Transl. Stroke Res. DOI 10.1007/s12975-014-0327-0

REVIEW ARTICLE

Traumatic Brain Injury Using Mouse Models Yi Ping Zhang & Jun Cai & Lisa B. E. Shields & Naikui Liu & Xiao-Ming Xu & Christopher B. Shields

Received: 3 August 2013 / Revised: 9 December 2013 / Accepted: 5 January 2014 # Springer Science+Business Media New York 2014

Abstract The use of mouse models in traumatic brain injury (TBI) has several advantages compared to other animal models including low cost of breeding, easy maintenance, and innovative technology to create genetically modified strains. Studies using knockout and transgenic mice demonstrating functional gain or loss of molecules provide insight into basic mechanisms of TBI. Mouse models provide powerful tools to screen for putative therapeutic targets in TBI. This article reviews currently available mouse models that replicate several clinical features of TBI such as closed head injuries (CHI), penetrating head injuries, and a combination of both. CHI may be caused by direct trauma creating cerebral concussion or contusion. Sudden acceleration–deceleration injuries of the head without direct trauma may also cause

intracranial injury by the transmission of shock waves to the brain. Recapitulation of temporary cavities that are induced by high-velocity penetrating objects in the mouse brain are difficult to produce, but slow brain penetration injuries in mice are reviewed. Synergistic damaging effects on the brain following systemic complications are also described. Advantages and disadvantages of CHI mouse models induced by weight drop, fluid percussion, and controlled cortical impact injuries are compared. Differences in the anatomy, biomechanics, and behavioral evaluations between mice and humans are discussed. Although the use of mouse models for TBI research is promising, further development of these techniques is warranted. Keywords Traumatic brain injury . Mouse . Animal models

Y. P. Zhang : L. B. E. Shields : C. B. Shields (*) Norton Neuroscience Institute, Norton Healthcare, 210 East Gray Street, Suite 1102, Louisville, KY 40202, USA e-mail: [email protected] Y. P. Zhang e-mail: [email protected] L. B. E. Shields e-mail: [email protected] J. Cai Department of Pediatrics, School of Medicine, University of Louisville, Donald Baxter Building, Suite 321B, 570 South Preston Street, Louisville, KY 40292, USA e-mail: [email protected] N. Liu : X.90 days [225]. Football and other sports injuries are common in humans, therefore, models of these injuries would be beneficial to study the effects of concussion/contusion related to sports injuries. A rat model using a 7.4- to 11.2-m/s velocity at impact using a 50-g impactor on a helmet-protected head replicating cerebral concussion arising from football injuries

Transl. Stroke Res.

has been described [75]. Gross pathological changes were observed in 11, 28, and 33 % of animals subjected to 7.4-, 9.3-, and 11.2-m/s single impacts, respectively. When a 100-g impactor was used, greater energy transfer further increased the frequency and severity of injury [75]. A blast injury model using mice has been developed to recapitulate the pathological changes of tau-body encephalopathy observed in football players [71]. Using the wild-type C57BL/6 mouse exposed to a single blast, phosphorylated tauopathy, myelinated axonopathy, microvasculopathy, chronic neuroinflammation, and neurodegeneration were observed in the absence of macroscopic tissue damage or hemorrhage. Memory deficits persisting for >1 month correlated directly with impaired axonal conduction along with defective long-term potentiation of synaptic transmission. Such models replicate injuries that occur in athletes and correlate the magnitude of head trauma to persistent impairments in neurophysiological function, learning, and memory. Head immobilization during blast exposure prevented blast-induced learning and memory deficits [71]. Rodent models of head injury require induction of anesthesthetic agents that limit immediate postinjury behavior assessment such as orientation, memory, and level of consciousness. Head injuries have uncommonly been created in rats without anesthetic agents [161]; however, animal rights laws have largely prevented such injuries without sedation or anesthesia. Cumulative Effects of Multiple Head Injuries A single impact to the head may cause minimal or no clinical symptoms in humans. Cumulative mild head injuries may become symptomatic following boxing and football injuries as well as battlefield trauma [92, 199]. Repetitive head injuries or blast injuries may increase the risk of developing major delayed neurological diseases such as Alzheimers disease, motor neuron disease, and Lewy body disease [143, 147]. The so-called second-impact syndrome may cause rapid neurological deterioration following a second cerebral concussion after the effects of the initial one have fully resolved, resulting in long-term cognitive or behavioral abnormalities [71] or even an abrupt increase in intracranial pressure which may lead to brain herniation and death [26, 142]. It is unknown why repetitive concussions causing chronic traumatic encephalopathy lead to cognitive deficits (memory loss and conceptual computation problems) and post-traumatic stress disorders (PTSD) [75, 213]. Using a rat model, repetitive blast injuries under general anesthesia induced a variety of PTSDrelated behavioral traits, such as increased anxiety, enhanced contextual fear conditioning, and altered response to predator scent assay, that lasted for several months after the blast exposure [56]. As these injuries were performed in anesthetized rats, we are unable to evaluate psychological factors in

