Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
BMJ Case Rep ; 14(2)2021 Feb 01.
Article En | MEDLINE | ID: mdl-33526531

This report describes two patients with acute-onset ptosis, oculomotor dysfunction, ataxia and drowsiness, referable to the midbrain tegmentum. Both patients had previously suffered severe closed head injuries requiring craniotomy for cerebral decompression. Serial brain scans in both cases revealed a newly developing cleft in the midbrain, with features suggestive of abnormal cerebrospinal fluid (CSF) flow across the aqueduct. A trial of acetazolamide was initiated to reduce CSF production, followed by a third ventriculostomy for CSF diversion in one patient, which resulted in arrested disease progression and partial recovery. There are only two previous reports in the literature of midbrain clefts that developed as remote sequelae of head trauma. We postulate that altered CSF flow dynamics in the aqueduct, possibly related to changes in brain compliance, may be contributory. Early recognition and treatment may prevent irreversible structural injury and possible death.


Brain Diseases/diagnostic imaging , Brain Injuries, Diffuse/diagnostic imaging , Cerebral Aqueduct/diagnostic imaging , Decompressive Craniectomy , Head Injuries, Closed/surgery , Mesencephalon/diagnostic imaging , Acetazolamide/therapeutic use , Ataxia/physiopathology , Blepharoptosis , Brain Diseases/physiopathology , Brain Diseases/therapy , Brain Injuries, Diffuse/physiopathology , Carbonic Anhydrase Inhibitors/therapeutic use , Cerebrospinal Fluid , Disease Progression , Dysarthria/physiopathology , Humans , Hydrodynamics , Magnetic Resonance Imaging , Male , Ocular Motility Disorders/physiopathology , Ventriculostomy , Young Adult
2.
J Neurosci Res ; 99(4): 1136-1160, 2021 04.
Article En | MEDLINE | ID: mdl-33319441

Elderly populations (≥65 years old) have the highest risk of developing Alzheimer's disease (AD) and/or obtaining a traumatic brain injury (TBI). Using translational mouse models, we investigated sleep disturbances and inflammation associated with normal aging, TBI and aging, and AD. We hypothesized that aging results in marked changes in sleep compared with adult mice, and that TBI and aging would result in sleep and inflammation levels similar to AD mice. We used female 16-month-old wild-type (WT Aged) and 3xTg-AD mice, as well as a 2-month-old reference group (WT Adult), to evaluate sleep changes. WT Aged mice received diffuse TBI by midline fluid percussion, and blood was collected from both WT Aged (pre- and post-TBI) and 3xTg-AD mice to evaluate inflammation. Cognitive behavior was tested, and tissue was collected for histology. Bayesian generalized additive and mixed-effects models were used for analyses. Both normal aging and AD led to increases in sleep compared with adult mice. WT Aged mice with TBI slept substantially more, with fragmented shorter bouts, than they did pre-TBI and compared with AD mice. However, differences between WT Aged and 3xTg-AD mice in immune cell populations and plasma cytokine levels were incongruous, cognitive deficits were similar, and cumulative sleep was not predictive of inflammation or behavior for either group. Our results suggest that in similarly aged individuals, TBI immediately induces more profound sleep alterations than in AD, although both diseases likely include cognitive impairments. Unique pathological sleep pathways may exist in elderly individuals who incur TBI compared with similarly aged individuals who have AD, which may warrant disease-specific treatments in clinical settings.


Alzheimer Disease/physiopathology , Brain Injuries, Diffuse/physiopathology , Inflammation/metabolism , Sleep/physiology , Aging/pathology , Aging/physiology , Animals , Cognitive Dysfunction , Cytokines/metabolism , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Microglia/metabolism , Monocytes
3.
J Vis Exp ; (156)2020 02 10.
Article En | MEDLINE | ID: mdl-32090988

