407. [PMID: 37059414 DOI: 10.1016/j.expneurol.2023.114407] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/16/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
Impairments in social and cognitive function are a common consequence of pediatric traumatic brain injury (TBI). Rehabilitation has the potential to promote optimal behavioral recovery. Here, we evaluated whether an enhanced social and/or cognitive environment could improve long-term outcomes in a preclinical model of pediatric TBI. Male C57Bl/6 J mice received a moderately-severe TBI or sham procedure at postnatal day 21. After one week, mice were randomized to different social conditions (minimal socialization, n = 2/cage; or social grouping, n = 6/cage), and housing conditions (standard cage, or environmental enrichment (EE), incorporating sensory, motor, and cognitive stimuli). After 8 weeks, neurobehavioral outcomes were assessed, followed by post-mortem neuropathology. We found that TBI mice exhibited hyperactivity, spatial memory deficits, reduced anxiety-like behavior, and reduced sensorimotor performance compared to age-matched sham controls. Pro-social and sociosexual behaviors were also reduced in TBI mice. EE increased sensorimotor performance, and the duration of sociosexual interactions. Conversely, social housing reduced hyperactivity and altered anxiety-like behavior in TBI mice, and reduced same-sex social investigation. TBI mice showed impaired spatial memory retention, except for TBI mice exposed to both EE and group housing. In the brain, while TBI led to significant regional tissue atrophy, social housing had modest neuroprotective effects on hippocampal volumes, neurogenesis, and oligodendrocyte progenitor numbers. In conclusion, manipulation of the post-injury environment has benefit for chronic behavioral outcomes, but the benefits are specific to the type of enrichment available. This study improves understanding of modifiable factors that may be harnessed to optimize long-term outcomes for survivors of early-life TBI.
Collapse
Affiliation(s)
- Larissa K Dill
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia; The Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Sadaf Teymornejad
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Rishabh Sharma
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Jennaya Christensen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Erskine Chu
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Sarah S Rewell
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Ali Shad
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC 3050, Australia.
| |
Collapse
|
7
|
Hossain SR, Karem H, Jafari Z, Kolb BE, Mohajerani MH. Tactile stimulation improves cognition, motor, and anxiety-like behaviors and attenuates the Alzheimer's disease pathology in adult APP NL-G-F/NL-G-F mice. Synapse 2023; 77:e22257. [PMID: 36255152 DOI: 10.1002/syn.22257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/05/2022] [Accepted: 09/28/2022] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is one of the largest health crises in the world. There are limited pharmaceutical interventions to treat AD, however, and most of the treatment options are not for cure or prevention, but rather to slow down the progression of the disease. The aim of this study was to examine the effect of tactile stimulation (TS) on AD-like symptoms and pathology in APPNL-G-F/NL-G-F mice, a mouse model of AD. The results show that TS reduces the AD-like symptoms on tests of cognition, motor, and anxiety-like behaviors and these improvements in behavior are associated with reduced AD pathology in APP mice. Thus, TS appears to be a promising noninvasive strategy for slowing the onset of dementia in aging animals.
Collapse
Affiliation(s)
- Shakhawat R Hossain
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Hadil Karem
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Zahra Jafari
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Bryan E Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| |
Collapse
|
8
|
Eyolfson E, Carr T, Fraunberger E, Khan A, Clark I, Mychasiuk R, Lohman AW. Repeated mild traumatic brain injuries in mice cause age- and sex-specific alterations in dendritic spine density. Exp Neurol 2022; 357:114172. [PMID: 35863503 DOI: 10.1016/j.expneurol.2022.114172] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/08/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022]
Abstract
Mild traumatic brain injuries (mTBI) plague the human population and their prevalence is increasing annually. More so, repeated mTBIs (RmTBI) are known to manifest and compound neurological deficits in vulnerable populations. Age at injury and sex are two important factors influencing RmTBI pathophysiology, but we continue to know little about the specific effects of RmTBI in youth and females. In this study, we directly quantified the effects of RmTBI on adolescent and adult, male and female mice, with a closed-head lateral impact model. We report age- and sex-specific neurobehavioural deficits in motor function and working memory, microglia responses to injury, and the subsequent changes in dendritic spine density in select brain regions. Specifically, RmTBI caused increased footslips in adult male mice as assessed in a beam walk assay and significantly reduced the time spent with a novel object in adolescent male and female mice. RmTBIs caused a significant reduction in microglia density in male mice in the motor cortex, but not female mice. Finally, RmTBI significantly reduced dendritic spine density in the agranular insular cortex (a region of the prefrontal cortex in mice) and increased dendritic spine density in the adolescent male motor cortex. Together, the data provided in this study sheds new light on the heterogeneity in RmTBI-induced behavioural, glial, and neuronal architecture changes dependent on age and sex.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada.
| | - Thomas Carr
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Erik Fraunberger
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Asher Khan
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Isabel Clark
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Alexander W Lohman
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
9
|
Li JL, Cao Y, Nie H. The Effect of Mild Hypothermia on Nogo-A and Neurological Function in the Brain after Cardiopulmonary Resuscitation in Rats. Fetal Pediatr Pathol 2022; 41:198-207. [PMID: 32589081 DOI: 10.1080/15513815.2020.1783407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
ObjectiveWe investigated the dynamic changes of Nogo-A protein in brain and the effects of mild therapeutic hypothermia (MTH) on its expression after cardiopulmonary resuscitation (CPR). Methods: Western-blotting and neurological scoring of 45 rats subjected to cardiac arrest and CPR with and without MTR were performed to investigate the changes in the expression of Nogo-A protein in the hippocampus and cortex over a period of time ranging from 6 h to 72 h after restoration of spontaneous circulation (ROSC). Results: Nogo-A expression levels were increased at 6 h after CPR in the hippocampus and cortex, peaked at 24 h in the cortex, and at 48 h in the hippocampus. The expression of Nogo-A in the MTR group was significantly lower at 12 h (p < 0.05) compared to those with no MTR after ROSC. Conclusions: MTR blunts the expression of Nogo-A protein in the hippocampus and cortex after cardiac arrest and resuscitation, and MTR may provide cerebral protection after ischemia.
