1
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 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] [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
|
Badaut J, Hippauf L, Malinconi M, Noarbe BP, Obenaus A, Dubois CJ. Endocannabinoid-mediated rescue of somatosensory cortex activity, plasticity and related behaviors following an early in life concussion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.577914. [PMID: 38352553 PMCID: PMC10862852 DOI: 10.1101/2024.01.30.577914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Due to the assumed plasticity of immature brain, early in life brain alterations are thought to lead to better recoveries in comparison to the mature brain. Despite clinical needs, how neuronal networks and associated behaviors are affected by early in life brain stresses, such as pediatric concussions, have been overlooked. Here we provide first evidence in mice that a single early in life concussion durably increases neuronal activity in the somatosensory cortex into adulthood, disrupting neuronal integration while the animal is performing sensory-related tasks. This represents a previously unappreciated clinically relevant mechanism for the impairment of sensory-related behavior performance. Furthermore, we demonstrate that pharmacological modulation of the endocannabinoid system a year post-concussion is well-suited to rescue neuronal activity and plasticity, and to normalize sensory-related behavioral performance, addressing the fundamental question of whether a treatment is still possible once post-concussive symptoms have developed, a time-window compatible with clinical treatment.
Collapse
Affiliation(s)
- J Badaut
- Univ. Bordeaux, CNRS, CRMSB, UMR 5536, F-33000 Bordeaux, France
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - L Hippauf
- Univ. Bordeaux, CNRS, CRMSB, UMR 5536, F-33000 Bordeaux, France
| | - M Malinconi
- Univ. Bordeaux, CNRS, CRMSB, UMR 5536, F-33000 Bordeaux, France
| | - B P Noarbe
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - A Obenaus
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - C J Dubois
- Univ. Bordeaux, CNRS, CRMSB, UMR 5536, F-33000 Bordeaux, France
| |
Collapse
|
3
|
Gorthy AS, Balleste AF, Placeres-Uray F, Atkins CM. Chronic Stress in Early Development and Effects on Traumatic Brain Injury Outcome. ADVANCES IN NEUROBIOLOGY 2024; 42:179-204. [PMID: 39432043 DOI: 10.1007/978-3-031-69832-3_9] [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
In recent years, significant advances have been made in the study of mild traumatic brain injury (mTBI). Complete recovery from mTBI normally requires days to weeks, yet a subset of the population suffers from symptoms for weeks to months after injury. The risk factors for these prolonged symptoms have not yet been fully understood. In this chapter, we address one proposed risk factor, early life stress (ELS) and its influence on mTBI recovery. To study the effects of ELS on mTBI recovery, accepted animal models of ELS, including maternal separation, limited bedding and nesting, and chronic unpredictable stress, have been implemented. Combining these ELS models with standardized mTBI models, such as fluid percussion injury or controlled cortical impact, has allowed for a deeper understanding of the neuronal, hormonal, and cognitive changes that occur after mTBI following ELS. These preclinical findings are being used to understand how adverse childhood experiences may predispose a subset of individuals to poorer recovery after mTBI.
Collapse
Affiliation(s)
- Aditi S Gorthy
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alyssa F Balleste
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fabiola Placeres-Uray
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Coleen M Atkins
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
4
|
Moschonas EH, Annas EM, Zamudio-Flores J, Jarvis JM, Lajud N, Bondi CO, Kline AE. Pediatric Traumatic Brain Injury: Models, Therapeutics, and Outcomes. ADVANCES IN NEUROBIOLOGY 2024; 42:147-163. [PMID: 39432041 DOI: 10.1007/978-3-031-69832-3_7] [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
Pediatric traumatic brain injury (TBI) is a significant healthcare issue, but potential treatments are absent despite robust investigation in several clinical trials. Factors attributed to clinical TBI, such as heterogeneity of injury and single-dose pharmacological treatments as well as timing of administration, may be reasons for the negative studies. Preclinical models of TBI can reduce some of the impediments by highlighting differences in injury depending on injury severity and location and by conducting dose response studies, thus providing better therapeutic targets and pharmacological profiles for clinical use. In this chapter, there were sufficient reports to make comparisons between the models in terms of pathophysiology, behavioral dysfunction, and the efficacy of therapeutic interventions. The models used to date include controlled cortical impact (CCI), weight drop, fluid percussion, and abusive head trauma. Several therapeutics were identified after CCI injury but none in the other models, which underscores the need for studies evaluating the therapies reported after CCI injury as well as novel potential approaches.
Collapse
Affiliation(s)
- Eleni H Moschonas
- Department of Physical Medicine & Rehabilitation, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ellen M Annas
- Department of Physical Medicine & Rehabilitation, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Zamudio-Flores
- Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - Jessica M Jarvis
- Department of Physical Medicine & Rehabilitation, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Naima Lajud
- Instituto de Investigaciones sobre los Recursos Naturales - Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacan, Mexico
| | - Corina O Bondi
- Departments of Physical Medicine & Rehabilitation and Neurobiology, Center for Neuroscience, and Safar Center for Resuscitation Research, John G. Rangos Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anthony E Kline
- Departments of Physical Medicine & Rehabilitation, Critical Care Medicine, and Psychology, and Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, John G. Rangos Research Center, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
van der Horn HJ, Dodd AB, Wick TV, Robertson‐Benta CR, McQuaid JR, Hittson AK, Ling JM, Zotev V, Ryman SG, Erhardt EB, Phillips JP, Campbell RA, Sapien RE, Mayer AR. Neural correlates of cognitive control deficits in pediatric mild traumatic brain injury. Hum Brain Mapp 2023; 44:6173-6184. [PMID: 37800467 PMCID: PMC10619369 DOI: 10.1002/hbm.26504] [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: 06/06/2023] [Revised: 08/18/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023] Open
Abstract
There is a growing body of research showing that cerebral pathophysiological processes triggered by pediatric mild traumatic brain injury (pmTBI) may extend beyond the usual clinical recovery timeline. It is paramount to further unravel these processes, because the possible long-term cognitive effects resulting from ongoing secondary injury in the developing brain are not known. In the current fMRI study, neural processes related to cognitive control were studied in 181 patients with pmTBI at sub-acute (SA; ~1 week) and early chronic (EC; ~4 months) stages post-injury. Additionally, a group of 162 age- and sex-matched healthy controls (HC) were recruited at equivalent time points. Proactive (post-cue) and reactive (post-probe) cognitive control were examined using a multimodal attention fMRI paradigm for either congruent or incongruent stimuli. To study brain network function, the triple-network model was used, consisting of the executive and salience networks (collectively known as the cognitive control network), and the default mode network. Additionally, whole-brain voxel-wise analyses were performed. Decreased deactivation was found within the default mode network at the EC stage following pmTBI during both proactive and reactive control. Voxel-wise analyses revealed sub-acute hypoactivation of a frontal area of the cognitive control network (left pre-supplementary motor area) during proactive control, with a reversed effect at the EC stage after pmTBI. Similar effects were observed in areas outside of the triple-network during reactive control. Group differences in activation during proactive control were limited to the visual domain, whereas for reactive control findings were more pronounced during the attendance of auditory stimuli. No significant correlations were present between task-related activations and (persistent) post-concussive symptoms. In aggregate, current results show alterations in neural functioning during cognitive control in pmTBI up to 4 months post-injury, regardless of clinical recovery. We propose that subacute decreases in activity reflect a general state of hypo-excitability due to the injury, while early chronic hyperactivation represents a compensatory mechanism to prevent default mode interference and to retain cognitive control.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Josef M. Ling
- The Mind Research Network/LBERIAlbuquerqueNew MexicoUSA
| | - Vadim Zotev
- The Mind Research Network/LBERIAlbuquerqueNew MexicoUSA
| | | | - Erik B. Erhardt
- Department of Mathematics and StatisticsUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | | | - Richard A. Campbell
- Department of Psychiatry & Behavioral SciencesUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Robert E. Sapien
- Department of Emergency MedicineUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Andrew R. Mayer
- The Mind Research Network/LBERIAlbuquerqueNew MexicoUSA
- Department of Psychiatry & Behavioral SciencesUniversity of New MexicoAlbuquerqueNew MexicoUSA
- Department of PsychologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
| |
Collapse
|
6
|
Hoogenboom WS, Rubin TG, Ambadipudi K, Cui MH, Ye K, Foster H, Elkouby E, Liu J, Branch CA, Lipton ML. Evolving brain and behaviour changes in rats following repetitive subconcussive head impacts. Brain Commun 2023; 5:fcad316. [PMID: 38046094 PMCID: PMC10691880 DOI: 10.1093/braincomms/fcad316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/26/2023] [Accepted: 11/19/2023] [Indexed: 12/05/2023] Open
Abstract
There is growing concern that repetitive subconcussive head impacts, independent of concussion, alter brain structure and function, and may disproportionately affect the developing brain. Animal studies of repetitive subconcussive head impacts are needed to begin to characterize the pathological basis and mechanisms underlying imaging and functional effects of repetitive subconcussive head impacts seen in humans. Since repetitive subconcussive head impacts have been largely unexplored in animals, we aimed to characterize the evolution of imaging, behavioural and pathological effects of repetitive subconcussive head impacts in awake adolescent rodents. Awake male and female Sprague Dawley rats (postnatal Day 35) received 140 closed-head impacts over the course of a week. Impacted and sham-impacted animals were restrained in a plastic cone, and unrestrained control animals were included to account for effects of restraint and normal development. Animals (n = 43) underwent repeated diffusion tensor imaging prior to and over 1 month following the final impact. A separate cohort (n = 53) was assessed behaviourally for fine motor control, emotional-affective behaviour and memory at acute and chronic time points. Histological and immunohistochemical analyses, which were exploratory in nature due to smaller sample sizes, were completed at 1 month following the final impact. All animals tolerated the protocol with no overt changes in behaviour or stigmata of traumatic brain injury, such as alteration of consciousness, intracranial haemorrhage or skull fracture. We detected longitudinal, sex-dependent diffusion tensor imaging changes (fractional anisotropy and axial diffusivity decline) in corpus callosum and external capsule of repetitive subconcussive head impact animals, which diverged from both sham and control. Compared to sham animals, repetitive subconcussive head impact animals exhibited acute but transient mild motor deficits. Repetitive subconcussive head impact animals also exhibited chronic anxiety and spatial memory impairment that differed from the control animals, but these effects were not different from those seen in the sham condition. We observed trends in the data for thinning of the corpus callosum as well as regions with elevated Iba-1 in the corpus callosum and cerebral white matter among repetitive subconcussive head impact animals. While replication with larger study samples is needed, our findings suggest that subconcussive head impacts cause microstructural tissue changes in the developing rat brain, which are detectable with diffusion tensor imaging, with suggestion of correlates in tissue pathology and behaviour. The results point to potential mechanisms underpinning consequences of subconcussive head impacts that have been described in humans. The congruence of our imaging findings with human subconcussive head impacts suggests that neuroimaging could serve as a translational bridge to advance study of injury mechanisms and development of interventions.
Collapse
Affiliation(s)
- Wouter S Hoogenboom
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Clinical Investigation, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Todd G Rubin
- Department of Neurology, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Kamalakar Ambadipudi
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Min-Hui Cui
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Kenny Ye
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Henry Foster
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Esther Elkouby
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Jinyuan Liu
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Craig A Branch
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Michael L Lipton
- Department of Radiology, Columbia University Irving Medical Center, NewYork, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, NewYork, NY 10032, USA
| |
Collapse
|
7
|
Raghupathi R, Prasad R, Fox D, Huh JW. Repeated mild closed head injury in neonatal rats results in sustained cognitive deficits associated with chronic microglial activation and neurodegeneration. J Neuropathol Exp Neurol 2023; 82:707-721. [PMID: 37390808 PMCID: PMC10357947 DOI: 10.1093/jnen/nlad048] [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] [Indexed: 07/02/2023] Open
Abstract
Abusive head trauma in infants is a consequence of multiple episodes of abuse and results in axonal injury, brain atrophy, and chronic cognitive deficits. Anesthetized 11-day-old rats, neurologically equivalent to infants, were subjected to 1 impact/day to the intact skull for 3 successive days. Repeated, but not single impact(s) resulted in spatial learning deficits (p < 0.05 compared to sham-injured animals) up to 5 weeks postinjury. In the first week following single or repetitive brain injury, axonal and neuronal degeneration, and microglial activation were observed in the cortex, white matter, thalamus, and subiculum; the extent of the histopathologic damage was significantly greater in the repetitive-injured animals compared to single-injured animals. At 40 days postinjury, loss of cortical, white matter and hippocampal tissue was evident only in the repetitive-injured animals, along with evidence of microglial activation in the white matter tracts and thalamus. Axonal injury and neurodegeneration were evident in the thalamus up to 40 days postinjury in the repetitive-injured rats. These data demonstrate that while single closed head injury in the neonate rat is associated with pathologic alterations in the acute post-traumatic period, repetitive closed head injury results in sustained behavioral and pathologic deficits reminiscent of infants with abusive head trauma.