Traumatic brain injury (TBI) is a leading cause of acquired epilepsy. TBI can result in a focal or diffuse brain injury. Focal injury is a result of direct mechanical forces, sometimes penetrating through the cranium, creating a direct lesion in the brain tissue. These are visible during brain imaging as areas with contusion, laceration, and hemorrhage. Focal lesions induce neuronal death and glial scar formation and are present in 20%-25% of all people who incur a TBI. However, in the majority of TBI cases, injury is caused by acceleration-deceleration forces and subsequent tissue shearing, resulting in nonfocal, diffuse damage. A subpopulation of TBI patients continues to develop post-traumatic epilepsy (PTE) after a latency period of months or years. Currently, it is impossible to predict which patients will develop PTE, and seizures in PTE patients are challenging to control, necessitating further research. Until recently, the field was limited to only two animal/rodent models with validated spontaneous post-traumatic seizures, both presenting with large focal lesions with massive tissue loss in the cortex and sometimes subcortical structures. In contrast to these approaches, it was determined that diffuse TBI induced using a modified weight drop model is sufficient to initiate development of spontaneous convulsive and non-convulsive seizures, even in the absence of focal lesions or tissue loss. Similar to human patients with acquired post-traumatic epilepsy, this model presents with a latency period after injury before seizure onset. In this protocol, the community will be provided with a new model of post-traumatic epilepsy, detailing how to induce diffuse non-lesional TBI followed by continuous long-term video-electroencephalographic animal monitoring over the course of several months. This protocol will detail animal handling, the weight drop procedure, the electrode placement for two acquisition systems, and the frequent challenges encountered during each of the steps of surgery, postoperative monitoring, and data acquisition.


Brain Injuries, Diffuse/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Epilepsy, Post-Traumatic/etiology , Epilepsy, Post-Traumatic/pathology , Animals , Brain/pathology , Brain/physiopathology , Brain Injuries, Diffuse/physiopathology , Brain Injuries, Traumatic/physiopathology , Cerebral Cortex/pathology , Epilepsy, Post-Traumatic/physiopathology , Male , Mice , Mice, Inbred C57BL , Seizures/etiology , Seizures/pathology
4.
Am J Respir Crit Care Med ; 201(2): 167-177, 2020 01 15.
Article En | MEDLINE | ID: mdl-31657946

Rationale: Older adults (≥65 yr old) account for an increasing proportion of patients with severe traumatic brain injury (TBI), yet clinical trials and outcome studies contain relatively few of these patients.Objectives: To determine functional status 6 months after severe TBI in older adults, changes in this status over 2 years, and outcome covariates.Methods: This was a registry-based cohort study of older adults who were admitted to hospitals in Victoria, Australia, between 2007 and 2016 with severe TBI. Functional status was assessed with Glasgow Outcome Scale Extended (GOSE) 6, 12, and 24 months after injury. Cohort subgroups were defined by admission to an ICU. Features associated with functional outcome were assessed from the ICU subgroup.Measurements and Main Results: The study included 540 older adults who had been hospitalized with severe TBI over the 10-year period; 428 (79%) patients died in hospital, and 456 (84%) died 6 months after injury. There were 277 patients who had not been admitted to an ICU; at 6 months, 268 (97%) had died, 8 (3%) were dependent (GOSE 2-4), and 1 (0.4%) was functionally independent (GOSE 5-8). There were 263 patients who had been admitted to an ICU; at 6 months, 188 (73%) had died, 39 (15%) were dependent, and 32 (12%) were functionally independent. These proportions did not change over longer follow-up. The only clinical features associated with a lower rate of functional independence were Injury Severity Score ≥25 (adjusted odds ratio, 0.24 [95% confidence interval, 0.09-0.67]; P = 0.007) and older age groups (P = 0.017).Conclusions: Severe TBI in older adults is a condition with very high mortality, and few recover to functional independence.