Collapse
Affiliation(s)
- Jia-Li Li
- Department of Emergency, The Second Affifiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, Sichuan, China
| | - Yu Cao
- Department of Emergency, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hu Nie
- Department of Emergency, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Giacometti LL, Huh JW, Raghupathi R. Sex and estrous-phase dependent alterations in depression-like behavior following mild traumatic brain injury in adolescent rats. J Neurosci Res 2021; 100:490-505. [PMID: 34850450 DOI: 10.1002/jnr.24989] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/04/2021] [Accepted: 10/10/2021] [Indexed: 12/21/2022]
Abstract
Following mild traumatic brain injury (TBI), high school and collegiate-aged females tend to report more emotional symptoms than males. Adolescent male and female rats (35 days old) were subjected to mild TBI and evaluated for anxiety- and depression-like behaviors using the elevated plus maze and forced swim test (FST), respectively, and cellular alterations. Injured brains did not exhibit an overt lesion, atrophy of tissue or astrocytic reactivity underneath the impact site at 6-week post-injury, suggestive of the mild nature of trauma. Neither male nor female brain-injured rats exhibited anxiety-like behavior at 2 or 6 weeks, regardless of estrous phase at the time of behavior testing. Brain-injured male rats did not exhibit any alterations in immobility, swimming and climbing times in the FST compared to sham-injured rats at either 2- or 6-week post-injury. Brain-injured female rats did, however, exhibit an increase in immobility (in the absence of changes in swimming and climbing times) in the FST at 6 weeks post-injury only during the estrus phase of the estrous cycle, suggestive of a depression-like phenotype. Combined administration of the estrogen receptor antagonist, tamoxifen, and the progesterone receptor antagonist, mifepristone, during proestrus was able to prevent the depression-like phenotype observed during estrus. Taken together, these data suggest that female rats may be more vulnerable to exhibiting behavioral deficits following mild TBI and that estrous phase may play a role in depression-like behavior.
Collapse
Affiliation(s)
- Laura L Giacometti
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, Pennsylvania, USA.,Graduate School of Biomedical Sciences and Professional Studies, College of Medicine, Drexel University, Philadelphia, Pennsylvania, USA
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ramesh Raghupathi
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, Pennsylvania, USA.,Graduate School of Biomedical Sciences and Professional Studies, College of Medicine, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Rose SC, Yeates KO, Nguyen JT, Ercole PM, Pizzimenti NM, McCarthy MT. Subconcussive Head Impacts and Neurocognitive Function Over 3 Seasons of Youth Football. J Child Neurol 2021; 36:768-775. [PMID: 33834862 DOI: 10.1177/08830738211004490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To determine the association between repetitive subconcussive head impacts and neurobehavioral outcomes in youth tackle football players. METHODS Using helmet-based sensors, we measured head impacts for 3 consecutive seasons of play in 29 male players age 9-11. Cumulative impact g's were calculated. Players completed a battery of outcome measures before and after each season, including neuropsychological testing, vestibular-ocular sensitivity, and self- and parent-reported measures of symptoms and attention-deficit hyperactivity disorder (ADHD). RESULTS Average cumulative impact over 3 seasons was 13 900g. High-intensity hits predicted worse change for self-reported social adjustment (P = .001). Cumulative impact did not predict change in any of the outcome measures. History of ADHD, anxiety, and depression predicted worse change for self-reported symptoms and social adjustment, independent of head impacts. When players were stratified into 3 groups based on cumulative impact across all 3 seasons, differences in outcome measures existed prior to the start of the first season. These differences did not further increase over the course of the 3 seasons. CONCLUSION Over 3 consecutive seasons of youth tackle football, we found no association between cumulative head impacts and neurobehavioral outcomes. Larger sample sizes and longer follow-up times would further assist in characterizing this relationship.
Collapse
Affiliation(s)
- Sean C Rose
- Division of Child Neurology, 2647Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Keith Owen Yeates
- Department of Psychology, Alberta Children's Hospital Research Institute, and Hotchkiss Brain Institute, 2129University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
12
|
Serpa RO, Ferguson L, Larson C, Bailard J, Cooke S, Greco T, Prins ML. Pathophysiology of Pediatric Traumatic Brain Injury. Front Neurol 2021; 12:696510. [PMID: 34335452 PMCID: PMC8319243 DOI: 10.3389/fneur.2021.696510] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022] Open
Abstract
The national incidence of traumatic brain injury (TBI) exceeds that of any other disease in the pediatric population. In the United States the Centers for Disease Control and Prevention (CDC) reports 697,347 annual TBIs in children ages 0–19 that result in emergency room visits, hospitalization or deaths. There is a bimodal distribution within the pediatric TBI population, with peaks in both toddlers and adolescents. Preclinical TBI research provides evidence for age differences in acute pathophysiology that likely contribute to long-term outcome differences between age groups. This review will examine the timecourse of acute pathophysiological processes during cerebral maturation, including calcium accumulation, glucose metabolism and cerebral blood flow. Consequences of pediatric TBI are complicated by the ongoing maturational changes allowing for substantial plasticity and windows of vulnerabilities. This review will also examine the timecourse of later outcomes after mild, repeat mild and more severe TBI to establish developmental windows of susceptibility and altered maturational trajectories. Research progress for pediatric TBI is critically important to reveal age-associated mechanisms and to determine knowledge gaps for future studies.
Collapse
Affiliation(s)
- Rebecka O Serpa
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lindsay Ferguson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cooper Larson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Julie Bailard
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha Cooke
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tiffany Greco
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mayumi L Prins
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Eyolfson E, Carr T, Khan A, Wright DK, Mychasiuk R, Lohman AW. Repetitive Mild Traumatic Brain Injuries in Mice during Adolescence Cause Sexually Dimorphic Behavioral Deficits and Neuroinflammatory Dynamics. J Neurotrauma 2020; 37:2718-2732. [DOI: 10.1089/neu.2020.7195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, Alberta, Canada
| | - Thomas Carr
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta, Canada
| | - Asher Khan
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta, Canada
| | - David K. Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, Alberta, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta, Canada
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Alexander W. Lohman
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
14
|
Lengel D, Sevilla C, Romm ZL, Huh JW, Raghupathi R. Stem Cell Therapy for Pediatric Traumatic Brain Injury. Front Neurol 2020; 11:601286. [PMID: 33343501 PMCID: PMC7738475 DOI: 10.3389/fneur.2020.601286] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
There has been a growing interest in the potential of stem cell transplantation as therapy for pediatric brain injuries. Studies in pre-clinical models of pediatric brain injury such as Traumatic Brain Injury (TBI) and neonatal hypoxia-ischemia (HI) have contributed to our understanding of the roles of endogenous stem cells in repair processes and functional recovery following brain injury, and the effects of exogenous stem cell transplantation on recovery from brain injury. Although only a handful of studies have evaluated these effects in models of pediatric TBI, many studies have evaluated stem cell transplantation therapy in models of neonatal HI which has a considerable overlap of injury pathology with pediatric TBI. In this review, we have summarized data on the effects of stem cell treatments on histopathological and functional outcomes in models of pediatric brain injury. Importantly, we have outlined evidence supporting the potential for stem cell transplantation to mitigate pathology of pediatric TBI including neuroinflammation and white matter injury, and challenges that will need to be addressed to incorporate these therapies to improve functional outcomes following pediatric TBI.