Collapse
Affiliation(s)
- Ramesh Raghupathi
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Rupal Prasad
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Douglas Fox
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Koza LA, Pena C, Russell M, Smith AC, Molnar J, Devine M, Serkova NJ, Linseman DA. Immunocal® limits gliosis in mouse models of repetitive mild-moderate traumatic brain injury. Brain Res 2023; 1808:148338. [PMID: 36966959 PMCID: PMC10258892 DOI: 10.1016/j.brainres.2023.148338] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
Successive traumatic brain injuries (TBIs) exacerbate neuroinflammation and oxidative stress. No therapeutics exist for populations at high risk of repetitive mild TBIs (rmTBIs). We explored the preventative therapeutic effects of Immunocal®, a cysteine-rich whey protein supplement and glutathione (GSH) precursor, following rmTBI and repetitive mild-moderate TBI (rmmTBI). Populations that suffer rmTBIs largely go undiagnosed and untreated; therefore, we first examined the potential therapeutic effect of Immunocal® long-term following rmTBI. Mice were treated with Immunocal® prior to, during, and following rmTBI induced by controlled cortical impact until analysis at 2 weeks, 2 months, and 6 months following the last rmTBI. Astrogliosis and microgliosis were measured in cortex at each time point and edema and macrophage infiltration by MRI were analyzed at 2 months post-rmTBI. Immunocal® significantly reduced astrogliosis at 2 weeks and 2 months post-rmTBI. Macrophage activation was observed at 2 months post-rmTBI but Immunocal® had no significant effect on this endpoint. We did not observe significant microgliosis or edema after rmTBI. The dosing regimen was repeated in mice subjected to rmmTBI; however, using this experimental paradigm, we examined the preventative therapeutic effects of Immunocal® at a much earlier timepoint because populations that suffer more severe rmmTBIs are more likely to receive acute diagnosis and treatment. Increases in astrogliosis, microgliosis, and serum neurofilament light (NfL), as well as reductions in the GSH:GSSG ratio, were observed 72 h post-rmmTBI. Immunocal® only significantly reduced microgliosis after rmmTBI. In summary, we report that astrogliosis persists for 2 months post-rmTBI and that inflammation, neuronal damage, and altered redox homeostasis present acutely following rmmTBI. Immunocal® significantly limited gliosis in these models; however, its neuroprotection was partially overwhelmed by repetitive injury. Treatments that modulate distinct aspects of TBI pathophysiology, used in combination with GSH precursors like Immunocal®, may show more protection in these repetitive TBI models.
Collapse
Affiliation(s)
- Lilia A Koza
- University of Denver, Department of Biological Sciences, Denver, CO 80208, United States; University of Denver, Knoebel Institute for Healthy Aging, Denver, CO 80208, United States
| | - Claudia Pena
- University of Denver, Department of Biological Sciences, Denver, CO 80208, United States; University of Denver, Knoebel Institute for Healthy Aging, Denver, CO 80208, United States
| | - Madison Russell
- University of Denver, Department of Biological Sciences, Denver, CO 80208, United States; University of Denver, Knoebel Institute for Healthy Aging, Denver, CO 80208, United States
| | - Alec C Smith
- University of Denver, Department of Biological Sciences, Denver, CO 80208, United States; University of Denver, Knoebel Institute for Healthy Aging, Denver, CO 80208, United States
| | - Jacob Molnar
- University of Denver, Department of Biological Sciences, Denver, CO 80208, United States; University of Denver, Knoebel Institute for Healthy Aging, Denver, CO 80208, United States
| | - Maeve Devine
- University of Denver, Department of Biological Sciences, Denver, CO 80208, United States; University of Denver, Knoebel Institute for Healthy Aging, Denver, CO 80208, United States
| | - Natalie J Serkova
- University of Colorado Cancer Center, Department of Radiology, Aurora, CO 80045, United States
| | - Daniel A Linseman
- University of Denver, Department of Biological Sciences, Denver, CO 80208, United States; University of Denver, Knoebel Institute for Healthy Aging, Denver, CO 80208, United States.
| |
Collapse
|
9
|
Sgro M, Ellens S, Kodila ZN, Christensen J, Li C, Mychasiuk R, Yamakawa GR. Repetitive mild traumatic brain injury alters central and peripheral clock gene expression in the adolescent rat. Neurobiol Sleep Circadian Rhythms 2023; 14:100090. [PMID: 36942266 PMCID: PMC10024151 DOI: 10.1016/j.nbscr.2023.100090] [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: 11/02/2022] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 03/11/2023] Open
Abstract
Mild traumatic brain injury (mTBI) or concussion is a common injury worldwide leading to substantial medical costs and a high burden on society. In adolescents, falls and sports related trauma are often the causes of mTBI. Importantly, critical brain growth and development occurs during this sensitive period making the prospect of a brain injury a worrying phenomenon. Upwards of 70% of patients report circadian disruption following these injuries and this has been shown to impede recovery. Therefore, we sought to determine if core circadian clock gene expression was disrupted in rat model of repetitive mTBI (RmTBI). Male and female adolescent rats (n = 129) received sham or RmTBI. The animals were then euthanized at different times throughout the day and night. Tissue from the hypothalamus, cerebellum, hippocampus, liver, and small intestine were evaluated for the expression of per1, per2, cry1, clock, bmal1 and rev-erb-α. We found most clock genes varied across the day/night indicating circadian expression patterns. In the hypothalamus we found RmTBI altered the expression of cry1 and bmal1 in addition to sex differences in per2, cry1, clock, bmal1 and rev-erb- α. In the cerebellum, per1, per2, cry1, clock, bmal1 and rev-erb-α rhythms were all knocked out by RmTBI in addition to sex differences in cry1, clock and bmal1 expression. We also detected a significant decrease in overall expression of all clock genes in males in the middle of the night. In the hippocampus we found that RmTBI changed the rhythm of rev-erb-α expression in addition to sex differences in bmal1 expression. In the liver we detected strong rhythms in all genes examined, however only per2 expression was knocked out by RmTBI, in addition we also detected sex differences in per2 and cry1. We also detected an overall decrease in female clock gene expression in the early night. In the small intestine, RmTBI altered cry1 expression and there were sex differences in rev-erb-α. These results indicate that RmTBI alters core circadian clock gene expression in the central and peripheral nervous system in a time, tissue and sex dependent manner. This may be disrupting important phase relationships between the brain and peripheral nervous system and contributing to post-injury symptomology and also highlights the importance for time and sex dependent assessment of injury outcomes.
Collapse
Affiliation(s)
- Marissa Sgro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Susanne Ellens
- Sport and Exercise Science, School of Allied Health, Human Services & Sport, La Trobe University, Melbourne, Australia
| | - Zoe N. Kodila
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jennaya Christensen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Crystal Li
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- 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
- Corresponding author. Department of Neuroscience, Central Clinical School, Monash University, 6th Floor, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
10
|
Alipour M, Tebianian M, Tofigh N, Taheri RS, Mousavi SA, Naseri A, Ahmadi A, Munawar N, Shahpasand K. Active immunotherapy against pathogenic Cis pT231-tau suppresses neurodegeneration in traumatic brain injury mouse models. Neuropeptides 2022; 96:102285. [PMID: 36087426 DOI: 10.1016/j.npep.2022.102285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 10/14/2022]
Abstract
Traumatic brain injury (TBI), characterized by acute neurological impairment, is associated with a higher incidence of neurodegenerative diseases, particularly chronic traumatic encephalopathy (CTE), Alzheimer's disease (AD), and Parkinson's disease (PD), whose hallmarks include hyperphosphorylated tau protein. Recently, phosphorylated tau at Thr231 has been shown to exist in two distinct cis and trans conformations. Moreover, targeted elimination of cis P-tau by passive immunotherapy with an appropriate mAb that efficiently suppresses tau-mediated neurodegeneration in severe TBI mouse models has proven to be a useful tool to characterize the neurotoxic role of cis P-tau as an early driver of the tauopathy process after TBI. Here, we investigated whether active immunotherapy can develop sufficient neutralizing antibodies to specifically target and eliminate cis P-tau in the brain of TBI mouse models. First, we explored the therapeutic efficacy of two different vaccines. C57BL/6 J mice were immunized with either cis or trans P-tau conformational peptides plus adjuvant. After rmTBI in mice, we found that cis peptide administration developed a specific Ab that precisely targeted and neutralized cis P-tau, inhibited the development of neuropathology and brain dysfunction, and restored various structural and functional sequelae associated with TBI in chronic phases. In contrast, trans P-tau peptide application not only lacked neuroprotective properties, but also contributed to a number of neuropathological features, including progressive TBI-induced neuroinflammation, widespread tau-mediated neurodegeneration, worsening functional deficits, and brain atrophy. Taken together, our results suggest that active immunotherapy strategies against pathogenic cis P-tau can halt the process of tauopathy and would have profound clinical implications.
Collapse
Affiliation(s)
- Masoume Alipour
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Faculty of Basic Science and Advanced Medical Technologies, Royan Institute, ACECR, Tehran, Iran
| | - Majid Tebianian
- Biotechnology Department, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Nahid Tofigh
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reyhaneh Sadat Taheri
- Department of Motor Behavior, Faculty of Physical Education and Sport Sciences, Allameh Tabataba'i University, Tehran, Iran
| | - Sayed Alireza Mousavi
- Department of Biology, Faculty of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Asal Naseri
- Department of Biology, Faculty of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Amin Ahmadi
- Department of Biomedical Sciences, Tabriz Medical University, Tabriz, Iran
| | - Nayla Munawar
- Department of Chemistry, United Arab Emirates University, United Arab Emirates
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
11
|
Celorrio M, Rhodes J, Shumilov K, Moritz J, Xiao S, Anabayan I, Sauerbeck A, Kummer T, Friess S. Recombinant human erythropoietin induces neuroprotection, activates MAPK/CREB pathway, and rescues fear memory after traumatic brain injury with delayed hypoxemia in mice. Brain Res 2022; 1795:148074. [PMID: 36075467 PMCID: PMC10515732 DOI: 10.1016/j.brainres.2022.148074] [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: 07/10/2022] [Revised: 07/29/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022]
Abstract
Therapeutic interventions targeting secondary insults, such as delayed hypoxemia, provide a unique opportunity for treatment in severe traumatic brain injury (TBI). Erythropoietin (EPO) is a hypoxia-responsive cytokine with important roles in neurodevelopment, neuroprotection and neuromodulation. We hypothesized that recombinant human erythropoietin (rhEPO) administration would mitigate injury in a combined injury model of TBI and delayed hypoxemia. Utilizing a clinically relevant murine model of TBI and delayed hypoxemia, we characterized how ongoing rhEPO administration influenced neurogenesis, neuroprotection, synaptic density and, behavioral outcomes early after TBI, and the impact on long-lasting outcomes 6 months after injury. We employed novel object recognition (NOR) and fear conditioning to assess long-term memory. At 1-month post-injury, we observed a significant increase in cued-fear memory response in the rhEPO-injured mice compared with vehicle-injured mice. This was associated with neuroprotection and neurogenesis in the hippocampus and mitogen-activated protein kinase (MAPK)/cAMP response element-binding protein (CREB) signaling activation and increased of excitatory synaptic density in the amygdala. Early rhEPO treatment after injury reduced neurodegeneration and increased excitatory synaptic density in the hippocampus and amygdala at 6 months post-injury. However at 6 months post-injury (4 months after discontinuation of rhEPO), we did not observe changes in behavioral assessments nor MAPK/CREB pathway activation. In summary, these data demonstrate that ongoing rhEPO treatment initiated at a clinically feasible time point improves neurological, cognitive, and histological outcomes after TBI in the setting of secondary hypoxemic insults.