Brain Injuries, Traumatic/physiopathology , Glasgow Outcome Scale , Hospital Mortality , Abbreviated Injury Scale , Accidental Falls , Accidents, Traffic , Activities of Daily Living , Age Factors , Aged , Aged, 80 and over , Brain Contusion/mortality , Brain Contusion/physiopathology , Brain Contusion/therapy , Brain Injuries, Diffuse/physiopathology , Brain Injuries, Traumatic/mortality , Brain Injuries, Traumatic/therapy , Cerebral Hemorrhage, Traumatic/mortality , Cerebral Hemorrhage, Traumatic/physiopathology , Cerebral Hemorrhage, Traumatic/therapy , Cerebral Intraventricular Hemorrhage/mortality , Cerebral Intraventricular Hemorrhage/physiopathology , Cerebral Intraventricular Hemorrhage/therapy , Cohort Studies , Female , Hematoma, Subdural/mortality , Hematoma, Subdural/physiopathology , Hematoma, Subdural/therapy , Humans , Injury Severity Score , Intensive Care Units , Male , Mortality , Neurosurgical Procedures , Odds Ratio , Registries , Respiration, Artificial , Skull Fractures/mortality , Skull Fractures/physiopathology , Skull Fractures/therapy , Subarachnoid Hemorrhage, Traumatic/mortality , Subarachnoid Hemorrhage, Traumatic/physiopathology , Subarachnoid Hemorrhage, Traumatic/therapy , Tracheostomy , Victoria
5.
Mil Med ; 184(Suppl 1): 195-205, 2019 03 01.
Article En | MEDLINE | ID: mdl-30901406

Blast-induced traumatic brain injury (bTBI) has become a signature casualty of recent military operations. In spite of significant clinical and preclinical TBI research, current understanding of injury mechanisms and short- and long-term outcomes is limited. Mathematical models of bTBI biomechanics may help in better understanding of injury mechanisms and in the development of improved neuroprotective strategies. Until present, bTBI has been analyzed as a single event of a blast pressure wave propagating through the brain. In many bTBI events, the loads on the body and the head are spatially and temporarily distributed, involving the primary intracranial pressure wave, followed by the head rotation and then by head impact on the ground. In such cases, the brain microstructures may experience time/space distributed (consecutive) damage and recovery events. The paper presents a novel multiscale simulation framework that couples the body/brain scale biomechanics with micro-scale mechanobiology to study the effects of micro-damage to neuro-axonal structures. Our results show that the micro-mechanical responses of neuro-axonal structures occur sequentially in time with "damage" and "relaxation" periods in different parts of the brain. A new integrated computational framework is described coupling the brain-scale biomechanics with micro-mechanical damage to axonal and synaptic structures.


Biomechanical Phenomena/physiology , Biophysics , Blast Injuries/complications , Brain Injuries, Traumatic/complications , Blast Injuries/classification , Brain Injuries, Diffuse/physiopathology , Brain Injuries, Traumatic/classification , Computer Simulation , Humans , Models, Theoretical , Time Factors
6.
Eur J Neurosci ; 50(2): 1972-1980, 2019 07.
Article En | MEDLINE | ID: mdl-30762917

Sympathetic hyperactivity occurs in a subgroup of patients after traumatic brain injury (TBI). The rostral ventrolateral medulla (RVLM) is a key region for the activity of sympathetic nervous system. Oxidative stress in the RVLM is proved to be responsible for the increased level of sympathetic activity in animal models of hypertension and heart failure. In this study, we investigated whether oxidative stress in the RVLM contributed to the development of sympathetic hyperactivity after TBI in rats. Model of diffuse axonal injury was induced using Sprague-Dawley rats, and level of mean arterial pressure (MAP) and plasma Norepinephrine (NE) was measured to evaluate the sympathetic activity. For the assessment of oxidative stress, expression of reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD) in the RVLM was determined. Microinjection of Tempol into the RVLM was performed to determine the effect of oxidative stress on sympathetic hyperactivity. According to the results, TBI led to elevated MAP and plasma NE in rats. It also induced a significantly increased level of ROS, MDA production and decreased level of SOD in the RVLM. The sympathetic activity, ROS, and MDA in the RVLM decreased significantly after microinjection of Tempol. Therefore, the present results suggested that oxidative stress in the RVLM was involved in the development of sympathetic hyperactivity following TBI.