producing PTSD-like injuries. A repetitive concussion model in rats was developed to study the effects of multiple brain injuries [34, 49, 151]. DeRoss et al. reported that multiple concussions in rats caused immediate transient impairment in spatial recognition as well as extended effects on baseline performance [49]. These injuries cause impairment in motor function, memory, and learning abilities [87]. Chen et al. developed a projectile concussive impact (PCI) model in rats that replicated cerebral concussion in humans causing significant sensorimotor abnormalities 1 to 24 h after PCI injury [31]. Repeated blast injuries exacerbate histopathological and functional outcomes in rat and mouse models [94, 114, 206, 212]. Repetitive TBI can be created experimentally [188]. Usually, the CHI method [95] is utilized, but combining CCI and CHI methods may be performed sequentially to produce such injuries [44]. The effects of multiple cerebral concussions/contusions are cumulative in the mouse, with cognitive impairment observed only after repetitive concussions [42, 46, 220]. Brain damage may be significant when mild repeatitive TBI occur within a brief time window or during a period of vulnerability [145, 167, 168]. Single cerebral concussions do not cause cognitive impairment in mice [220]. Using a rat model of two consecutive head injuries using the mild cortical impact model, brain injuries 1 or 3 days apart revealed significantly greater lesion volumes observed on MRI compared with a single mild TBI and shams [85]. This finding was further supported by increased cortical tissue damage, extravascular iron, and glial activation in the multiple head injured rats with an interval of 1 and 3 days between impact. Behavioral changes at 1 month postinjury included reduced exploratory behavior and spatial learning impairment [85]. A model of repetitive closed TBI in mice has been developed with 24 h between controlled rubber impact injuries [186]. The injuries produced deficits in the Morris water maze performance which lasted as long as 7 weeks postinjury, with extensive abnormalities observed in the corpus callosum, bilateral external capsule, and ipsilateral cerebral cortex and thalamus noted on silver stain. A mouse model of repetitive mild TBI has been developed using the modified Marmarou WD method under mild anesthesia [95]. These animals were not restrained which allowed rapid acceleration of the head and torso. A scalp incision was not required. The procedure can be completed in 1–2 min. Minor deficits in motor coordination and locomotion hyperactivity recovered over time. This method is simple, cost effective, and is ideal for high throughput screening of potential new therapies [95]. Additional evidence of increased cognitive effects following multiple concussive brain injuries in mice has been reported with improvement of cognition following vitamin E administration [39, 42, 71]. Head immobilization during blast exposure prevented blast-induced learning and memory deficits in mice [71].

Transl. Stroke Res.

Brain Injury Without Direct Head Impact Forces arising from the shock wave of an explosion or from a sudden acceleration–deceleration motion of the head may cause a TBI constituting a double hit [135].

brain produced a cumulative effect on the pathological appearance of the brain and behavioral recovery [29, 105, 213]. Use of single and repeated blast exposures to the entire mouse has caused changes in several plasma enzymes suggesting that these enzymes may be used as a marker of trauma severity [9].