Collapse
Affiliation(s)
- Dana Lengel
- Graduate Program in Neuroscience, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Cruz Sevilla
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Zoe L Romm
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ramesh Raghupathi
- Graduate Program in Neuroscience, Drexel University College of Medicine, Philadelphia, PA, United States.,Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
15
|
Eyolfson E, Khan A, Mychasiuk R, Lohman AW. Microglia dynamics in adolescent traumatic brain injury. J Neuroinflammation 2020; 17:326. [PMID: 33121516 PMCID: PMC7597018 DOI: 10.1186/s12974-020-01994-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Repetitive, mild traumatic brain injuries (RmTBIs) are increasingly common in adolescents and encompass one of the largest neurological health concerns in the world. Adolescence is a critical period for brain development where RmTBIs can substantially impact neurodevelopmental trajectories and life-long neurological health. Our current understanding of RmTBI pathophysiology suggests key roles for neuroinflammation in negatively regulating neural health and function. Microglia, the brain’s resident immune population, play important roles in brain development by regulating neuronal number, and synapse formation and elimination. In response to injury, microglia activate to inflammatory phenotypes that may detract from these normal homeostatic, physiological, and developmental roles. To date, however, little is known regarding the impact of RmTBIs on microglia function during adolescent brain development. This review details key concepts surrounding RmTBI pathophysiology, adolescent brain development, and microglia dynamics in the developing brain and in response to injury, in an effort to formulate a hypothesis on how the intersection of these processes may modify long-term trajectories.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N1N4, Canada.,Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, NW, Calgary, AB, T2N4N1, Canada
| | - Asher Khan
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, NW, Calgary, AB, T2N4N1, Canada
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N1N4, Canada.,Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, NW, Calgary, AB, T2N4N1, Canada.,Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Alexander W Lohman
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, NW, Calgary, AB, T2N4N1, Canada. .,Department of Cell Biology and Anatomy, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.
| |
Collapse
|
16
|
Davis CK, Vemuganti R. DNA damage and repair following traumatic brain injury. Neurobiol Dis 2020; 147:105143. [PMID: 33127471 DOI: 10.1016/j.nbd.2020.105143] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 01/05/2023] Open
Abstract
Traumatic brain injury (TBI) is known to promote significant DNA damage irrespective of age, sex, and species. Chemical as well as structural DNA modification start within minutes and persist for days after TBI. Although several DNA repair pathways are induced following TBI, the simultaneous downregulation of some of the genes and proteins of these pathways leads to an aberrant overall DNA repair process. In many instances, DNA damages escape even the most robust repair mechanisms, especially when the repair process becomes overwhelmed or becomes inefficient by severe or repeated injuries. The persisting DNA damage and/or lack of DNA repair contributes to long-term functional deficits. In this review, we discuss the mechanisms of TBI-induced DNA damage and repair. We further discussed the putative experimental therapies that target the members of the DNA repair process for improved outcome following TBI.
Collapse
Affiliation(s)
- Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA.
| |
Collapse
|
17
|
The behavioural and pathophysiological effects of the ketogenic diet on mild traumatic brain injury in adolescent rats. Behav Brain Res 2019; 376:112225. [DOI: 10.1016/j.bbr.2019.112225] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/16/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
|
18
|
Christensen J, Yamakawa GR, Salberg S, Wang M, Kolb B, Mychasiuk R. Caffeine consumption during development alters spine density and recovery from repetitive mild traumatic brain injury in young adult rats. Synapse 2019; 74:e22142. [PMID: 31705690 DOI: 10.1002/syn.22142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 11/10/2022]
Abstract
Caffeine is the most commonly used psychostimulant throughout the world, with its consumption being especially prevalent among adolescents and young adults, as over 75% of this group consumes caffeine daily. Similarly, the adolescent and young adult age group exhibit the highest incidence of traumatic brain injury (TBI). Given that both caffeine consumption and mild TBI (mTBI) are more prevalent among the late adolescent/young adult age group and that changes in dendritic spine morphology during this developmental period are poorly understood, this study sought to examine the effects of caffeine consumption during late adolescence/early adulthood on recovery from repetitive mTBI (RmTBI). The study specifically focused on changes to neuronal dendritic morphology as synaptic changes likely underlie long-term behavioral outcomes. The results demonstrate that during young adulthood caffeine consumption differentially affects the RmTBI outcomes of males and females, where the effects of caffeine and RmTBI were often additive in males while being equally detrimental, but rarely additive, in females. In general, caffeine and RmTBI induced the greatest impairments in males on cognitive and motor tasks whereas in females the most significant detriments were on pain-related tasks. Both caffeine and RmTBI increased spine density in the Cg3 (medial prefrontal cortex [mPFC]), AID (orbitofrontal cortex [OFC]), and nucleus accumbens (NAc), which is proposed to reflect an impairment in the normal pruning processes. Overall, despite caffeine's neuroprotective abilities among other age groups, this study offers concerning results regarding the detrimental effects of caffeine and RmTBI, in isolation, and especially in combination, in this susceptible population.