Brain Injuries, Diffuse , Hyperkinesis , Medulla Oblongata , Oxidative Stress , Psychomotor Agitation , Sympathetic Nervous System , Animals , Brain Injuries, Diffuse/metabolism , Brain Injuries, Diffuse/physiopathology , Disease Models, Animal , Hyperkinesis/metabolism , Hyperkinesis/physiopathology , Male , Medulla Oblongata/metabolism , Medulla Oblongata/physiopathology , Oxidative Stress/physiology , Psychomotor Agitation/metabolism , Psychomotor Agitation/physiopathology , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology
7.
Neuroscience ; 388: 152-170, 2018 09 15.
Article En | MEDLINE | ID: mdl-30036662

Changes in inhibition following traumatic brain injury (TBI) appear to be one of the major factors that contribute to excitation:inhibition imbalance. Neuron pathology, interneurons in particular evolves from minutes to weeks post injury and follows a complex time course. Previously, we showed that in the long-term in diffuse TBI (dTBI), there was select reduction of specific dendrite-targeting neurons in sensory cortex and hippocampus while in motor cortex there was up-regulation of specific dendrite-targeting neurons. We now investigated the time course of dTBI effects on interneurons in neocortex and hippocampus. Brains were labeled with antibodies against calbindin (CB), parvalbumin (PV), calretinin (CR) neuropeptide Y (NPY), and somatostatin (SOM) at 24 h and 2 weeks post dTBI. We found time-dependent, brain area-specific changes in inhibition at 24 h and 2 weeks. At 24 h post-injury, reduction of dendrite-targeting inhibitory neurons occurred in sensory cortex and hippocampus. At 2 weeks, we found compensatory changes in the somatosensory cortex and CA2/3 of hippocampus affected at 24 h, with affected interneuronal populations returning to sham levels. However, DG of hippocampus now showed reduction of dendrite-targeting inhibitory neurons. Finally, with respect to motor cortex, there was an upregulation of dendrite-targeting interneurons in the supragranular layers at 24 h returning to normal levels by 2 weeks. Overall, our findings reconfirm that dendritic inhibition is particularly susceptible to brain trauma, but also show that there are complex brain-area-specific changes in inhibitory neuronal numbers and in compensatory changes, rather than a simple monotonic progression of changes post-dTBI.


Brain Injuries, Diffuse/physiopathology , Brain Injuries, Traumatic/physiopathology , Cerebral Cortex/physiopathology , Hippocampus/physiopathology , Neurons/physiology , Animals , Brain Injuries, Diffuse/pathology , Brain Injuries, Traumatic/pathology , Cerebral Cortex/pathology , Disease Models, Animal , Disease Progression , Hippocampus/pathology , Male , Neural Inhibition/physiology , Neurons/pathology , Random Allocation , Rats, Sprague-Dawley , Time Factors
8.
Sci Rep ; 7(1): 13211, 2017 10 16.
Article En | MEDLINE | ID: mdl-29038483

Determining regions of altered brain physiology after diffuse brain injury is challenging. Microglia, brain immune cells with ramified and dynamically moving processes, constantly surveil the parenchyma for dysfunction which, when present, results in a changed morphology. Our purpose was to define the spatiotemporal changes in microglia morphology over 28 days following rat midline fluid percussion injury (mFPI) as a first step in exploiting microglia morphology to reflect altered brain physiology. Microglia morphology was quantified from histological sections using Image J skeleton and fractal analysis procedures at three time points and in three regions post-mFPI: impact site, primary somatosensory cortex barrel field (S1BF), and a remote region. Microglia ramification (process length/cell and endpoints/cell) decreased in the impact and S1BF but not the remote region (p < 0.05). Microglia complexity was decreased in the S1BF (p = 0.003) and increased in the remote region (p < 0.02). Rod-shaped microglia were present in the S1BF and had a 1.8:1.0 length:width ratio. An in-depth quantitative morphologic analysis revealed diverse and widespread changes to microglia morphology in the cortex post-mFPI. Due to their close link to neuronal function, changes in microglia morphology, summarized in this study, likely reflect altered physiology with diverse and widespread impact on neuronal and circuit function.