Collapse
Affiliation(s)
- Marta Celorrio
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - James Rhodes
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Kirill Shumilov
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Jennie Moritz
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Sophia Xiao
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Ilakkia Anabayan
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Andrew Sauerbeck
- Department of Neurology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Terrance Kummer
- Department of Neurology, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Stuart Friess
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
12
|
Bills SE, Schatz J, Hunt E, Varanasi S, Johnston J, Bradshaw J. Neurodevelopmental Outcomes in Preterm Children with Sickle Cell Disease. J Int Neuropsychol Soc 2022; 28:1039-1049. [PMID: 34839840 PMCID: PMC9148381 DOI: 10.1017/s1355617721001338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVES To explore the combined effect of pediatric sickle cell disease (SCD) and preterm birth on cognitive functioning. METHODS Cognitive functioning was examined in children ages 6-8 with high risk SCD genotypes born preterm (n = 20) and full-term (n = 59) and lower risk SCD genotypes/no SCD born preterm (n = 11) and full-term (n = 99) using tests previously shown to be sensitive to SCD-related neurocognitive deficits. Factorial ANOVAs and log linear analyses were conducted to examine the relationship between SCD risk, preterm birth status, and cognitive outcomes. Continuous scores were examined for specific tests. Children were categorized as having an abnormal screening outcome if at least one cognitive score was ≥1.5 standard deviations below the population mean. RESULTS Children with elevated risk due to high risk SCD and preterm birth performed worse than other groups on a test of expressive language but not on tests that emphasize processing speed and working memory. There was a three-way interaction between preterm status, SCD risk, and abnormal screening outcome, which was largely driven by the increased likelihood of abnormal cognitive scores for children with high risk SCD born preterm. CONCLUSIONS The combination of SCD and preterm birth may confer increased risk for language deficits and elevated rates of abnormal cognitive screenings. This suggests that neurodevelopmental risk imparted by comorbid SCD and preterm birth may manifest as heterogenous, rather than specific, patterns of cognitive deficits. Future studies are needed to clarify the domains of cognitive functioning most susceptible to disease-related effects of comorbid SCD and preterm birth.
Collapse
Affiliation(s)
- Sarah E. Bills
- Department of Psychology, University of South Carolina, Columbia, SC, 29208, USA
| | - Jeffrey Schatz
- Department of Psychology, University of South Carolina, Columbia, SC, 29208, USA
| | - Erin Hunt
- Department of Psychology, University of South Carolina, Columbia, SC, 29208, USA
| | - Sreya Varanasi
- Department of Psychology, University of South Carolina, Columbia, SC, 29208, USA
| | - Julia Johnston
- Department of Psychology, University of South Carolina, Columbia, SC, 29208, USA
| | - Jessica Bradshaw
- Department of Psychology, University of South Carolina, Columbia, SC, 29208, USA
| |
Collapse
|
13
|
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: 7] [Impact Index Per Article: 3.5] [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
|
14
|
Velayudhan PS, Schwab N, Hazrati LN, Wheeler AL. Temporal patterns of microglial activation in white matter following experimental mild traumatic brain injury: a systematic literature review. Acta Neuropathol Commun 2021; 9:197. [PMID: 34924026 PMCID: PMC8684664 DOI: 10.1186/s40478-021-01297-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022] Open
Abstract
Mild traumatic brain injuries (mTBIs) are a prevalent form of injury that can result in persistent neurological impairments. Microglial activation has become increasingly recognized as a key process regulating the pathology of white matter in a wide range of brain injury and disease contexts. As white matter damage is known to be a major contributor to the impairments that follow mTBI, microglia have rightfully become a common target of investigation for the development of mTBI therapies and biomarkers. Recent work has demonstrated that the efficacy of microglial manipulation as a therapeutic intervention following injury or disease is highly time-sensitive, emphasizing the importance of advancing our understanding of the dynamics of post-mTBI microglial activation from onset to resolution. Current reporting of microglial activation in experimental studies of mTBI is non-standardized, which has limited our ability to identify concrete patterns of post-mTBI microglial activation over time. In this review, we examine preclinical studies of mTBI that report on microglial activation in white matter regions to summarize our current understanding of these patterns. Specifically, we summarize timecourses of post-mTBI microglial activation in white matter regions of the brain, identify factors that influence this activation, examine the temporal relationship between microglial activation and other post-mTBI assessments, and compare the relative sensitivities of various methods for detecting microglial activation. While the lack of replicated experimental conditions has limited the extent of conclusions that can confidently be drawn, we find that microglia are activated over a wide range of timecourses following mTBI and that microglial activation is a long-lasting outcome of mTBI that may resolve after most typical post-mTBI assessments, with the exception of those measuring oligodendrocyte lineage cell integrity. We identify several understudied parameters of post-mTBI microglial activation in white matter, such as the inclusion of female subjects. This review summarizes our current understanding of the progression of microglial activation in white matter structures following experimental mTBI and offers suggestions for important future research directions.
Collapse
Affiliation(s)
- Prashanth S Velayudhan
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Nicole Schwab
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Lili-Naz Hazrati
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anne L Wheeler
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
15
|
de la Tremblaye PB, Wellcome JL, Wiley K, Lomahan CA, Moschonas EH, Cheng JP, Bondi CO, Kline AE. Chronic unpredictable stress during adolescence protects against adult traumatic brain injury-induced affective and cognitive deficits. Brain Res 2021; 1767:147544. [PMID: 34090883 PMCID: PMC8349874 DOI: 10.1016/j.brainres.2021.147544] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 01/06/2023]
Abstract
Pre-clinical early-life stress paradigms model early adverse events in humans. However, the long-term behavioral consequences of early-life adversities after traumatic brain injury (TBI) in adults have not been examined. In addition, endocannabinoids may protect against TBI neuropathology. Hence, the current study assessed the effects of adverse stress during adolescence on emotional and cognitive performance in rats sustaining a TBI as adults, and how cannabinoid receptor 1 (CB1) activation impacts the outcome. On postnatal days (PND) 30-60, adolescent male rats were exposed to four weeks of chronic unpredictable stress (CUS), followed by four weeks of no stress (PND 60-90), or no stress at any time (Control), and then anesthetized and provided a cortical impact of moderate severity (2.8 mm tissue deformation at 4 m/s) or sham injury. TBI and Sham rats (CUS and Control) were administered either arachidonyl-2'-chloroethylamide (ACEA; 1 mg/kg, i.p.), a CB1 receptor agonist, or vehicle (VEH; 1 mL/kg, i.p.) immediately after surgery and once daily for 7 days. Anxiety-like behavior was assessed in an open field test (OFT) and learning and memory in novel object recognition (NOR) and Morris water maze (MWM) tasks. No differences were revealed among the Sham groups in any behavioral assessment and thus the groups were pooled. In the ACEA and VEH-treated TBI groups, CUS increased exploration in the OFT, enhanced NOR focus, and decreased the time to reach the escape platform in the MWM, suggesting decreased anxiety and enhanced learning and memory relative to the Control group receiving VEH (p < 0.05). ACEA also enhanced NOR and MWM performance in the Control + TBI group (p < 0.05). These data suggest that 4 weeks of CUS provided during adolescence may provide protection against TBI acquired during adulthood and/or induce adaptive behavioral responses. Moreover, CB1 receptor agonism produces benefits after TBI independent of CUS protection.
Collapse
Affiliation(s)
- Patricia B de la Tremblaye
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States; Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - JoDy L Wellcome
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Kaitlyn Wiley
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Carolyn A Lomahan
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Eleni H Moschonas
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Jeffrey P Cheng
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Corina O Bondi
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States; Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Anthony E Kline
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, United States; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, United States; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, United States; Psychology, University of Pittsburgh, Pittsburgh, PA 15213, United States.
| |
Collapse
|
16
|
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: 8.7] [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
|
17
|
Ndode-Ekane XE, Puigferrat Pérez MDM, Di Sapia R, Lapinlampi N, Pitkänen A. Reorganization of Thalamic Inputs to Lesioned Cortex Following Experimental Traumatic Brain Injury. Int J Mol Sci 2021; 22:6329. [PMID: 34199241 PMCID: PMC8231773 DOI: 10.3390/ijms22126329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/27/2021] [Accepted: 06/10/2021] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) disrupts thalamic and cortical integrity. The effect of post-injury reorganization and plasticity in thalamocortical pathways on the functional outcome remains unclear. We evaluated whether TBI causes structural changes in the thalamocortical axonal projection terminals in the primary somatosensory cortex (S1) that lead to hyperexcitability. TBI was induced in adult male Sprague Dawley rats with lateral fluid-percussion injury. A virus carrying the fluorescent-tagged opsin channel rhodopsin 2 transgene was injected into the ventroposterior thalamus. We then traced the thalamocortical pathways and analyzed the reorganization of their axonal terminals in S1. Next, we optogenetically stimulated the thalamocortical relays from the ventral posterior lateral and medial nuclei to assess the post-TBI functionality of the pathway. Immunohistochemical analysis revealed that TBI did not alter the spatial distribution or lamina-specific targeting of projection terminals in S1. TBI reduced the axon terminal density in the motor cortex by 44% and in S1 by 30%. A nematic tensor-based analysis revealed that in control rats, the axon terminals in layer V were orientated perpendicular to the pial surface (60.3°). In TBI rats their orientation was more parallel to the pial surface (5.43°, difference between the groups p < 0.05). Moreover, the level of anisotropy of the axon terminals was high in controls (0.063) compared with TBI rats (0.045, p < 0.05). Optical stimulation of the sensory thalamus increased alpha activity in electroencephalography by 312% in controls (p > 0.05) and 237% (p > 0.05) in TBI rats compared with the baseline. However, only TBI rats showed increased beta activity (33%) with harmonics at 5 Hz. Our findings indicate that TBI induces reorganization of thalamocortical axonal terminals in the perilesional cortex, which alters responses to thalamic stimulation.
Collapse
Affiliation(s)
- Xavier Ekolle Ndode-Ekane
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (M.d.M.P.P.); (R.D.S.); (N.L.); (A.P.)
| | | | | | | | | |
Collapse
|
18
|
Celorrio M, Abellanas MA, Rhodes J, Goodwin V, Moritz J, Vadivelu S, Wang L, Rodgers R, Xiao S, Anabayan I, Payne C, Perry AM, Baldridge MT, Aymerich MS, Steed A, Friess SH. Gut microbial dysbiosis after traumatic brain injury modulates the immune response and impairs neurogenesis. Acta Neuropathol Commun 2021; 9:40. [PMID: 33691793 PMCID: PMC7944629 DOI: 10.1186/s40478-021-01137-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/26/2021] [Indexed: 12/14/2022] Open
Abstract
The influence of the gut microbiota on traumatic brain injury (TBI) is presently unknown. This knowledge gap is of paramount clinical significance as TBI patients are highly susceptible to alterations in the gut microbiota by antibiotic exposure. Antibiotic-induced gut microbial dysbiosis established prior to TBI significantly worsened neuronal loss and reduced microglia activation in the injured hippocampus with concomitant changes in fear memory response. Importantly, antibiotic exposure for 1 week after TBI reduced cortical infiltration of Ly6Chigh monocytes, increased microglial pro-inflammatory markers, and decreased T lymphocyte infiltration, which persisted through 1 month post-injury. Moreover, microbial dysbiosis was associated with reduced neurogenesis in the dentate gyrus 1 week after TBI. By 3 months after injury (11 weeks after discontinuation of the antibiotics), we observed increased microglial proliferation, increased hippocampal neuronal loss, and modulation of fear memory response. These data demonstrate that antibiotic-induced gut microbial dysbiosis after TBI impacts neuroinflammation, neurogenesis, and fear memory and implicate gut microbial modulation as a potential therapeutic intervention for TBI.