Collapse
Affiliation(s)
- Jennaya Christensen
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Melinda Wang
- Department of Psychology, University of Calgary, Calgary, AB, Canada
| | - Bryan Kolb
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW In the quest for understanding the pathophysiological processes underlying degeneration of nervous systems, synapses are emerging as sites of great interest as synaptic dysfunction is thought to play a role in the initiation and progression of neuronal loss. In particular, the synapse is an interesting target for the effects of epigenetic mechanisms in neurodegeneration. Here, we review the recent advances on epigenetic mechanisms driving synaptic compromise in major neurodegenerative disorders. RECENT FINDINGS Major developments in sequencing technologies enabled the mapping of transcriptomic patterns in human postmortem brain tissues in various neurodegenerative diseases, and also in cell and animal models. These studies helped identify changes in classical neurodegeneration pathways and discover novel targets related to synaptic degeneration. Identifying epigenetic patterns indicative of synaptic defects prior to neuronal degeneration may provide the basis for future breakthroughs in the field of neurodegeneration.
Collapse
Affiliation(s)
- Mary Xylaki
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Benedict Atzler
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany.
- Max Planck Institute for Experimental Medicine, 37075, Göttingen, Germany.
- Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
20
|
Cannella LA, McGary H, Ramirez SH. Brain interrupted: Early life traumatic brain injury and addiction vulnerability. Exp Neurol 2019; 317:191-201. [PMID: 30862466 PMCID: PMC6544498 DOI: 10.1016/j.expneurol.2019.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/27/2019] [Accepted: 03/08/2019] [Indexed: 12/20/2022]
Abstract
Recent reports provide evidence for increased risk of substance use disorders (SUD) among patients with a history of early-life traumatic brain injury (TBI). Preclinical research utilizing animal models of TBI have identified injury-induced inflammation, blood-brain barrier permeability, and changes to synapses and neuronal networks within regions of the brain associated with the perception of reward. Importantly, these reward pathway networks are underdeveloped during childhood and adolescence, and early-life TBI pathology may interrupt ongoing maturation. As such, maladaptive changes induced by juvenile brain injury may underlie increased susceptibility to SUD. In this review, we describe the available clinical and preclinical evidence that identifies SUD as a persistent psychiatric consequence of pediatric neurotrauma by discussing (1) the incidence of early-life TBI, (2) how preclinical studies model TBI and SUD, (3) TBI-induced neuropathology and neuroinflammation in the corticostriatal regions of the brain, and (4) the link between childhood or adolescent TBI and addiction in adulthood. In summary, preclinical research utilizes an innovative combination of models of early-life TBI and SUD to recapitulate clinical features and to determine how TBI promotes a risk for the development of SUD. However, causal processes that link TBI and SUD remain unclear. Additional research to identify and therapeutically target underlying mechanisms of aberrant reward pathway development will provide a launching point for TBI and SUD treatment strategies.
Collapse
Affiliation(s)
- Lee Anne Cannella
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hannah McGary
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Servio H Ramirez
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; Shriners Hospitals Pediatric Research Center, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
21
|
Bodnar CN, Roberts KN, Higgins EK, Bachstetter AD. A Systematic Review of Closed Head Injury Models of Mild Traumatic Brain Injury in Mice and Rats. J Neurotrauma 2019; 36:1683-1706. [PMID: 30661454 PMCID: PMC6555186 DOI: 10.1089/neu.2018.6127] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mild TBI (mTBI) is a significant health concern. Animal models of mTBI are essential for understanding mechanisms, and pathological outcomes, as well as to test therapeutic interventions. A variety of closed head models of mTBI that incorporate different aspects (i.e., biomechanics) of the mTBI have been reported. The aim of the current review was to compile a comprehensive list of the closed head mTBI rodent models, along with the common data elements, and outcomes, with the goal to summarize the current state of the field. Publications were identified from a search of PubMed and Web of Science and screened for eligibility following PRISMA guidelines. Articles were included that were closed head injuries in which the authors classified the injury as mild in rats or mice. Injury model and animal-specific common data elements, as well as behavioral and histological outcomes, were collected and compiled from a total of 402 articles. Our results outline the wide variety of methods used to model mTBI. We also discovered that female rodents and both young and aged animals are under-represented in experimental mTBI studies. Our findings will aid in providing context comparing the injury models and provide a starting point for the selection of the most appropriate model of mTBI to address a specific hypothesis. We believe this review will be a useful starting place for determining what has been done and what knowledge is missing in the field to reduce the burden of mTBI.
Collapse
Affiliation(s)
- Colleen N. Bodnar
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kelly N. Roberts
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Emma K. Higgins
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Adam D. Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
22
|
Hiskens MI, Angoa-Pérez M, Schneiders AG, Vella RK, Fenning AS. Modeling sports-related mild traumatic brain injury in animals-A systematic review. J Neurosci Res 2019; 97:1194-1222. [PMID: 31135069 DOI: 10.1002/jnr.24472] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Sports-related head trauma has emerged as an important public health issue, as mild traumatic brain injuries (mTBIs) may result in neurodegenerative disorders such as chronic traumatic encephalopathy (CTE). Research into mTBI and CTE pathophysiology are difficult to undertake in athletes, with observational trials and post-mortem analysis the current mainstays. Thus, animal models play an important role in the study of mTBI, however, traditional animal models have focused on acute, severe injuries rather than the more typical mTBI's seen in sport injuries. Recently, a number of animal models have been developed that are both appropriately scaled and biomechanically relevant to the forces sustained by athletes. This review aimed to examine the literature for variables included in these animal models, and the resulting neurotrauma as evidenced by pathology and behavioral deficits. A systematic search of the literature was performed in multiple electronic databases. The inclusion criteria required mimicry of athlete mTBI conditions: freedom of head movement, lack of surgical alteration of the skull, and application of direct contact force. Studies were analyzed for variables including apparatus design features (impact force, change in animal head velocity, and kinetic energy transfer to the head), demonstrated pathology (phosphorylated tau, TDP-43 aggregation, diffuse axonal injury, gliosis, cytokine inflammation response, and genetic integrity), and behavioral changes. These studies suggested that appropriate animal models can assist in understanding the pathological and functional outcomes of athlete mTBI, and could be used as a platform for future studies of diagnostic/prognostic markers and in the development of treatment interventions.