Brain Injuries, Diffuse/pathology , Microglia/pathology , Somatosensory Cortex/pathology , Animals , Brain Injuries, Diffuse/physiopathology , Fractals , Male , Percussion , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/physiopathology
9.
Am J Emerg Med ; 35(11): 1788.e5-1788.e6, 2017 Nov.
Article En | MEDLINE | ID: mdl-28801041

Diffuse axonal injury is usually caused by head trauma, and patients have significant clinical symptoms during admission to the emergency department. In our case, we present a five-year-old patient who was involved in a car accident. During admission to the emergency department, the patient had no symptoms of trauma. However, 6 h after admission to emergency service, neurological symptoms occurred, and mental status changed. Diffuse axonal injury (DAI) is characterized by diffuse nerve axon injury in the brain and brainstem. This is one of the worst results of a head trauma and occurs in one-third of the patients admitted to the hospital with head trauma. In some studies, it has been reported that diffuse axonal injury is permanent in accelerated and decelerated head traumas without accompanying loss of consciousness. Neurological sequels have occurred in the recovery phase of some patients with diffuse axonal damage. In this study, we present a delayed diffuse axonal injury case accompanying a head trauma.


Accidents, Traffic , Brain Injuries, Diffuse/diagnostic imaging , Brain Injuries, Diffuse/physiopathology , Child, Preschool , Electroencephalography , Humans , Magnetic Resonance Imaging , Male , Time Factors
10.
Unfallchirurg ; 120(9): 728-733, 2017 Sep.
Article De | MEDLINE | ID: mdl-28812113

Traumatic brain injury (TBI) constitutes a heterogeneous condition that affects the most complex organ of the human body. It is commonly classified by its location as focal injury (e.g. epidural hematoma) and diffuse injury (e.g. diffuse axonal shearing injury) as well as by primary and secondary tissue injury. Accordingly, direct mechanical force causes the primary insult. The tissue damage occurring afterwards is subsumed under the term secondary brain damage. Some of these processes are overlapping and include in the early phase local cerebral ischemia resulting in excitotoxicity, which together with the triggered neuroinflammatory cascade causes the formation of cerebral edema and ultimately increased intracranial pressure once the intracranial compliance is exhausted. In survivors the long-term sequelae of the late stage include seizures caused by synaptic reorganization (incidence depending on the severity of TBI), persistent neuroinflammation promoting further neurodegeneration and increased risk for Alzheimer's disease probably because of TBI-related protein misfolding (tauopathy). Acute phase biomarkers of TBI should ideally originate from the injured brain. They should help distinguish disease severity and predict morbidity and mortality; however, the most commonly used biomarkers (S-100ß and neurone-specific enolase) show a low specificity. In theory their successors (i. e. GFAP, pNF-H) seem more specific; however, these "new kids on the block" still need to be thoroughly investigated in large scale studies.


Brain Injuries, Traumatic/physiopathology , Biomarkers/metabolism , Brain/physiopathology , Brain Damage, Chronic/physiopathology , Brain Edema/classification , Brain Edema/physiopathology , Brain Injuries, Diffuse/physiopathology , Brain Injuries, Traumatic/classification , Glial Fibrillary Acidic Protein/metabolism , Hematoma, Epidural, Cranial/classification , Hematoma, Epidural, Cranial/physiopathology , Hematoma, Subdural/classification , Hematoma, Subdural/physiopathology , Humans , Intracranial Pressure/physiology , Neurofilament Proteins/metabolism , Phosphopyruvate Hydratase/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Synapses/physiology , Tauopathies/physiopathology
11.
J Neurotrauma ; 34(14): 2315-2319, 2017 07 15.
Article En | MEDLINE | ID: mdl-28249552

Traumatic brain injury (TBI) was shown to impair pressure-induced myogenic response of cerebral arteries, which is associated with vascular and neural dysfunction and increased mortality of TBI patients. Hypertension was shown to enhance myogenic tone of cerebral arteries via increased vascular production of 20-hydroxyeicosatrienoic acid (HETE). This adaptive mechanism protects brain tissue from pressure/volume overload; however, it can also lead to increased susceptibility to cerebral ischemia. Although both effects may potentiate the detrimental vascular consequences of TBI, it is not known how hypertension modulates the effect of TBI on myogenic responses of cerebral vessels. We hypothesized that in hypertensive rats, the enhanced myogenic cerebrovascular response is preserved after TBI. Therefore, we investigated the myogenic responses of isolated middle cerebral arteries (MCA) of normotensive and spontaneously hypertensive rats (SHR) after severe impact acceleration diffuse brain injury. TBI diminished myogenic constriction of MCAs isolated from normotensive rats, whereas the 20-HETE-mediated enhanced myogenic response of MCAs isolated from SHRs was not affected by TBI. These results suggest that the optimal cerebral perfusion pressure values and vascular signaling pathways can be different and, therefore, should be targeted differently in normotensive and hypertensive patients following TBI.