Collapse
Affiliation(s)
- Marta Celorrio
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Miguel A Abellanas
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
- Departamento de Bioquímica Y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain
- CIMA, Programa de Neurociencias, Universidad de Navarra, Pamplona, Spain
| | - James Rhodes
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Victoria Goodwin
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Jennie Moritz
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Sangeetha Vadivelu
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Leran Wang
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Rachel Rodgers
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Sophia Xiao
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Ilakkia Anabayan
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Camryn Payne
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Alexandra M Perry
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Megan T Baldridge
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Maria S Aymerich
- Departamento de Bioquímica Y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain
- CIMA, Programa de Neurociencias, Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Ashley Steed
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Stuart H Friess
- Division of Critical Care Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, USA.
| |
Collapse
|
19
|
Traumatic brain injury in adolescence: A review of the neurobiological and behavioural underpinnings and outcomes. DEVELOPMENTAL REVIEW 2021. [DOI: 10.1016/j.dr.2020.100943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
20
|
Popovitz J, Mysore SP, Adwanikar H. Neural Markers of Vulnerability to Anxiety Outcomes after Traumatic Brain Injury. J Neurotrauma 2020; 38:1006-1022. [PMID: 33050836 DOI: 10.1089/neu.2020.7320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Anxiety outcomes after traumatic brain injury (TBI) are complex, and the underlying neural mechanisms are poorly understood. Here, we developed a multi-dimensional behavioral profiling approach to investigate anxiety-like outcomes in mice that takes into account individual variability. Departing from the tradition of comparing outcomes in TBI versus sham groups, we identified a subgroup within the TBI group that is vulnerable to anxiety dysfunction, and present increased exploration of the anxiogenic zone compared to sham controls or resilient injured animals, by applying dimensionality reduction, clustering, and post hoc validation to behavioral data obtained from multiple assays for anxiety at several post-injury time points. These vulnerable animals expressed distinct molecular profiles in the corticolimbic network, with downregulation in gamma-aminobutyric acid and glutamate and upregulation in neuropeptide Y markers. Indeed, among vulnerable animals, not resilient or sham controls, severity of anxiety-related outcomes correlated strongly with expression of molecular markers. Our results establish a foundational approach, with predictive power, for reliably identifying maladaptive anxiety outcomes after TBI and uncovering neural signatures of vulnerability to anxiety.
Collapse
Affiliation(s)
- Juliana Popovitz
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shreesh P Mysore
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hita Adwanikar
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Shultz SR, McDonald SJ, Corrigan F, Semple BD, Salberg S, Zamani A, Jones NC, Mychasiuk R. Clinical Relevance of Behavior Testing in Animal Models of Traumatic Brain Injury. J Neurotrauma 2020; 37:2381-2400. [DOI: 10.1089/neu.2018.6149] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Frances Corrigan
- Department of Anatomy, University of South Australia, Adelaide, South Australia, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
22
|
Wang Z, Zhang M, Sun C, Wang S, Cao J, Wang KKW, Gan S, Huang W, Niu X, Zhu Y, Sun Y, Bai L. Single Mild Traumatic Brain Injury Deteriorates Progressive Interhemispheric Functional and Structural Connectivity. J Neurotrauma 2020; 38:464-473. [PMID: 30931824 DOI: 10.1089/neu.2018.6196] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The present study examined dynamic interhemispheric structural and functional connectivity in mild traumatic brain injury (mTBI) patients with longitudinal observations from early subacute to chronic stages within 1 year of injury. Forty-two mTBI patients and 42 matched healthy controls underwent clinical and neuropsychological evaluations, diffusion tensor imaging, and resting-state functional magnetic resonance imaging. All mTBI patients were initially evaluated within 14 d post-injury (T-1) and at 3 months (T-2) and 6-12 months (T-3) follow-ups. Separate transcallosal fiber tracts in the corpus callosum (CC) with respect to their specific interhemispheric cortical projections were derived with fiber tracking and voxel-mirrored homotopic connectivity analyses. With diffusion tensor imaging-based tractography, five vertical segments of the CC (I-V) were distinguished. Correlation analyses were performed to evaluate relationships between structural and functional imaging measures as well as imaging indices and neuropsychological measures. The loss of integrity in the CC demonstrated saliently persistent and time-dependent regional specificity after mTBI. The impairment spanned multiple segments from CC II at T-1 and CC I, II, VI, and V at T-2 to all subregions at T-3. Moreover, loss of interhemispheric structural connectivity through the CC corresponded well to regions presenting altered interhemispheric functional connectivity. Decreased functional connectivity in the dorsolateral prefrontal cortex thereafter contributed to poor executive function in mTBI patients. The current study provides further evidence that the CC is a sign to interhemispheric highways underpinning the widespread cerebral pathology typifying mTBI syndrome.
Collapse
Affiliation(s)
- Zhuonan Wang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ming Zhang
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chuanzhu Sun
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shan Wang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jieli Cao
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics, and Biomarker Research, Departments of Emergency Medicine, Psychiatry, and Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Shuoqiu Gan
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenmin Huang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xuan Niu
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanan Zhu
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yingxiang Sun
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lijun Bai
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
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: 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: 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
|
24
|
Pinkowski NJ, Guerin J, Zhang H, Carpentier ST, McCurdy KE, Pacheco JM, Mehos CJ, Brigman JL, Morton RA. Repeated mild traumatic brain injuries impair visual discrimination learning in adolescent mice. Neurobiol Learn Mem 2020; 175:107315. [PMID: 32980477 DOI: 10.1016/j.nlm.2020.107315] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/10/2020] [Accepted: 09/18/2020] [Indexed: 12/19/2022]
Abstract
Cognitive deficits following a mild traumatic brain injury (mTBI) are common and are associated with learning deficits in school-age children. Some of these deficits include problems with long-term memory, working memory, processing speeds, attention, mental fatigue, and executive function. Processing speed deficits have been associated with alterations in white matter, but the underlying mechanisms of many of the other deficits are unclear. Without a clear understanding of the underlying mechanisms we cannot effectively treat these injuries. The goal of these studies is to validate a translatable touchscreen discrimination/reversal task to identify deficits in executive function following a single or repeated mTBIs. Using a mild closed skull injury model in adolescent mice we were able to identify clear deficits in discrimination learning following repeated injuries that were not present from a single mTBI. The repeated injuries were not associated with any deficits in motor-based behavior but did induce a robust increase in astrocyte activation. These studies provide an essential platform to interrogate the underlying neurological dysfunction associated with these injuries.
Collapse
Affiliation(s)
- Natalie J Pinkowski
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, United States
| | - Juliana Guerin
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, United States
| | - Haikun Zhang
- Center for Brain Recovery and Repair, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Sydney T Carpentier
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, United States
| | - Kathryn E McCurdy
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, United States
| | - Johann M Pacheco
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, United States
| | - Carissa J Mehos
- Center for Brain Recovery and Repair, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, United States; Center for Brain Recovery and Repair, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Russell A Morton
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, United States; Center for Brain Recovery and Repair, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
25
|
Gazdzinski LM, Mellerup M, Wang T, Adel SAA, Lerch JP, Sled JG, Nieman BJ, Wheeler AL. White Matter Changes Caused by Mild Traumatic Brain Injury in Mice Evaluated Using Neurite Orientation Dispersion and Density Imaging. J Neurotrauma 2020; 37:1818-1828. [PMID: 32242488 DOI: 10.1089/neu.2020.6992] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is common and can lead to persistent cognitive and behavioral symptoms. Although diffusion tensor imaging (DTI) has demonstrated some sensitivity to changes in white matter following mTBI, recent studies have suggested that more complex geometric models of diffusion, including the neurite orientation dispersion and density imaging (NODDI) model, may be more sensitive and specific. Here, we evaluate microstructural changes in white matter following mTBI using DTI and NODDI in a mouse model, and compare the time course of these changes to behavioral impairment and recovery. We also assess volumetric changes for a comprehensive picture of the structural alterations in the brain and histological staining to identify cellular changes that may contribute to the differences detected in the imaging data. Increased orientation dispersion index (ODI) was observed in the optic tracts of mTBI mice compared with shams. Changes in fractional anisotropy (FA) were not statistically significant. Volume deficits were detected in the optic tract as well as in several gray matter regions: the lateral geniculate nuclei of the thalamus, the entorhinal cortex, and the superior colliculi. Glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) staining was increased in the optic tracts of mTBI brains, and this staining correlated with ODI values. A transient impairment in working memory was observed, which resolved by 6 weeks, whereas increased ODI, GFAP, and Iba1 persisted to 18 weeks post-injury. We conclude that the optic tracts are particularly vulnerable to damage from the closed-skull impact model used in this study, and that ODI may be a more sensitive metric to this damage than FA. Differences in ODI and in histological measures of astrogliosis, neuroinflammation, and axonal degeneration persist beyond behavioral impairment in this model.
Collapse
Affiliation(s)
- Lisa M Gazdzinski
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Miranda Mellerup
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and University of Toronto, Toronto, Ontario, Canada
| | - Tong Wang
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and University of Toronto, Toronto, Ontario, Canada
| | - Seyed Amir Ali Adel
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and University of Toronto, Toronto, Ontario, Canada
| | - Jason P Lerch
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Wellcome Centre for Integrative Neuroimaging, Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - John G Sled
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada.,Mouse Imaging Centre at The Centre for Phenogenomics, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Brian J Nieman
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada.,Mouse Imaging Centre at The Centre for Phenogenomics, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Anne L Wheeler
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
History of Concussion Diagnosis, Differences in Concussion Reporting Behavior, and Self-Described Reasons for Non-Report. JOURNAL OF CLINICAL SPORT PSYCHOLOGY 2020. [DOI: 10.1123/jcsp.2017-0036] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Purpose: Assess whether athletes with a prior concussion diagnosis are more likely to continue play with a possible concussion. Additionally, explore whether reasons for concussion under-reporting are different among athletes with a prior concussion when compared to other athletes. Methods: Cross-sectional survey of 328 collegiate athletes. Results: Athletes with a prior concussion diagnosis had significantly greater relative risk of continuing play while symptomatic of a possible concussion during their most recent season compared to athletes without prior concussion diagnosis. Significant differences exist in the reasons that athletes provided for not reporting by history of concussion. Conclusions: Findings suggest that learning may have occurred as a result of the prior diagnosis; however, this learning did not appear to result in safer reporting behavior. Additional research is necessary to clarify why athletes who have been previously diagnosed with a concussion are more likely to continue playing while experiencing concussion symptoms.