Collapse
Affiliation(s)
- Matthew I Hiskens
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| | - Mariana Angoa-Pérez
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan.,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Anthony G Schneiders
- School of Health, Medical and Applied Sciences, Central Queensland University, Branyan, Australia
| | - Rebecca K Vella
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| | - Andrew S Fenning
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| |
Collapse
|
23
|
Tabor J, Collins R, Debert CT, Shultz SR, Mychasiuk R. Neuroendocrine Whiplash: Slamming the Breaks on Anabolic-Androgenic Steroids Following Repetitive Mild Traumatic Brain Injury in Rats May Worsen Outcomes. Front Neurol 2019; 10:481. [PMID: 31133974 PMCID: PMC6517549 DOI: 10.3389/fneur.2019.00481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/23/2019] [Indexed: 11/13/2022] Open
Abstract
Sport-related concussion is an increasingly common injury among adolescents, with repetitive mild traumatic brain injuries (RmTBI) being a significant risk factor for long-term neurobiological and psychological consequences. It is not uncommon for younger professional athletes to consume anabolic-androgenic steroids (AAS) in an attempt to enhance their performance, subjecting their hormonally sensitive brains to potential impairment during neurodevelopment. Furthermore, RmTBI produces acute neuroendocrine dysfunction, specifically in the anterior pituitary, disrupting the hypothalamic-pituitary adrenal axis, lowering cortisol secretion that is needed to appropriately respond to injury. Some AAS users exhibit worse symptoms post-RmTBI if they quit their steroid regime. We sought to examine the pathophysiological outcomes associated with the abrupt cessation of the commonly abused AAS, Metandienone (Met) on RmTBI outcomes in rats. Prior to injury, adolescent male rats received either Met or placebo, and exercise. Rats were then administered RmTBIs or sham injuries, followed by steroid and exercise cessation (SEC) or continued treatment. A behavioral battery was conducted to measure outcomes consistent with clinical representations of post-concussion syndrome and chronic AAS exposure, followed by analysis of serum hormone levels, and qRT-PCR for mRNA expression and telomere length. RmTBI increased loss of consciousness and anxiety-like behavior, while also impairing balance and short-term working memory. SEC induced hyperactivity while Met treatment alone increased depressive-like behavior. There were cumulative effects whereby RmTBI and SEC exacerbated anxiety and short-term memory outcomes. mRNA expression in the prefrontal cortex, amygdala, hippocampus, and pituitary were modified in response to Met and SEC. Analysis of telomere length revealed the negative impact of SEC while Met and SEC produced changes in serum levels of testosterone and corticosterone. We identified robust changes in mRNA to serotonergic circuitry, neuroinflammation, and an enhanced stress response. Interestingly, Met treatment promoted glucocorticoid secretion after injury, suggesting that maintained AAS may be more beneficial than abstaining after mTBI.
Collapse
Affiliation(s)
- Jason Tabor
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Reid Collins
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Chantel T Debert
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Division of Physical Medicine and Rehabilitation, Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Sta Maria NS, Sargolzaei S, Prins ML, Dennis EL, Asarnow RF, Hovda DA, Harris NG, Giza CC. Bridging the gap: Mechanisms of plasticity and repair after pediatric TBI. Exp Neurol 2019; 318:78-91. [PMID: 31055004 DOI: 10.1016/j.expneurol.2019.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/09/2019] [Accepted: 04/25/2019] [Indexed: 01/25/2023]
Abstract
Traumatic brain injury is the leading cause of death and disability in the United States, and may be associated with long lasting impairments into adulthood. The multitude of ongoing neurobiological processes that occur during brain maturation confer both considerable vulnerability to TBI but may also provide adaptability and potential for recovery. This review will examine and synthesize our current understanding of developmental neurobiology in the context of pediatric TBI. Delineating this biology will facilitate more targeted initial care, mechanism-based therapeutic interventions and better long-term prognostication and follow-up.
Collapse
Affiliation(s)
- Naomi S Sta Maria
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, 1501 San Pablo Street, ZNI115, Los Angeles, CA 90033, United States of America.
| | - Saman Sargolzaei
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America.
| | - Mayumi L Prins
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Steve Tisch BrainSPORT Program, University of California at Los Angeles, Los Angeles, CA, United States of America.
| | - Emily L Dennis
- Brigham and Women's Hospital/Harvard University and Department of Psychology, Stanford University, 1249 Boylston Street, Boston, MA 02215, United States of America.
| | - Robert F Asarnow
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Box 951759, 760 Westwood Plaza, 48-240C Semel Institute, Los Angeles, CA 90095-1759, United States of America.
| | - David A Hovda
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Department of Medical and Molecular Pharmacology, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562 & Semel 18-228A, Los Angeles, CA 90095-6901, United States of America.
| | - Neil G Harris
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States of America.
| | - Christopher C Giza
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Steve Tisch BrainSPORT Program, University of California at Los Angeles, Los Angeles, CA, United States of America; Division of Pediatric Neurology, Mattel Children's Hospital - UCLA, Los Angeles, CA, United States of America.
| |
Collapse
|
25
|
Salberg S, Yamakawa G, Christensen J, Kolb B, Mychasiuk R. Assessment of a nutritional supplement containing resveratrol, prebiotic fiber, and omega-3 fatty acids for the prevention and treatment of mild traumatic brain injury in rats. Neuroscience 2017; 365:146-157. [PMID: 28988852 DOI: 10.1016/j.neuroscience.2017.09.053] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 12/18/2022]
Abstract
Children and adolescents have the highest rates of traumatic brain injury (TBI), with mild TBI (mTBI) accounting for most of these injuries. Adolescents are particularly vulnerable and often suffer from post-injury symptomologies that may persist for months. We hypothesized that the combination of resveratrol (RES), prebiotic fiber (PBF), and omega-3 fatty acids (docosahexaenoic acid (DHA)) would be an effective therapeutic supplement for the mitigation of mTBI outcomes in the developing brain. Adolescent male and female Sprague-Dawley rats were randomly assigned to the supplement (3S) or control condition, which was followed by a mTBI or sham insult. A behavioral test battery designed to examine symptomologies commonly associated with mTBI was administered. Following the test battery, tissue was collected from the prefrontal cortex (PFC) and primary auditory cortex for Golgi-Cox analysis of spine density, and for changes in expression of 6 genes (Aqp4, Gfap, Igf1, Nfl, Sirt1, and Tau). 3S treatment altered the behavioral performance of sham animals indicating that dietary manipulations modify premorbid characteristics. 3S treatment prevented injury-related deficits in the longer-term behavior measures, medial prefrontal cortex (mPFC) spine density, and levels of Aqp4, Gfap, Igf1, Nfl, and Sirt1 expression in the PFC. Although not fully protective, treatment with the supplement significantly improved post-mTBI function and warrants further investigation.