Brain Injuries, Diffuse/physiopathology , Brain Injuries, Traumatic/physiopathology , Cerebrovascular Circulation/physiology , Hydroxyeicosatetraenoic Acids/metabolism , Hypertension/physiopathology , Middle Cerebral Artery/physiopathology , Muscle, Smooth, Vascular/physiopathology , Animals , Brain Injuries, Diffuse/metabolism , Brain Injuries, Traumatic/metabolism , Disease Models, Animal , Hypertension/metabolism , Middle Cerebral Artery/metabolism , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY
12.
Dev Neurosci ; 38(3): 195-205, 2016.
Article En | MEDLINE | ID: mdl-27449121

Development and aging are influenced by external factors with the potential to impact health throughout the life span. Traumatic brain injury (TBI) can initiate and sustain a lifetime of physical and mental health symptoms. Over 1.7 million TBIs occur annually in the USA alone, with epidemiology suggesting a higher incidence for young age groups. Additionally, increasing life spans mean more years to age with TBI. While there is ongoing research of experimental pediatric and adult TBI, few studies to date have incorporated animal models of pediatric, adolescent, and adult TBI to understand the role of age at injury across the life span. Here, we explore repeated behavioral performance between rats exposed to diffuse TBI at five different ages. Our aim was to follow neurological morbidities across the rodent life span with respect to age at injury. A single cohort of male Sprague-Dawley rats (n = 69) was received at postnatal day (PND) 10. Subgroups of this cohort (n = 11-12/group) were subjected to a single moderate midline fluid percussion injury at age PND 17, PND 35, 2 months, 4 months, or 6 months. A control group of naïve rats (n = 12) was assembled from this cohort. The entire cohort was assessed for motor function by beam walk at 1.5, 3, 5, and 7 months of age. Anxiety-like behavior was assessed with the open field test at 8 months of age. Cognitive performance was assessed using the novel object location task at 8, 9, and 10 months of age. Depression-like behavior was assessed using the forced swim test at 10 months of age. Age at injury and time since injury differentially influenced motor, cognitive, and affective behavioral outcomes. Motor and cognitive deficits occurred in rats injured at earlier developmental time points, but not in rats injured in adulthood. In contrast, rats injured during adulthood showed increased anxiety-like behavior compared to uninjured control rats. A single diffuse TBI did not result in chronic depression-like behaviors or changes in body weight among any groups. The interplay of age at injury and aging with an injury are translationally important factors that influence behavioral performance as a quality of life metric. More complete understanding of these factors can direct rehabilitative efforts and personalized medicine for TBI survivors.


Anxiety/physiopathology , Brain Injuries, Diffuse/physiopathology , Brain Injuries, Traumatic/physiopathology , Cognition Disorders/physiopathology , Aging , Animals , Behavior, Animal/physiology , Male , Maze Learning , Rats, Sprague-Dawley
13.
Mil Med ; 181(5 Suppl): 23-7, 2016 05.
Article En | MEDLINE | ID: mdl-27168549