Collapse
|
27
|
Eyolfson E, Yamakawa GR, Griep Y, Collins R, Carr T, Wang M, Lohman AW, Mychasiuk R. Examining the Progressive Behavior and Neuropathological Outcomes Associated with Chronic Repetitive Mild Traumatic Brain Injury in Rats. Cereb Cortex Commun 2020; 1:tgaa002. [PMID: 34296084 PMCID: PMC8152839 DOI: 10.1093/texcom/tgaa002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/24/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
While the physical and behavioral symptomologies associated with a single mild traumatic brain injury (mTBI) are typically transient, repetitive mTBIs (RmTBI) have been associated with persisting neurological deficits. Therefore, this study examined the progressive changes in behavior and the neuropathological outcomes associated with chronic RmTBI through adolescence and adulthood in male and female Sprague Dawley rats. Rats experienced 2 mTBIs/week for 15 weeks and were periodically tested for changes in motor behavior, cognitive function, emotional disturbances, and aggression. Brain tissue was examined for neuropathological changes in ventricle size and presentation of Iba1 and GFAP. We did not see progressively worse behavioral impairments with the accumulation of injuries or time, but did find evidence for neurological and functional change (motor disturbance, reduced exploration, reduced aggression, alteration in depressive-like behavior, deficits in short-term working memory). Neuropathological assessment of RmTBI animals identified an increase in ventricle size, prolonged changes in GFAP, and sex differences in Iba1, in the corpus callosum, thalamus, and medial prefrontal cortex. Telomere length reduced exponentially as the injury load increased. Overall, chronic RmTBI did not result in accumulating behavioral impairment, and there is a need to further investigate progressive behavioral changes associated with repeated injuries in adolescence and young adulthood.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Yannick Griep
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Division of Epidemiology, Stress Research Institute, Stockholm University, 106 91 Stockholm, Sweden
- Behavioral Science Institute, Radbound University, 9104, 6500 HE, Nijmegen, The Netherlands
| | - Reid Collins
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Thomas Carr
- Department of Cell Biology and Anatomy, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Melinda Wang
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Alexander W Lohman
- Department of Cell Biology and Anatomy, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Richelle Mychasiuk
- Department of Psychology, Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Psychology, Hotchkiss Brain Institute, The University of Calgary, Calgary, AB, T2N 1N4, Canada
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| |
Collapse
|
28
|
Phenotyping Cardiac Arrest: Bench and Bedside Characterization of Brain and Heart Injury Based on Etiology. Crit Care Med 2019. [PMID: 29533310 DOI: 10.1097/ccm.0000000000003070] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Cardiac arrest etiology may be an important source of between-patient heterogeneity, but the impact of etiology on organ injury is unknown. We tested the hypothesis that asphyxial cardiac arrest results in greater neurologic injury than cardiac etiology cardiac arrest (ventricular fibrillation cardiac arrest), whereas ventricular fibrillation cardiac arrest results in greater cardiovascular dysfunction after return of spontaneous circulation. DESIGN Prospective observational human and randomized animal study. SETTING University laboratory and ICUs. PATIENTS Five-hundred forty-three cardiac arrest patients admitted to ICU. SUBJECTS Seventy-five male Sprague-Dawley rats. INTERVENTIONS We examined neurologic and cardiovascular injury in Isoflurane-anesthetized rat cardiac arrest models matched by ischemic time. Hemodynamic and neurologic outcomes were assessed after 5 minutes no flow asphyxial cardiac arrest or ventricular fibrillation cardiac arrest. Comparison was made to injury patterns observed after human asphyxial cardiac arrest or ventricular fibrillation cardiac arrest. MEASUREMENTS AND MAIN RESULTS In rats, cardiac output (20 ± 10 vs 45 ± 9 mL/min) and pH were lower and lactate higher (9.5 ± 1.0 vs 6.4 ± 1.3 mmol/L) after return of spontaneous circulation from ventricular fibrillation cardiac arrest versus asphyxial cardiac arrest (all p < 0.01). Asphyxial cardiac arrest resulted in greater early neurologic deficits, 7-day neuronal loss, and reduced freezing time (memory) after conditioned fear (all p < 0.05). Brain antioxidant reserves were more depleted following asphyxial cardiac arrest. In adjusted analyses, human ventricular fibrillation cardiac arrest was associated with greater cardiovascular injury based on peak troponin (7.8 ng/mL [0.8-57 ng/mL] vs 0.3 ng/mL [0.0-1.5 ng/mL]) and ejection fraction by echocardiography (20% vs 55%; all p < 0.0001), whereas asphyxial cardiac arrest was associated with worse early neurologic injury and poor functional outcome at hospital discharge (n = 46 [18%] vs 102 [44%]; p < 0.0001). Most ventricular fibrillation cardiac arrest deaths (54%) were the result of cardiovascular instability, whereas most asphyxial cardiac arrest deaths (75%) resulted from neurologic injury (p < 0.0001). CONCLUSIONS In transcending rat and human studies, we find a consistent phenotype of heart and brain injury after cardiac arrest based on etiology: ventricular fibrillation cardiac arrest produces worse cardiovascular dysfunction, whereas asphyxial cardiac arrest produces worsened neurologic injury associated with greater oxidative stress.
Collapse
|
29
|
Weber B, Lackner I, Haffner-Luntzer M, Palmer A, Pressmar J, Scharffetter-Kochanek K, Knöll B, Schrezenemeier H, Relja B, Kalbitz M. Modeling trauma in rats: similarities to humans and potential pitfalls to consider. J Transl Med 2019; 17:305. [PMID: 31488164 PMCID: PMC6728963 DOI: 10.1186/s12967-019-2052-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/29/2019] [Indexed: 12/27/2022] Open
Abstract
Trauma is the leading cause of mortality in humans below the age of 40. Patients injured by accidents frequently suffer severe multiple trauma, which is life-threatening and leads to death in many cases. In multiply injured patients, thoracic trauma constitutes the third most common cause of mortality after abdominal injury and head trauma. Furthermore, 40-50% of all trauma-related deaths within the first 48 h after hospital admission result from uncontrolled hemorrhage. Physical trauma and hemorrhage are frequently associated with complex pathophysiological and immunological responses. To develop a greater understanding of the mechanisms of single and/or multiple trauma, reliable and reproducible animal models, fulfilling the ethical 3 R's criteria (Replacement, Reduction and Refinement), established by Russell and Burch in 'The Principles of Human Experimental Technique' (published 1959), are required. These should reflect both the complex pathophysiological and the immunological alterations induced by trauma, with the objective to translate the findings to the human situation, providing new clinical treatment approaches for patients affected by severe trauma. Small animal models are the most frequently used in trauma research. Rattus norvegicus was the first mammalian species domesticated for scientific research, dating back to 1830. To date, there exist numerous well-established procedures to mimic different forms of injury patterns in rats, animals that are uncomplicated in handling and housing. Nevertheless, there are some physiological and genetic differences between humans and rats, which should be carefully considered when rats are chosen as a model organism. The aim of this review is to illustrate the advantages as well as the disadvantages of rat models, which should be considered in trauma research when selecting an appropriate in vivo model. Being the most common and important models in trauma research, this review focuses on hemorrhagic shock, blunt chest trauma, bone fracture, skin and soft-tissue trauma, burns, traumatic brain injury and polytrauma.
Collapse
Affiliation(s)
- Birte Weber
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Ina Lackner
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma-Immunology, University of Ulm, Ulm, Germany
| | - Jochen Pressmar
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | | | - Bernd Knöll
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Hubert Schrezenemeier
- Institute of Transfusion Medicine, University of Ulm and Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, Frankfurt, Germany
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| |
Collapse
|
30
|
Kulkarni P, Morrison TR, Cai X, Iriah S, Simon N, Sabrick J, Neuroth L, Ferris CF. Neuroradiological Changes Following Single or Repetitive Mild TBI. Front Syst Neurosci 2019; 13:34. [PMID: 31427931 PMCID: PMC6688741 DOI: 10.3389/fnsys.2019.00034] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 07/10/2019] [Indexed: 11/13/2022] Open
Abstract
Objectives To test the hypothesis that there are differences in neuroradiological measures between single and repeated mild traumatic brain injury using multimodal MRI. Methods A closed-head momentum exchange model was used to produce one or three mild head injuries in young adult male rats compared to non-injured, age and weight-matched controls. Six-seven weeks post-injury, rats were studied for deficits in cognitive and motor function. Seven-eight weeks post-injury changes in brain anatomy and function were evaluated through analysis of high resolution T2 weighted images, resting-state BOLD functional connectivity, and diffusion weighted imaging with quantitative anisotropy. Results Head injuries occurred without skull fracture or signs of intracranial bleeding or contusion. There were no significant differences in cognitive or motors behaviors between experimental groups. With a single mild hit, the affected areas were limited to the caudate/putamen and central amygdala. Rats hit three times showed altered diffusivity in white matter tracts, basal ganglia, central amygdala, brainstem, and cerebellum. Comparing three hits to one hit showed a similar pattern of change underscoring a dose effect of repeated head injury on the brainstem and cerebellum. Disruption of functional connectivity was pronounced with three mild hits. The midbrain dopamine system, hippocampus, and brainstem/cerebellum showed hypoconnectivity. Interestingly, rats exposed to one hit showed enhanced functional connectivity (or hyperconnectivity) across brain sites, particularly between the olfactory system and the cerebellum. Interpretation Neuroradiological evidence of altered brain structure and function, particularly in striatal and midbrain dopaminergic areas, persists long after mild repetitive head injury. These changes may serve as biomarkers of neurodegeneration and risk for dementia later in life.
Collapse
Affiliation(s)
- Praveen Kulkarni
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Thomas R Morrison
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Xuezhu Cai
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Sade Iriah
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Neal Simon
- Azevan Pharmaceuticals, Bethlehem, PA, United States.,Department of Biological Sciences, College of Arts and Sciences, Lehigh University, Bethlehem, PA, United States
| | - Julia Sabrick
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Lucas Neuroth
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Craig F Ferris
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| |
Collapse
|
31
|
Curry AE, Arbogast KB, Metzger KB, Kessler RS, Breiding MJ, Haarbauer-Krupa J, DePadilla L, Greenspan A, Master CL. Risk of Repeat Concussion Among Patients Diagnosed at a Pediatric Care Network. J Pediatr 2019; 210:13-19.e2. [PMID: 31101406 PMCID: PMC6645379 DOI: 10.1016/j.jpeds.2019.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To quantify the risk of repeat concussions for children and identify demographic and clinical aspects of the index concussion associated with repeat injury. STUDY DESIGN For this retrospective cohort study, we queried the Children's Hospital of Philadelphia healthcare network's unified electronic health record to identify all 5- to 15-year-old patients who had their first clinical visit for an index concussion at a Children's Hospital of Philadelphia location from July 2012 through June 2013. A 25% random sample (n = 536) were selected. Clinical data were abstracted for their index concussion and all concussion-related visits for 2 years following the index concussion. RESULTS Overall, 16.2% (n = 87) of patients experienced at least 1 repeat concussion within 2 years of their index concussion. The risk of repeat concussion increased with patient age (9.5% for ages 5-8 years; 10.7% for ages 9-11 years; and 19.8% for ages 12-15 years). After we adjusted for other factors, risk was particularly heightened among patients whose index concussion had a longer clinical course (>30 vs 0-7 days, adjusted risk ratio 1.65 [1.01-2.69]) and greater symptom burden (>11 vs 0-2 symptoms, adjusted risk ratio 2.12 [1.12-3.72]). CONCLUSIONS We estimate that 1 in 6 youth diagnosed with a concussion are diagnosed with a subsequent concussion within 2 years and that several clinical characteristics of the index concussion increase this risk. Identifying factors associated with a repeat injury is essential to inform the clinical management of concussion and direct injury prevention efforts.
Collapse
Affiliation(s)
- Allison E Curry
- Center for Injury Research and Prevention, Children's Hospital of Philadelphia, Philadelphia, PA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
| | - Kristy B Arbogast
- Center for Injury Research and Prevention, Children's Hospital of Philadelphia, Philadelphia, PA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kristina B Metzger
- Center for Injury Research and Prevention, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Ronni S Kessler
- Center for Injury Research and Prevention, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Matthew J Breiding
- National Center for Injury Prevention and Control, Centers for Disease Control and Prevention, Atlanta, GA
| | - Juliet Haarbauer-Krupa
- National Center for Injury Prevention and Control, Centers for Disease Control and Prevention, Atlanta, GA
| | - Lara DePadilla
- National Center for Injury Prevention and Control, Centers for Disease Control and Prevention, Atlanta, GA
| | - Arlene Greenspan
- National Center for Injury Prevention and Control, Centers for Disease Control and Prevention, Atlanta, GA
| | - Christina L Master
- Center for Injury Research and Prevention, Children's Hospital of Philadelphia, Philadelphia, PA; Sports Medicine and Performance Center, Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
32
|
Kinder HA, Baker EW, Howerth EW, Duberstein KJ, West FD. Controlled Cortical Impact Leads to Cognitive and Motor Function Deficits that Correspond to Cellular Pathology in a Piglet Traumatic Brain Injury Model. J Neurotrauma 2019; 36:2810-2826. [PMID: 31084390 DOI: 10.1089/neu.2019.6405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the United States, with children who sustain a TBI having a greater risk of developing long-lasting cognitive, behavioral, and motor function deficits. This has led to increased interest in utilizing large animal models to study pathophysiologic and functional changes after injury in hopes of identifying novel therapeutic targets. In the present study, a controlled cortical impact (CCI) piglet TBI model was utilized to evaluate cognitive, motor, and histopathologic outcomes. CCI injury (4 m/sec velocity, 9 mm depression, 400 msec dwell time) was induced at the parietal cortex. Compared with normal pigs (n = 5), TBI pigs (n = 5) exhibited appreciable cognitive deficiencies, including significantly impaired spatial memory in spatial T-maze testing and a significant decrease in exploratory behaviors followed by marked hyperactivity in open field testing. Additionally, gait analysis revealed significant increases in cycle time and stance percent, significant decreases in hind reach, and a shift in the total pressure index from the front to the hind limb on the affected side, suggesting TBI impairs gait and balance. Pigs were sacrificed 28 days post-TBI and histological analysis revealed that TBI lead to a significant decrease in neurons and a significant increase in microglia activation and astrogliosis/astrocytosis at the perilesional area, a significant loss in neurons at the dorsal hippocampus, and significantly increased neuroblast proliferation at the subventricular zone. These data demonstrate a strong relationship between TBI-induced cellular changes and functional outcomes in our piglet TBI model that lay the framework for future studies that assess the ability of therapeutic interventions to contribute to functional improvements.