Collapse
Affiliation(s)
- Sabrina Salberg
- Department of Psychology, The University of Calgary, Calgary, AB, Canada
| | - Glenn Yamakawa
- Department of Psychology, The University of Calgary, Calgary, AB, Canada
| | | | - Bryan Kolb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Richelle Mychasiuk
- Department of Psychology, The University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
26
|
Candy S, Ma I, McMahon JM, Farrell M, Mychasiuk R. Staying in the game: a pilot study examining the efficacy of protective headgear in an animal model of mild traumatic brain injury (mTBI). Brain Inj 2017; 31:1521-1529. [PMID: 28972405 DOI: 10.1080/02699052.2017.1363407] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PRIMARY OBJECTIVE Rugby is one of the few contact sports that do not mandate protective headgear, possibly because studies have shown poor efficacy for protection related to concussion pathology with existing headguards. RESEARCH DESIGN Following innovative material technology utilization to produce headgear believed to have protective capabilities, this study examined the effects of a soft-shell headgear constructed from a novel viscoelastic material, on both behaviour and serum biomarkers after high and average impact force mild traumatic brain injuries (mTBI). METHODS AND PROCEDURES Seventy-five male Sprague Dawley rats were divided into five groups: control, average - 37G impact, with and without headgear, and high - 106G impact, with and without headgear. Rats were sacrificed at 3 or 48 hours and serum samples were analyzed for levels of TNF-α, NEF-L, and GFAP. Animals sacrificed at 48 hours also underwent testing for balance and motor coordination, and exploratory/locomotor behaviour. MAIN OUTCOMES AND RESULTS The novel headgear offered significant protection against mTBI symptomology and biomarkers in the group that experienced an average impact force, but only moderated protection for the animals in the high impact group. CONCLUSIONS This innovative headgear may prevent some of the negative sequel associated with concussion pathology.
Collapse
Affiliation(s)
- Sydney Candy
- a Hotchkiss Brain Institute , University of Calgary , Calgary AB.,b Alberta Children's Hospital Research Institute , Canada
| | - Irene Ma
- b Alberta Children's Hospital Research Institute , Canada.,c Department of Psychology , University of Calgary , Calgary AB
| | - Jill M McMahon
- d Galway Neuroscience Centre , School of Natural Sciences, National University of Ireland Galway , Galway , Ireland
| | - Michael Farrell
- e Department of Neuropathology , Beaumont Hospital , Dublin , Ireland
| | - Richelle Mychasiuk
- a Hotchkiss Brain Institute , University of Calgary , Calgary AB.,b Alberta Children's Hospital Research Institute , Canada.,c Department of Psychology , University of Calgary , Calgary AB
| |
Collapse
|
27
|
Yu K, Seal ML, Reyes J, Godfrey C, Anderson V, Adamson C, Ryan NP, Hearps SJC, Catroppa C. Brain volumetric correlates of inhibition and cognitive flexibility 16 years following childhood traumatic brain injury. J Neurosci Res 2017; 96:642-651. [PMID: 28675465 DOI: 10.1002/jnr.24087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/06/2017] [Accepted: 05/01/2017] [Indexed: 01/08/2023]
Abstract
Executive functions (EFs), such as inhibition and cognitive flexibility, are essential for everyday functioning, including regulation of socially appropriate emotional responses. These skills develop during childhood and continue maturing into early adulthood. The current study aimed to investigate the very long-term impact of childhood traumatic brain injury (TBI) on inhibition and cognitive flexibility, and to examine whether global white matter is associated with these abilities. Twenty-eight young adult survivors of childhood TBI (mean age at 16-year follow-up = 21.67 years, SD = 2.70) and 16 typically developing controls (TDCs), group-matched for age, sex, and socioeconomic status, completed tests of inhibition and cognitive flexibility and underwent structural MRI. Survivors of childhood TBI did not significantly differ from TDCs on EF or white matter volume. However, the relationship between EF and white matter volume differed between survivors of TBI and TDCs. Survivors of TBI did not mimic the brain behavior relationship that characterized EF in TDCs. The inverse brain behavior relationship, exhibited by childhood TBI survivors, suggests disruptions in the whole brain underpinning EF following childhood TBI.
Collapse
Affiliation(s)
- Kelleynne Yu
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Marc L Seal
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Jonathan Reyes
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Celia Godfrey
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Vicki Anderson
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Department of Psychology, Royal Children's Hospital, Parkville, Victoria, Australia.,Melbourne School of Psychological Sciences (MSPS), University of Melbourne, Parkville, Victoria, Australia
| | - Chris Adamson
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Nicholas P Ryan
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Melbourne School of Psychological Sciences (MSPS), University of Melbourne, Parkville, Victoria, Australia
| | - Stephen J C Hearps
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Cathy Catroppa
- Clinical Sciences, Murdoch Childrens Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Department of Psychology, Royal Children's Hospital, Parkville, Victoria, Australia.,Melbourne School of Psychological Sciences (MSPS), University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
28
|
Manipulating cognitive reserve: Pre-injury environmental conditions influence the severity of concussion symptomology, gene expression, and response to melatonin treatment in rats. Exp Neurol 2017; 295:55-65. [PMID: 28579327 DOI: 10.1016/j.expneurol.2017.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/21/2017] [Accepted: 06/01/2017] [Indexed: 11/22/2022]
Abstract
In an effort to understand the factors that contribute to heterogeneity in outcomes often associated with mTBI in youth, this study examined the role of premorbid differences in cognitive reserve on post-concussive symptoms (PCS), molecular markers, and treatment response. Male and female rats matured in one of three environmental conditions (Stress, Enrichment, Control), received a mTBI in adolescence, and were randomized to melatonin or placebo treatment. All animals underwent a behavioural test battery designed to examine PCS. Using prefrontal cortex and hippocampus tissue, expression of 9 genes was assessed in an effort to determine how the brain's epigenome was influenced by cognitive reserve, mTBI, and melatonin. Enrichment increased cognitive reserve (CR) and prevented lingering symptoms. Conversely, stress was associated with progressive worsening and manifestation of PCS in the longer-term. Melatonin was able to restore baseline function for control and enriched animals, but was ineffective for the stress condition. Epigenetic change in the prefrontal cortex was largely driven by the injury, while gene expression changes in the hippocampus were dependent upon cognitive reserve. The occurrence and severity of PCS is dependent upon a complex and multifaceted array of factors that modify behavioural and epigenetic responses to mTBI and its treatment.