Traumatic brain injury (TBI) secondary to blast exposure is a common injury in the Global War on Terrorism, but little is known about the acute effects, recovery, pathophysiology, and neuropathology of blast-induced mild TBI (mTBI) in humans in a battlefield environment. Moreover, there is ongoing debate whether blast-induced mTBI is a different injury with a unique pathophysiology compared with mTBI from blunt trauma. In the case series reported here from Craig Joint Theater Hospital at Bagram Airfield, Afghanistan, 15 military service members with acute concussion/mTBI associated with blast exposure were evaluated within the first 24 hours after concussion and on days 2, 3, 5, and 7 with a Graded Symptom Checklist and a balance assessment, the Balance Error Scoring System. These data suggest that the recovery in blast-induced mTBI follows the pattern of recovery in sports-related concussion reported in The National Collegiate Athletic Association Concussion Study. In this retrospective case series, we provide the first description of the natural history of acute recovery in blast-induced mTBI, and we suspect, given our experience treating military service members, that further observations of the natural history of recovery in blast-induced mTBI will continue to mirror the natural history of recovery in sports concussion.


Athletic Injuries/rehabilitation , Brain Concussion/rehabilitation , Military Personnel/psychology , Adult , Afghan Campaign 2001- , Afghanistan , Brain Concussion/complications , Brain Injuries/physiopathology , Brain Injuries, Diffuse/physiopathology , Female , Humans , Male , Neuropsychological Tests/statistics & numerical data , Retrospective Studies
14.
J Neurotrauma ; 33(8): 734-40, 2016 Apr 15.
Article En | MEDLINE | ID: mdl-26414329

We have developed and implemented a noninvasive, objective neurofunctional assessment for evaluating the sustained effects of traumatic brain injury (TBI) in piglets with both diffuse and focal injury types. Derived from commercial actigraphy methods in humans, this assessment continuously monitors the day/night activity of piglets using close-fitting jackets equipped with tri-axial accelerometers to monitor movements of the thorax. Acceleration metrics were correlated (N = 7 naïve piglets) with video images to define values associated with a range of activities, from recumbancy (rest) to running. Both focal (N = 8) and diffuse brain injury (N = 9) produced alterations in activity that were significant 4 days post-TBI. Compared to shams (N = 6) who acclimated to the animal facility 4 days after an anesthesia experience by blurring the distinction between day and night activity, post-TBI time-matched animals had larger fractions of inactive periods during the daytime than nighttime, and larger fractions of active time in the night were spent in high activity (e.g., constant walking, intermittent running) than during the day. These persistent disturbances in rest and activity are similar to those observed in human adults and children post-TBI, establishing actigraphy as a translational metric, used in both humans and large animals, for assessment of injury severity, progressions, and intervention.


Actigraphy/methods , Brain Injuries, Diffuse/physiopathology , Circadian Rhythm/physiology , Motor Activity/physiology , Animals , Animals, Newborn , Brain Injuries/physiopathology , Female , Humans , Swine
15.
Brain Inj ; 30(2): 217-24, 2016.
Article En | MEDLINE | ID: mdl-26646974

PRIMARY OBJECTIVE: A dynamic relationship exists between diffuse traumatic brain injury and changes to the neurovascular unit. The purpose of this study was to evaluate vascular changes during the first week following diffuse TBI. It was hypothesized that pathology is associated with modification of the vasculature. METHODS: Male Sprague-Dawley rats underwent either midline fluid percussion injury or sham-injury. Brain tissue was collected 1, 2 or 7 days post-injury or sham-injury (n = 3/time point). Tissue was collected and stained by de Olmos amino-cupric silver technique to visualize neuropathology or animals were perfused with AltaBlue casting resin before high-resolution vascular imaging. The average volume, surface area, radius, branching and tortuosity of the vessels were evaluated across three regions of interest. RESULTS: In M2, average vessel volume (p < 0.01) and surface area (p < 0.05) were significantly larger at 1 day relative to 2 days, 7 days and sham. In S1BF and VPM, no significant differences in the average vessel volume or surface area at any of the post-injury time points were observed. No significant changes in average radius, branching or tortuosity were observed. CONCLUSIONS: Preliminary findings suggest gross morphological changes within the vascular network likely represent an acute response to mechanical forces of injury, rather than delayed or chronic pathological processes.


Brain Injuries, Diffuse/physiopathology , Animals , Brain/pathology , Brain Injuries, Diffuse/anatomy & histology , Brain Injuries, Diffuse/injuries , Disease Models, Animal , Male , Neuropathology , Rats , Rats, Sprague-Dawley
...