Collapse
Affiliation(s)
- Holly A Kinder
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Emily W Baker
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Elizabeth W Howerth
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Pathology, University of Georgia, Athens, Georgia
| | - Kylee J Duberstein
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| | - Franklin D West
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia.,Department of Animal and Dairy Science, University of Georgia, Athens, Georgia
| |
Collapse
|
33
|
Abstract
The underlying mechanisms that result in neurophysiological changes and cognitive sequelae in the context of repetitive mild traumatic brain injury (rmTBI) remain poorly understood. Animal models provide a unique opportunity to examine cellular and molecular responses using histological assessment, which can give important insights on the neurophysiological changes associated with the evolution of brain injury. To better understand the potential cumulative effects of multiple concussions, the focus of animal models is shifting from single to repetitive head impacts. With a growing body of literature on this subject, a review and discussion of current findings is valuable to better understand the neuropathology associated with rmTBI, to evaluate the current state of the field, and to guide future research efforts. Despite variability in experimental settings, existing animal models of rmTBI have contributed to our understanding of the underlying mechanisms following repeat concussion. However, how to reconcile the various impact methods remains one of the major challenges in the field today.
Collapse
Affiliation(s)
- Wouter S Hoogenboom
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Clinical Investigation, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA.
| | - Craig A Branch
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Physiology and Biophysics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| | - Michael L Lipton
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Departments of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| |
Collapse
|
34
|
Maynard ME, Underwood EL, Redell JB, Zhao J, Kobori N, Hood KN, Moore AN, Dash PK. Carnosic Acid Improves Outcome after Repetitive Mild Traumatic Brain Injury. J Neurotrauma 2019; 36:2147-2152. [PMID: 30672378 DOI: 10.1089/neu.2018.6155] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In the majority of cases, the cognitive and behavioral impairments resulting from a mild traumatic brain injury (TBI) (also referred to as concussion) wane within days to weeks. In contrast, these impairments can persist for months to years after repetitive mild TBI (rmTBI). The cellular and molecular mechanisms underlying these impairments are not well understood. In the present study, we examined the consequences of rmTBI (three weight drops each separated by 72 h) on brain tissue respiration, pathology, and cognitive performance in mice. The transcription factor nuclear factor-erythroid 2-realted factor 2 (Nrf2) has been demonstrated to enhance the expression of numerous cytoprotective genes. Carnosic acid (CA) has been shown to activate Nrf2 and suppress the proinflammatory transcription factor nuclear factor kappa B (NF-κB). Because contemporaneous activation of cytoprotective genes and inhibition of proinflammatory genes can be beneficial, we questioned whether CA can be used to mitigate the pathobiology of rmTBI. The rmTBI increased hippocampal adenosine triphosphate-linked tissue respiration and proton leak that were unaffected by CA treatment. The rmTBI also caused significant motor and cognitive dysfunction, as tested using the foot fault, Barnes maze, and novel object recognition tasks. These impairments occurred in the absence of visible neuronal or dendritic loss. Post-rmTBI administration of CA significantly improved motor and cognitive function, and decreased Gfap and Iba1 immunoreactivities within white matter tracks. Taken together, these results show that rmTBI can cause cognitive impairments in the absence of overt neuronal pathologies, and post-injury treatment with CA can lessen some of these impairments.
Collapse
Affiliation(s)
- Mark E Maynard
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Erica L Underwood
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - John B Redell
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Jing Zhao
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Nobuhide Kobori
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Kimberly N Hood
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, the University of Texas McGovern Medical School, Houston, Texas
| |
Collapse
|
35
|
Smith AC, Holden RC, Rasmussen SM, Hoane MR, Hylin MJ. Effects of nicotinamide on spatial memory and inflammation after juvenile traumatic brain injury. Behav Brain Res 2019; 364:123-132. [PMID: 30771366 DOI: 10.1016/j.bbr.2019.02.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 01/23/2023]
Abstract
Age is a consistent predictor of outcome following traumatic brain injury (TBI). Although children and adolescents have the highest rate of hospitalizations and long-term disabilities, few preclinical studies have attempted to model and treat TBI in this population. Studies using nicotinamide (NAM), a soluble B-group vitamin, in older animals (3-6 months) have shown improved functional recovery in experimental models of TBI. The purpose of this study was two-fold: to examine the preclinical efficacy of NAM at different doses on behavioral outcomes in juvenile rats and examine the microglial response over time. Groups of juvenile rats (PND 28-60) were assigned to sham, NAM (125 mg/kg, 500 mg/kg, or 1000 mg/kg) or saline (1 mL/kg) and received unilateral cortical contusion injuries (CCI) and received injections at 15 min, 24 h, and 72 h after injury. Animals treated with NAM demonstrated no significant behavioral improvements over saline treatments. NAM treatments did however show slowed cortical loss and reduced microglia compared to saline treated animals. In summary, the preclinical efficacy of NAM as a treatment following CCI in juvenile animals differs from that previously documented in older rat models. While NAM treatments did reduce microglial activity and slowed progression of cortical loss, it did not reduce the total cortical volume lost nor did it improve behavioral outcomes. The findings of this study emphasize the need to examine potential treatments for TBI utilizing juvenile populations and may explain why so many treatments have failed in clinical trials.
Collapse
Affiliation(s)
- Aidan C Smith
- Neurotrauma and Rehabilitation Laboratory, Southern Illinois University, Carbondale, IL, United States
| | - Ryan C Holden
- Neurotrauma and Rehabilitation Laboratory, Southern Illinois University, Carbondale, IL, United States
| | - Sherry M Rasmussen
- Neurotrauma and Rehabilitation Laboratory, Southern Illinois University, Carbondale, IL, United States
| | - Michael R Hoane
- Restorative Neuroscience Laboratory, Southern Illinois University, Carbondale, IL, United States
| | - Michael J Hylin
- Neurotrauma and Rehabilitation Laboratory, Southern Illinois University, Carbondale, IL, United States.
| |
Collapse
|
36
|
Walker A, Kim J, Wyatt J, Terlouw A, Balachandran K, Wolchok J. Repeated In Vitro Impact Conditioning of Astrocytes Decreases the Expression and Accumulation of Extracellular Matrix. Ann Biomed Eng 2019; 47:967-979. [PMID: 30706307 DOI: 10.1007/s10439-019-02219-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/24/2019] [Indexed: 12/19/2022]
Abstract
Pathological changes to the physical and chemical properties of brain extracellular matrix (ECM) occur following injury. It is generally assumed that astrocytes play an important role in these changes. What remain unclear are the triggers that lead to changes in the regulation of ECM by astrocytes following injury. We hypothesize that mechanical stimulation triggers genotypic and phenotypic changes to astrocytes that could ultimately reshape the ECM composition of the central nervous system following injury. To explore astrocyte mechanobiology, an in vitro drop test bioreactor was employed to condition primary rat astrocytes using short duration (10 ms), high deceleration (150G) and strain (20%) impact stimuli. Experiments were designed to explore the effect of single and repeated impact (single vs. double) on mechano-sensitive behavior including cell viability; ECM gene (collagens I and IV, fibronectin, neurocan, versican) and reactivity gene [glial fibrillary acidic protein (GFAP), S100B, vimentin] expression; matrix regulatory cytokine secretion [matrix metalloproteinase 2 (MMP-2), tissue inhibitor of metalloproteinases 1 (TIMP1), transforming growth factor beta 1 (TGFβ1)]; and matrix accumulation [collagen and glycosaminoglycan (GAG)]. Experiments revealed that both ECM and reactive gliosis gene expression was significantly decreased in response to impact conditioning. The decreases for several genes, including collagen, versican, and GFAP were sensitive to impact number, suggesting mechano-sensitivity to repeated impact conditioning. The measured decreases in ECM gene expression were supported by longer-term in vitro experiments that demonstrated significant decreases in chondroitin sulfate proteoglycan (CSPG) and collagen accumulation within impact conditioned 3-D scaffolds accompanied by a 25% decrease in elastic modulus. Overall, the general trend across all samples was towards altered ECM and reactive gliosis gene expression in response to impact. These results suggest that the regulation of ECM production by astrocytes is sensitive to mechanical stimuli, and that repeated impact conditioning may increase this sensitivity.
Collapse
Affiliation(s)
- Addison Walker
- Department of Biomedical Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA
| | - Johntaehwan Kim
- Department of Biomedical Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA
| | - Joseph Wyatt
- Department of Biomedical Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA
| | - Abby Terlouw
- Department of Biomedical Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA
| | - Jeffrey Wolchok
- Department of Biomedical Engineering, University of Arkansas, 125 Engineering Hall, Fayetteville, AR, 72701, USA.
| |
Collapse
|
37
|
Proton Magnetic Resonance Spectroscopy (H1-MRS) Study of the Ketogenic Diet on Repetitive Mild Traumatic Brain Injury in Adolescent Rats and Its Effect on Neurodegeneration. World Neurosurg 2018; 120:e1193-e1202. [DOI: 10.1016/j.wneu.2018.09.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 11/21/2022]
|
38
|
Lee JB, Affeldt BM, Gamboa Y, Hamer M, Dunn JF, Pardo AC, Obenaus A. Repeated Pediatric Concussions Evoke Long-Term Oligodendrocyte and White Matter Microstructural Dysregulation Distant from the Injury. Dev Neurosci 2018; 40:358-375. [PMID: 30466074 DOI: 10.1159/000494134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/30/2018] [Indexed: 11/19/2022] Open
Abstract
Concussion or mild traumatic brain injury (mTBI) is often accompanied by long-term behavioral and neuropsychological deficits. Emerging data suggest that these deficits can be exacerbated following repeated injuries. However, despite the overwhelming prevalence of mTBI in children due to falls and sports-related activities, the effects of mTBI on white matter (WM) structure and its development in children have not been extensively examined. Moreover, the effect of repeated mTBI (rmTBI) on developing WM has not yet been studied, despite the possibility of exacerbated outcomes with repeat injuries. To address this knowledge gap, we investigated the long-term effects of single (s)mTBI and rmTBI on the WM in the pediatric brain, focusing on the anterior commissure (AC), a WM structure distant to the injury site, using diffusion tensor imaging (DTI) and immunohistochemistry (IHC). We hypothesized that smTBI and rmTBI to the developing mouse brain would lead to abnormalities in microstructural integrity and impaired oligodendrocyte (OL) development. We used a postnatal day 14 Ascl1-CreER: ccGFP mouse closed head injury (CHI) model with a bilateral repeated injury. We demonstrate that smTBI and rmTBI differentially lead to myelin-related diffusion changes in the WM and to abnormal OL development in the AC, which are accompanied by behavioral deficits 2 months after the initial injury. Our results suggest that mTBIs elicit long-term behavioral alterations and OL-associated WM dysregulation in the developing brain. These findings warrant additional research into the development of WM and OL as key components of pediatric TBI pathology and potential therapeutic targets.
Collapse
Affiliation(s)
- Jeong Bin Lee
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Bethann M Affeldt
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Yaritxa Gamboa
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Mary Hamer
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Jeff F Dunn
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrea C Pardo
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andre Obenaus
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA, .,Department of Pediatrics, University of California Irvine School of Medicine, Irvine, California, USA,
| |
Collapse
|
39
|
Semple BD, Zamani A, Rayner G, Shultz SR, Jones NC. Affective, neurocognitive and psychosocial disorders associated with traumatic brain injury and post-traumatic epilepsy. Neurobiol Dis 2018; 123:27-41. [PMID: 30059725 DOI: 10.1016/j.nbd.2018.07.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/08/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
Survivors of traumatic brain injury (TBI) often develop chronic neurological, neurocognitive, psychological, and psychosocial deficits that can have a profound impact on an individual's wellbeing and quality of life. TBI is also a common cause of acquired epilepsy, which is itself associated with significant behavioral morbidity. This review considers the clinical and preclinical evidence that post-traumatic epilepsy (PTE) acts as a 'second-hit' insult to worsen chronic behavioral outcomes for brain-injured patients, across the domains of emotional, cognitive, and psychosocial functioning. Surprisingly, few well-designed studies have specifically examined the relationship between seizures and behavioral outcomes after TBI. The complex mechanisms underlying these comorbidities remain incompletely understood, although many of the biological processes that precipitate seizure occurrence and epileptogenesis may also contribute to the development of chronic behavioral deficits. Further, the relationship between PTE and behavioral dysfunction is increasingly recognized to be a bidirectional one, whereby premorbid conditions are a risk factor for PTE. Clinical studies in this arena are often challenged by the confounding effects of anti-seizure medications, while preclinical studies have rarely examined an adequately extended time course to fully capture the time course of epilepsy development after a TBI. To drive the field forward towards improved treatment strategies, it is imperative that both seizures and neurobehavioral outcomes are assessed in parallel after TBI, both in patient populations and preclinical models.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Akram Zamani
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia.
| | - Genevieve Rayner
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre (Austin Campus), Heidelberg, VIC, Australia; Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, VIC, Australia; Comprehensive Epilepsy Program, Alfred Health, Australia.