Collapse
|
29
|
Sex-dependent changes in neuronal morphology and psychosocial behaviors after pediatric brain injury. Behav Brain Res 2016; 319:48-62. [PMID: 27829127 DOI: 10.1016/j.bbr.2016.10.045] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 10/28/2016] [Accepted: 10/28/2016] [Indexed: 12/12/2022]
Abstract
Chronic social behavior problems after pediatric traumatic brain injury (TBI) significantly contribute to poor quality of life for survivors. Using a well-characterized mouse model of early childhood TBI, we have previously demonstrated that young brain-injured mice develop social deficits by adulthood. As biological sex may influence both normal and aberrant social development, we here evaluated potential sex differences in post-TBI psychosocial deficits by comparing the behavior of male and female mice at adulthood (8 weeks post-injury). Secondly, we hypothesized that pediatric TBI would influence neuronal morphology identified by Golgi-Cox staining in the hippocampus and prefrontal cortex, regions involved in social cognition and behavior, before the onset of social problems (3 weeks post-injury). Morphological analysis of pyramidal neurons in the ipsilateral prefrontal cortex and granule cells of the hippocampal dentate gyrus revealed a reduction in dendritic complexity after pediatric TBI. This was most apparent in TBI males, whereas neurons from females were less affected. At adulthood, consistent with previous studies, TBI males showed deficits in sociability and social recognition. TBI females also showed a reduction in sociability, but intact social recognition and increased sociosexual avoidance. Together, these findings indicate that sex is a determinant of regional neuroplasticity and social outcomes after pediatric TBI. Reduced neuronal complexity in the prefrontal cortex and hippocampus, several weeks after injury in male mice, appears to precede the subsequent emergence of social deficits. Sex-specific alterations in the social brain network are thus implicated as an underlying mechanism of social dysfunction after pediatric TBI.
Collapse
|
30
|
Transcriptional and Epigenetic Regulation in Injury-Mediated Neuronal Dendritic Plasticity. Neurosci Bull 2016; 33:85-94. [PMID: 27730386 DOI: 10.1007/s12264-016-0071-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/27/2016] [Indexed: 12/26/2022] Open
Abstract
Injury to the nervous system induces localized damage in neural structures and neuronal death through the primary insult, as well as delayed atrophy and impaired plasticity of the delicate dendritic fields necessary for interneuronal communication. Excitotoxicity and other secondary biochemical events contribute to morphological changes in neurons following injury. Evidence suggests that various transcription factors are involved in the dendritic response to injury and potential therapies. Transcription factors play critical roles in the intracellular regulation of neuronal morphological plasticity and dendritic growth and patterning. Mounting evidence supports a crucial role for epigenetic modifications via histone deacetylases, histone acetyltransferases, and DNA methyltransferases that modify gene expression in neuronal injury and repair processes. Gene regulation through epigenetic modification is of great interest in neurotrauma research, and an early picture is beginning to emerge concerning how injury triggers intracellular events that modulate such responses. This review provides an overview of injury-mediated influences on transcriptional regulation through epigenetic modification, the intracellular processes involved in the morphological consequences of such changes, and potential approaches to the therapeutic manipulation of neuronal epigenetics for regulating gene expression to facilitate growth and signaling through dendritic arborization following injury.
Collapse
|
31
|
Polimanti R, Chen CY, Ursano RJ, Heeringa SG, Jain S, Kessler RC, Nock MK, Smoller JW, Sun X, Gelernter J, Stein MB. Cross-Phenotype Polygenic Risk Score Analysis of Persistent Post-Concussive Symptoms in U.S. Army Soldiers with Deployment-Acquired Traumatic Brain Injury. J Neurotrauma 2016; 34:781-789. [PMID: 27439997 DOI: 10.1089/neu.2016.4550] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) contributes to the increased rates of suicide and post-traumatic stress disorder in military personnel and veterans, and it is also associated with the risk for neurodegenerative and psychiatric disorders. A cross-phenotype high-resolution polygenic risk score (PRS) analysis of persistent post-concussive symptoms (PCS) was conducted in 845 U.S. Army soldiers who sustained TBI during their deployment. We used a prospective longitudinal survey of three brigade combat teams to assess deployment-acquired TBI and persistent physical, cognitive, and emotional PCS. PRS was derived from summary statistics of large genome-wide association studies of Alzheimer's disease, Parkinson's disease, schizophrenia, bipolar disorder, and major depressive disorder (MDD); and for years of schooling, college completion, childhood intelligence, infant head circumference (IHC), and adult intracranial volume. Although our study had more than 95% of statistical power to detect moderate-to-large effect sizes, no association was observed with neurodegenerative and psychiatric disorders, suggesting that persistent PCS does not share genetic components with these traits to a moderate-to-large degree. We observed a significant finding: subjects with high IHC PRS recovered better from cognitive/emotional persistent PCS than the other individuals (R2 = 1.11%; p = 3.37 × 10-3). Enrichment analysis identified two significant Gene Ontology (GO) terms related to this result: GO:0050839∼Cell adhesion molecule binding (p = 8.9 × 10-6) and GO:0050905∼Neuromuscular process (p = 9.8 × 10-5). In summary, our study indicated that the genetic predisposition to persistent PCS after TBI does not have substantial overlap with neurodegenerative and psychiatric diseases, but mechanisms related to early brain growth may be involved.
Collapse
Affiliation(s)
- Renato Polimanti
- 1 Department of Psychiatry, Yale School of Medicine and VA CT Healthcare Center , West Haven, Connecticut
| | - Chia-Yen Chen
- 2 Department of Psychiatry, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard , Cambridge, Massachusetts
| | - Robert J Ursano
- 3 Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Steven G Heeringa
- 4 Institute for Social Research, University of Michigan , Ann Arbor, Michigan
| | - Sonia Jain
- 5 Department of Family Medicine and Public Health, University of California , La Jolla, California
| | - Ronald C Kessler
- 6 Department of Health Care Policy, Harvard Medical School , Boston, Massachusetts
| | - Matthew K Nock
- 7 Department of Psychology, Harvard University , Cambridge, Massachusetts
| | - Jordan W Smoller
- 2 Department of Psychiatry, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard , Cambridge, Massachusetts
| | - Xiaoying Sun
- 5 Department of Family Medicine and Public Health, University of California , La Jolla, California
| | - Joel Gelernter
- 8 Departments of Psychiatry, Genetics, and Neuroscience, Yale School of Medicine and VA CT Healthcare Center , West Haven, Connecticut
| | - Murray B Stein
- 5 Department of Family Medicine and Public Health, University of California , La Jolla, California.,9 Department of Psychiatry, University of California , La Jolla, California.,10 VA San Diego Healthcare System , San Diego, California
| |
Collapse
|
32
|
Cetin I, Tezdig I, Tarakcioglu MC, Kadak MT, Demirel OF, Ozer OF. Serum levels of glial fibrillary acidic protein and Nogo-A in children with autism spectrum disorders. Biomarkers 2016; 21:614-8. [DOI: 10.3109/1354750x.2016.1171901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ihsan Cetin
- Department of Nutrition and Dietetics, School of Health, Batman University, Batman, Turkey
| | - Ihsan Tezdig
- Department of Chemistry, Institute of Science, Batman University, Batman, Turkey
| | - Mahmut Cem Tarakcioglu
- Clinic of Child and Adolescent Psychiatry, Bakirköy Dr Sadi Konuk Training and Educatin Hospital, Istanbul University, İstanbul, Turkey
| | - Muhammed Tayyib Kadak
- Department of Child and Adolescent Psychiatry, Cerrahpaşa School of Medicine, Istanbul University, İstanbul, Turkey
| | - Omer Faruk Demirel
- Department of Psychiatry, Cerrahpaşa School of Medicine, Istanbul University, İstanbul, Turkey
| | - Omer Faruk Ozer
- Department of Biochemistry, BezmiAlem Vakif University, İstanbul, Turkey
| |
Collapse
|
33
|
Hehar H, Mychasiuk R. The use of telomere length as a predictive biomarker for injury prognosis in juvenile rats following a concussion/mild traumatic brain injury. Neurobiol Dis 2016; 87:11-8. [DOI: 10.1016/j.nbd.2015.12.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/23/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023] Open
|
34
|
Abstract
Due to a high incidence of traumatic brain injury (TBI) in children and adolescents, age-specific studies are necessary to fully understand the long-term consequences of injuries to the immature brain. Preclinical and translational research can help elucidate the vulnerabilities of the developing brain to insult, and provide model systems to formulate and evaluate potential treatments aimed at minimizing the adverse effects of TBI. Several experimental TBI models have therefore been scaled down from adult rodents for use in juvenile animals. The following chapter discusses these adapted models for pediatric TBI, and the importance of age equivalence across species during model development and interpretation. Many neurodevelopmental processes are ongoing throughout childhood and adolescence, such that neuropathological mechanisms secondary to a brain insult, including oxidative stress, metabolic dysfunction and inflammation, may be influenced by the age at the time of insult. The long-term evaluation of clinically relevant functional outcomes is imperative to better understand the persistence and evolution of behavioral deficits over time after injury to the developing brain. Strategies to modify or protect against the chronic consequences of pediatric TBI, by supporting the trajectory of normal brain development, have the potential to improve quality of life for brain-injured children.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Jaclyn Carlson
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Linda J Noble-Haeusslein
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
- Department of Physical Therapy and Rehabilitation Science, University of California School of Medicine, 513 Parnassus Ave., HSE 814, San Francisco, CA, 94143, USA.
| |
Collapse
|
35
|
Hehar H, Yeates K, Kolb B, Esser MJ, Mychasiuk R. Impulsivity and Concussion in Juvenile Rats: Examining Molecular and Structural Aspects of the Frontostriatal Pathway. PLoS One 2015; 10:e0139842. [PMID: 26448536 PMCID: PMC4598031 DOI: 10.1371/journal.pone.0139842] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/17/2015] [Indexed: 01/09/2023] Open
Abstract
Impulsivity and poor executive control have been implicated in the pathogenesis of many developmental and neuropsychiatric disorders. Similarly, concussions/mild traumatic brain injuries (mTBI) have been associated with increased risk for neuropsychiatric disorders and the development of impulsivity and inattention. Researchers and epidemiologists have therefore considered whether or not concussions induce symptoms of attention-deficit/hyperactivity disorder (ADHD), or merely unmask impulsive tendencies that were already present. The purpose of this study was to determine if a single concussion in adolescence could induce ADHD-like impulsivity and impaired response inhibition, and subsequently determine if inherent impulsivity prior to a pediatric mTBI would exacerbate post-concussion symptomology with a specific emphasis on impulsive and inattentive behaviours. As these behaviours are believed to be associated with the frontostriatal circuit involving the nucleus accumbens (NAc) and the prefrontal cortex (PFC), the expression patterns of 8 genes (Comt, Drd2, Drd3, Drd4, Maoa, Sert, Tph1, and Tph2) from these two regions were examined. In addition, Golgi-Cox staining of medium spiny neurons in the NAc provided a neuroanatomical examination of mTBI-induced structural changes. The study found that a single early brain injury could induce impulsivity and impairments in response inhibition that were more pronounced in males. Interestingly, when animals with inherent impulsivity experienced mTBI, injury-related deficits were exacerbated in female animals. The single concussion increased dendritic branching, but reduced synaptic density in the NAc, and these changes were likely associated with the increase in impulsivity. Finally, mTBI-induced impulsivity was associated with modifications to gene expression that differed dramatically from the gene expression pattern associated with inherent impulsivity, despite very similar behavioural phenotypes. Our findings suggest the need to tailor treatment strategies for mTBI in light of an individual's premorbid characteristics, given significant differences in molecular profiles of the frontostriatal circuits that depend upon sex and the etiology of the behavioural phenotype.
Collapse
Affiliation(s)
- Harleen Hehar
- Alberta Children’s Hospital Research Institute, University of Calgary, Faculty of Medicine, Calgary, Canada
| | - Keith Yeates
- Alberta Children’s Hospital Research Institute, University of Calgary, Department of Psychology, Calgary, Canada
| | - Bryan Kolb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Canada
| | - Michael J. Esser
- Alberta Children’s Hospital Research Institute, University of Calgary, Faculty of Medicine, Calgary, Canada
| | - Richelle Mychasiuk
- Alberta Children’s Hospital Research Institute, University of Calgary, Faculty of Medicine, Calgary, Canada
- * E-mail:
| |
Collapse
|