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| | - Nigel C Jones
- Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, VIC, Australia.
| |
Collapse
|
40
|
Aldag M, Armstrong RC, Bandak F, Bellgowan PSF, Bentley T, Biggerstaff S, Caravelli K, Cmarik J, Crowder A, DeGraba TJ, Dittmer TA, Ellenbogen RG, Greene C, Gupta RK, Hicks R, Hoffman S, Latta RC, Leggieri MJ, Marion D, Mazzoli R, McCrea M, O'Donnell J, Packer M, Petro JB, Rasmussen TE, Sammons-Jackson W, Shoge R, Tepe V, Tremaine LA, Zheng J. The Biological Basis of Chronic Traumatic Encephalopathy following Blast Injury: A Literature Review. J Neurotrauma 2018; 34:S26-S43. [PMID: 28937953 DOI: 10.1089/neu.2017.5218] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The United States Department of Defense Blast Injury Research Program Coordinating Office organized the 2015 International State-of-the-Science meeting to explore links between blast-related head injury and the development of chronic traumatic encephalopathy (CTE). Before the meeting, the planning committee examined articles published between 2005 and October 2015 and prepared this literature review, which summarized broadly CTE research and addressed questions about the pathophysiological basis of CTE and its relationship to blast- and nonblast-related head injury. It served to inform participants objectively and help focus meeting discussion on identifying knowledge gaps and priority research areas. CTE is described generally as a progressive neurodegenerative disorder affecting persons exposed to head injury. Affected individuals have been participants primarily in contact sports and military personnel, some of whom were exposed to blast. The symptomatology of CTE overlaps with Alzheimer's disease and includes neurological and cognitive deficits, psychiatric and behavioral problems, and dementia. There are no validated diagnostic criteria, and neuropathological evidence of CTE has come exclusively from autopsy examination of subjects with histories of exposure to head injury. The perivascular accumulation of hyperphosphorylated tau (p-tau) at the depths of cortical sulci is thought to be unique to CTE and has been proposed as a diagnostic requirement, although the contribution of p-tau and other reported pathologies to the development of clinical symptoms of CTE are unknown. The literature on CTE is limited and is focused predominantly on head injuries unrelated to blast exposure (e.g., football players and boxers). In addition, comparative analyses of clinical case reports has been challenging because of small case numbers, selection biases, methodological differences, and lack of matched controls, particularly for blast-exposed individuals. Consequently, the existing literature is not sufficient to determine whether the development of CTE is associated with head injury frequency (e.g., single vs. multiple exposures) or head injury type (e.g., impact, nonimpact, blast-related). Moreover, the incidence and prevalence of CTE in at-risk populations is unknown. Future research priorities should include identifying additional risk factors, pursuing population-based longitudinal studies, and developing the ability to detect and diagnose CTE in living persons using validated criteria.
Collapse
Affiliation(s)
- Matt Aldag
- 1 Booz Allen Hamilton , McLean, Virginia
| | - Regina C Armstrong
- 2 Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Faris Bandak
- 3 Defense Advanced Research Projects Agency , Arlington, Virginia
| | | | | | - Sean Biggerstaff
- 6 Office of the Assistant Secretary of Defense , Health Affairs, Falls Church, Virginia
| | | | - Joan Cmarik
- 7 Office of the Principal Assistant for Acquisition, United States Army Medical Research and Materiel Command , Frederick, Maryland
| | - Alicia Crowder
- 8 Combat Casualty Care Research Program , United States Army Medical Research and Materiel Command, Fort Detrick, Maryland
| | | | | | - Richard G Ellenbogen
- 10 Departments of Neurological Surgery and Global Health Medicine, University of Washington , Seattle, Washington
| | - Colin Greene
- 11 Joint Trauma Analysis and Prevention of Injuries in Combat Program, Frederick, Maryland
| | - Raj K Gupta
- 12 Department of Defense Blast Injury Research Program Coordinating Office, United States Army Medical Research and Materiel Command , Frederick, Maryland
| | | | | | | | - Michael J Leggieri
- 12 Department of Defense Blast Injury Research Program Coordinating Office, United States Army Medical Research and Materiel Command , Frederick, Maryland
| | - Donald Marion
- 16 Defense and Veterans Brain Injury Center , Silver Spring, Maryland
| | | | | | | | - Mark Packer
- 20 Hearing Center of Excellence , Lackland, Texas
| | - James B Petro
- 21 Office of the Assistant Secretary of Defense, Research and Engineering, Arlington, Virginia
| | - Todd E Rasmussen
- 8 Combat Casualty Care Research Program , United States Army Medical Research and Materiel Command, Fort Detrick, Maryland
| | - Wendy Sammons-Jackson
- 22 Office of the Principal Assistant for Research and Technology , United States Army Medical Research and Materiel Command, Fort Detrick, Maryland
| | - Richard Shoge
- 23 Military Operational Medicine Research Program, United States Army Medical Research and Materiel Command , Fort Detrick, Maryland
| | | | | | - James Zheng
- 25 Program Executive Office Soldier , Fort Belvoir, Virginia
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW This article summarizes the impact and complications of mild traumatic brain injury and concussion in children and outlines the recent evidence for its assessment and early management. Useful evidence-based management strategies are provided for children who have a typical recovery following concussion as well as for those who have persistent postconcussion syndrome. Cases are used to demonstrate the commonly encountered pathologies of headache, cognitive issues, and mood disturbances following injury. RECENT FINDINGS A clinical risk score using risk factors for poor recovery (eg, female sex, adolescence, previous migraine, and a high degree of acute symptoms) can be used to help the clinician plan follow-up in the community. Prolonged periods of physical and cognitive rest should be avoided. Multidisciplinary treatment plans are often required in the management of persistent postconcussion syndrome. SUMMARY A paucity of research exists for the treatment of postconcussion syndrome. Current treatments target individual symptoms.
Collapse
|
42
|
Fidan E, Foley LM, New LA, Alexander H, Kochanek PM, Hitchens TK, Bayır H. Metabolic and Structural Imaging at 7 Tesla After Repetitive Mild Traumatic Brain Injury in Immature Rats. ASN Neuro 2018; 10:1759091418770543. [PMID: 29741097 PMCID: PMC5944144 DOI: 10.1177/1759091418770543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 03/03/2018] [Accepted: 03/20/2018] [Indexed: 11/15/2022] Open
Abstract
Mild traumatic brain injury (mTBI) in children is a common and serious public health problem. Traditional neuroimaging findings in children who sustain mTBI are often normal, putting them at risk for repeated mTBI (rmTBI). There is a need for more sensitive imaging techniques capable of detecting subtle neurophysiological alterations after injury. We examined neurochemical and white matter changes using diffusion tensor imaging of the whole brain and proton magnetic resonance spectroscopy of the hippocampi at 7 Tesla in 18-day-old male rats at 7 days after mTBI and rmTBI. Traumatic axonal injury was assessed by beta-amyloid precursor protein accumulation using immunohistochemistry. A significant decrease in fractional anisotropy and increase in axial and radial diffusivity were observed in several brain regions, especially in white matter regions, after a single mTBI versus sham and more prominently after rmTBI. In addition, we observed accumulation of beta-amyloid precursor protein in the external capsule after mTBI and rmTBI. mTBI and rmTBI reduced the N-acetylaspartate/creatine ratio (NAA/Cr) and increased the myoinositol/creatine ratio (Ins/Cr) versus sham. rmTBI exacerbated the reduction in NAA/Cr versus mTBI. The choline/creatine (Cho/Cr) and (lipid/Macro Molecule 1)/creatine (Lip/Cr) ratios were also decreased after rmTBI versus sham. Diffusion tensor imaging findings along with the decrease in Cho and Lip after rmTBI may reflect damage to axonal membrane. NAA and Ins are altered at 7 days after mTBI and rmTBI likely reflecting neuro-axonal damage and glial response, respectively. These findings may be relevant to understanding the extent of disability following mTBI and rmTBI in the immature brain and may identify possible therapeutic targets.
Collapse
Affiliation(s)
- Emin Fidan
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, PA, USA
| | - Lesley M. Foley
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, PA, USA
- Animal Imaging Center, University of Pittsburgh, PA, USA
| | - Lee Ann New
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, PA, USA
| | - Henry Alexander
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, PA, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, PA, USA
| | - T. Kevin Hitchens
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, PA, USA
- Animal Imaging Center, University of Pittsburgh, PA, USA
| | - Hülya Bayır
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, PA, USA
- Children's Neuroscience Institute
| |
Collapse
|
43
|
Pre-clinical models in pediatric traumatic brain injury-challenges and lessons learned. Childs Nerv Syst 2017; 33:1693-1701. [PMID: 29149385 PMCID: PMC5909721 DOI: 10.1007/s00381-017-3474-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 05/30/2017] [Indexed: 12/31/2022]
Abstract
PURPOSE Despite the enormity of the problem and the lack of new therapies, research in the pre-clinical arena specifically using pediatric traumatic brain injury (TBI) models is limited. In this review, some of the key models addressing both the age spectrum of pediatric TBI and its unique injury mechanisms will be highlighted. Four topics will be addressed, namely, (1) unique facets of the developing brain important to TBI model development, (2) a description of some of the most commonly used pre-clinical models of severe pediatric TBI including work in both rodents and large animals, (3) a description of the pediatric models of mild TBI and repetitive mild TBI that are relatively new, and finally (4) a discussion of challenges, gaps, and potential future directions to further advance work in pediatric TBI models. METHODS This narrative review on the topic of pediatric TBI models was based on review of PUBMED/Medline along with a synthesis of information on key factors in pre-clinical and clinical developmental brain injury that influence TBI modeling. RESULTS In the contemporary literature, six types of models have been used in rats including weight drop, fluid percussion injury (FPI), impact acceleration, controlled cortical impact (CCI), mechanical shaking, and closed head modifications of CCI. In mice, studies are largely restricted to CCI. In large animals, FPI and rotational injury have been used in piglets and shake injury has also been used in lambs. Most of the studies have been in severe injury models, although more recently, studies have begun to explore mild and repetitive mild injuries to study concussion. CONCLUSIONS Given the emerging importance of TBI in infants and children, the morbidity and mortality that is produced, along with its purported link to the development of chronic neurodegenerative diseases, studies in these models merit greater systematic investigations along with consortium-type approaches and long-term follow-up to translate new therapies to the bedside.
Collapse
|
44
|
Glenn DE, Acheson DT, Geyer MA, Nievergelt CM, Baker DG, Risbrough VB. Fear learning alterations after traumatic brain injury and their role in development of posttraumatic stress symptoms. Depress Anxiety 2017; 34:723-733. [PMID: 28489272 DOI: 10.1002/da.22642] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/20/2017] [Accepted: 04/02/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND It is unknown how traumatic brain injury (TBI) increases risk for posttraumatic stress disorder (PTSD). One potential mechanism is via alteration of fear-learning processes that could affect responses to trauma memories and cues. We utilized a prospective, longitudinal design to determine if TBI is associated with altered fear learning and extinction, and if fear processing mediates effects of TBI on PTSD symptom change. METHODS Eight hundred fifty two active-duty Marines and Navy Corpsmen were assessed before and after deployment. Assessments included TBI history, PTSD symptoms, combat trauma and deployment stress, and a fear-potentiated startle task of fear acquisition and extinction. Startle response and self-reported expectancy and anxiety served as measures of fear conditioning, and PTSD symptoms were measured with the Clinician-Administered PTSD Scale. RESULTS Individuals endorsing "multiple hit" exposure (both deployment TBI and a prior TBI) showed the strongest fear acquisition and highest fear expression compared to groups without multiple hits. Extinction did not differ across groups. Endorsing a deployment TBI was associated with higher anxiety to the fear cue compared to those without deployment TBI. The association of deployment TBI with increased postdeployment PTSD symptoms was mediated by postdeployment fear expression when recent prior-TBI exposure was included as a moderator. TBI associations with increased response to threat cues and PTSD symptoms remained when controlling for deployment trauma and postdeployment PTSD diagnosis. CONCLUSIONS Deployment TBI, and multiple-hit TBI in particular, are associated with increases in conditioned fear learning and expression that may contribute to risk for developing PTSD symptoms.
Collapse
Affiliation(s)
- Daniel E Glenn
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | - Dean T Acheson
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, CA, USA.,Research Service, VA San Diego Healthcare System, CA, USA
| | - Caroline M Nievergelt
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | - Dewleen G Baker
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | - Victoria B Risbrough
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | -
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| |
Collapse
|
45
|
Bai R, Gao H, Han Z, Ge X, Huang S, Chen F, Lei P. Long-Term Kinetics of Immunologic Components and Neurological Deficits in Rats Following Repetitive Mild Traumatic Brain Injury. Med Sci Monit 2017; 23:1707-1718. [PMID: 28390198 PMCID: PMC5395134 DOI: 10.12659/msm.901124] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Despite growing awareness of repetitive mild traumatic brain injury (rmTBI), understanding of the involvement of long-term kinetics of immunologic components in the central and peripheral immune system took part remains incomplete. The present study aimed to provide a quantitative assay for certain immune system parameters in rmTBI rats. Material/Methods Neurological functions were assessed by modified Neurological Severity Score (mNSS) and Morris Water Maze (MWM), immunologic components from brain and peripheral blood were analyzed by flow cytometry (FCM), and concentrations of inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10 were measure by enzyme-linked immunosorbent assay (ELISA). Results Neurological functions of rmTBI rats were seriously impaired. In the brain, T cells were up-regulated and peaked at week 1. The percentage of CD4+ T cells decreased from week 1 to week 4, while CD8+ T cells notably decreased at week 1, then increased until week 4. The infiltration proportion of Treg cells was reduced at week 1 and peaked at week 2. CD86+/CD11b+ M1 peaked at week 4 and CD206+/CD11b+ M2 rose at week 1. IL-6/IL-10 showed a similar pattern, whose rise corresponded to the decrease in TNF-α at week 2 after rmTBI. FCM demonstrated peripheral immune dysfunction after rmTBI. Conclusions mNSS and MWM demonstrated neuronal deficits in rmTBI rats, and central and peripheral immune systems were implicated in the pathophysiological processes of rmTBI. Long-term immune response may play dual roles in injury and repair of rmTBI.
Collapse
Affiliation(s)
- Ruojing Bai
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China (mainland).,Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin, China (mainland).,Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China (mainland).,Key Laboratory of Injuries, Variation and Regeneration of Nervous System, Tianjin, China (mainland).,Laboratory of Neuro-Trauma, Tianjin Neurological Institute, Tianjin, China (mainland)
| | - Huabin Gao
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China (mainland).,Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin, China (mainland).,Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China (mainland).,Key Laboratory of Injuries, Variation and Regeneration of Nervous System, Tianjin, China (mainland).,Laboratory of Neuro-Trauma, Tianjin Neurological Institute, Tianjin, China (mainland)
| | - Zhaoli Han
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China (mainland)
| | - Xintong Ge
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China (mainland).,Key Laboratory of Injuries, Variation and Regeneration of Nervous System, Tianjin, China (mainland).,Laboratory of Neuro-Trauma, Tianjin Neurological Institute, Tianjin, China (mainland).,Department of Neurosurgery, Tianjin Neurological Institute General Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Shan Huang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China (mainland).,Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin, China (mainland).,Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China (mainland).,Key Laboratory of Injuries, Variation and Regeneration of Nervous System, Tianjin, China (mainland).,Laboratory of Neuro-Trauma, Tianjin Neurological Institute, Tianjin, China (mainland)
| | - Fanglian Chen
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China (mainland).,Key Laboratory of Injuries, Variation and Regeneration of Nervous System, Tianjin, China (mainland).,Laboratory of Neuro-Trauma, Tianjin Neurological Institute, Tianjin, China (mainland)
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China (mainland).,Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin, China (mainland)
| |
Collapse
|
46
|
Ascenzi P, di Masi A, Leboffe L, Fiocchetti M, Nuzzo MT, Brunori M, Marino M. Neuroglobin: From structure to function in health and disease. Mol Aspects Med 2016; 52:1-48. [DOI: 10.1016/j.mam.2016.10.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 01/01/2023]
|
47
|
Air-puff induced vocalizations: A novel approach to detecting negative affective state following concussion in rats. J Neurosci Methods 2016; 275:45-49. [PMID: 27984100 DOI: 10.1016/j.jneumeth.2016.10.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/27/2016] [Accepted: 10/28/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND Negative emotional states resulting from concussion are of increasing concern. In the current study, we developed a model to investigate negative affect following concussion in the projectile concussive impact (PCI) model. High frequency ultrasonic vocalizations (22kHz USVs) are associated with negative affective stimuli in rats. Changes in negative affective state were examined following PCI using a mild air-puff stimulus to elicit 22kHz USVs. NEW METHOD Forty-eight hours post-injury, animals were placed into a clean acrylic box lined with bedding. A 5min baseline recording was followed by 15 air puffs (55psi) spaced 15s apart aimed at the upper back and neck. RESULTS Injured animals produced on average 153.5±55.13 more vocalizations than shams, vocalizing on average 4min longer than shams. Additionally, concussed animals vocalized to fewer air-puffs, exhibiting a 1.5 fold lower threshold for the expression of negative affect. COMPARISON WITH EXISTING METHODS Studies currently used to test negative affective states following concussion in animals, such as the elevated plus maze and forced swim task have, as of yet, been unsuccessful in demonstrating injury effects in the PCI model. While the air-puff test has been applied in other fields, to our knowledge it has not been utilized to study traumatic brain injury. CONCLUSION The current study demonstrates that the air-puff vocalization test may be a valuable tool in assessing negative mood states following concussion in rat models and may be used to evaluate novel therapies following brain injury for the treatment of mood dysfunction.
Collapse
|
48
|
Abstract
Traumatic brain injury (TBI) has come to the forefront of both the scientific and popular culture. Specifically, sports-related concussions or mild TBI (mTBI) has become the center of scientific scrutiny with a large amount of research focusing on the long-term sequela of this type of injury. As the populace continues to age, the impact of TBI on the aging brain will become clearer. Currently, reports have come to light that link TBI to neurodegenerative disorders such as Alzheimer's and Parkinson's diseases, as well as certain psychiatric diseases. Whether these associations are causations, however, is yet to be determined. Other long-term sequelae, such as chronic traumatic encephalopathy (CTE), appear to be associated with repetitive injuries. Going forward, as we gain better understanding of the pathophysiological process involved in TBI and subclinical head traumas, and individual traits that influence susceptibility to neurocognitive diseases, a clearer, more comprehensive understanding of the connection between brain injury and resultant disease processes in the aging brain will become evident.
Collapse
|
49
|
Montenigro PH, Alosco ML, Martin BM, Daneshvar DH, Mez J, Chaisson CE, Nowinski CJ, Au R, McKee AC, Cantu RC, McClean MD, Stern RA, Tripodis Y. Cumulative Head Impact Exposure Predicts Later-Life Depression, Apathy, Executive Dysfunction, and Cognitive Impairment in Former High School and College Football Players. J Neurotrauma 2016; 34:328-340. [PMID: 27029716 DOI: 10.1089/neu.2016.4413] [Citation(s) in RCA: 342] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The term "repetitive head impacts" (RHI) refers to the cumulative exposure to concussive and subconcussive events. Although RHI are believed to increase risk for later-life neurological consequences (including chronic traumatic encephalopathy), quantitative analysis of this relationship has not yet been examined because of the lack of validated tools to quantify lifetime RHI exposure. The objectives of this study were: 1) to develop a metric to quantify cumulative RHI exposure from football, which we term the "cumulative head impact index" (CHII); 2) to use the CHII to examine the association between RHI exposure and long-term clinical outcomes; and 3) to evaluate its predictive properties relative to other exposure metrics (i.e., duration of play, age of first exposure, concussion history). Participants included 93 former high school and collegiate football players who completed objective cognitive and self-reported behavioral/mood tests as part of a larger ongoing longitudinal study. Using established cutoff scores, we transformed continuous outcomes into dichotomous variables (normal vs. impaired). The CHII was computed for each participant and derived from a combination of self-reported athletic history (i.e., number of seasons, position[s], levels played), and impact frequencies reported in helmet accelerometer studies. A bivariate probit, instrumental variable model revealed a threshold dose-response relationship between the CHII and risk for later-life cognitive impairment (p < 0.0001), self-reported executive dysfunction (p < 0.0001), depression (p < 0.0001), apathy (p = 0.0161), and behavioral dysregulation (p < 0.0001). Ultimately, the CHII demonstrated greater predictive validity than other individual exposure metrics.
Collapse
Affiliation(s)
- Philip H Montenigro
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts.,2 Department of Anatomy and Neurobiology, Boston University School of Medicine , Boston, Massachusetts
| | - Michael L Alosco
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts
| | - Brett M Martin
- 3 Data Coordinating Center, Boston University School of Public Health , Boston, Massachusetts
| | - Daniel H Daneshvar
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts
| | - Jesse Mez
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts.,4 Department of Neurology, Boston University School of Medicine , Boston, Massachusetts
| | - Christine E Chaisson
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts.,3 Data Coordinating Center, Boston University School of Public Health , Boston, Massachusetts.,5 Department of Biostatistics, Boston University School of Public Health , Boston, Massachusetts
| | - Christopher J Nowinski
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts.,6 Concussion Legacy Foundation , Waltham, Massachusetts
| | - Rhoda Au
- 4 Department of Neurology, Boston University School of Medicine , Boston, Massachusetts.,7 Framingham Heart Study, Boston University School of Medicine , Boston, Massachusetts
| | - Ann C McKee
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts.,4 Department of Neurology, Boston University School of Medicine , Boston, Massachusetts.,8 Department of Pathology, Boston University School of Medicine , Boston, Massachusetts.,9 VA Boston Healthcare System , Boston, Massachusetts
| | - Robert C Cantu
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts.,6 Concussion Legacy Foundation , Waltham, Massachusetts.,10 Department Neurosurgery, Boston University School of Medicine , Boston, Massachusetts.,11 Department of Neurosurgery, Emerson Hospital , Concord, Massachusetts
| | - Michael D McClean
- 12 Environmental Health, Boston University School of Public Health , Boston, Massachusetts
| | - Robert A Stern
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts.,2 Department of Anatomy and Neurobiology, Boston University School of Medicine , Boston, Massachusetts.,4 Department of Neurology, Boston University School of Medicine , Boston, Massachusetts.,10 Department Neurosurgery, Boston University School of Medicine , Boston, Massachusetts
| | - Yorghos Tripodis
- 1 Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine , Boston, Massachusetts.,5 Department of Biostatistics, Boston University School of Public Health , Boston, Massachusetts
| |
Collapse
|
50
|
A Narrative Review of Pharmacologic and Non-pharmacologic Interventions for Disorders of Consciousness Following Brain Injury in the Pediatric Population. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2016; 4:56-70. [PMID: 27280064 DOI: 10.1007/s40141-016-0108-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Traumatic brain injury (TBI) is the most common cause of long-term disability in the United States. A significant proportion of children who experience a TBI will have moderate or severe injuries, which includes a period of decreased responsiveness. Both pharmacological and non-pharmacological modalities are used for treating disorders of consciousness after TBI in children. However, the evidence supporting the use of potential therapies is relatively scant, even in adults, and overall, there is a paucity of study in pediatrics. The goal of this review is to describe the state of the science for use of pharmacologic and non-pharmacologic interventions for disorders of consciousness in the pediatric population.
Collapse
|