Blast-Induced Traumatic Brain Injury

Acceleration–Deceleration Induced Brain Injuries

Explosive forces may cause brain injury without direct impact to the head. Mechanisms of such an injury may be due to: (1) shock waves to the brain emanating from the explosion, (2) projectiles jettisoned from the explosive device causing brain trauma, (3) uncontrolled movement of the head in any direction (acceleration–deceleration), and (4) secondary damage to the brain arising from hemorrhagic shock, burns, and respiratory damage [30, 135, 153]. The first mechanism of blastinduced traumatic brain injury (bTBI) is unique and frequently occurs on the battlefield. Veterans may present with PTSD and other behavioral brain disturbances. These injuries occur most frequently following exposure to multiple explosions [7, 33, 48, 65, 71, 115, 122, 196, 214]. The mechanisms of brain damage caused by shock waves are unclear, however, it has been suggested that (1) shock waves cause an acoustic impedance mismatch that induces a shock-bubble effect in brain tissue and (2) the blast may cause increased pressure in body cavities that induce a blood surge to the brain that produces brain dysfunction [41], and (3) mitochondrial dysfunction [8]. Experimental bTBI has been studied in animals by subjecting them to a Friedlander blast wave that instigates a positive pressure wave followed immediately by a negative suction force. Rat models may consist of a “shock tube” that is closed at one end and open at the other with a shock wave generated by detonation of an explosive device or release of compressed air [29, 171, 194]. The shock wave is created by rupturing a membrane separating two compartments with different air pressures. An animal is placed in the chamber with the lower pressure and is subjected to a brief, sudden high-pressure shock wave when the membrane is punctured [171]. Rodents sustain a bTBI after the shock wave is transmitted to it. Shock waves may be produced by the release of compressed air generated by an oxygen–hydrogen combustion system or by an explosive detonation as demonstrated in several animal models: rats [108, 122, 170, 171, 173] and swine [14]. The peak and duration of the pressure wave may be measured which creates a scalable injury in these bTBI models. Severe high-pressure shock waves may kill the animal as a result of lung damage. However, this effect may be mitigated by a protective vest applied to animals [122]. Mild nonrepetitive blast injuries have replicated the behavior, cognitive, and MRI changes compatible with diffuse mild head injuries observed clinically [175]. These deficits may persist during the subacute and chronic phases following injury. Mouse bTBI models demonstrate that multiple exposures of shock waves to the

Violent repetitive shaking of the head may induce brain injury due to unsynchronized motion of the brain within the skull without skull impact or any external evidence of head trauma (Fig. 2) [160, 183]. Shaken baby syndrome (SBS) refers to abusive shaking of a child’s trunk that is often overlooked because of the lack of scalp trauma [183]. Neck muscles of infants and children are weak and unable to limit acceleration– deceleration or rotational movement of the head [160]. Although SBS is usually observed in children less than 2 years of age, it has been reported in older and heavier children up to 7 years old [178]. This injury is caused by the brain striking the internal bony surfaces of the skull. SBS usually causes the pathological picture of subdural, subarachnoid and retinal hemorrhages, and encephalopathy that may result in impairment of vision as well as motor and cognitive function [191]. A similar injury is frequently observed in adults caused by unanticipated flexion–extension movement of the unrestrained head when protective effects of neck muscles are overcome by major forces occurring in motor vehicle accidents (whiplash injury) [68]. Short-term effects of brain injury consist of behavioral regression, whereas long-term effects include mental health disorders and chronic neuropsychiatric complaints throughout life [24]. Models of TBI caused by acceleration–deceleration injuries have been developed to investigate mechanisms of brain damage and measures of biomarkers [174]. Models of TBI caused by acceleration–deceleration injuries have been developed to investigate mechanisms of brain damage producing effects that can be seen immediately or long term. Sudden deceleration of the head in monkeys creates a whiplash-like injury that produces brain damage similar to that seen in humans [106, 132, 157]. Rats subjected to rotational acceleration–deceleration head motion in a sagittal rotational direction (≤30° in 0.4 ms) caused significant brain injury [45, 211], transient impairment in walking and memory, and elevation of several serum biomarkers (neurofilament heavy chain, Tau, S100B, and myelin basic protein) at 1, 3, and 13 days postinjury [174]. Inertial forces required to create brain damage by acceleration–deceleration and rotational forces are dependent on a combination of brain mass and acceleration vectors. The brain of the adult mouse is approximately 1/3,000 the weight and 1/30 the radius of the human brain and, therefore, the production of a brain injury in the mouse is more difficult to create than in larger animals. The severity of the brain injury caused

Transl. Stroke Res. Fig. 2 Coup and contrecoup injuries in humans are caused by either a lateral (top left) or frontal (top right) head impact. The brain beneath the initial impact site is contused (large arrow) along with contusion of the contralateral side (small arrow). Inertial forces develop during head acceleration–deceleration injuries that cause remote (contralateral) damage in humans. The greater the volume of the brain, the greater the inertial forces will be. In mice (lower two figures), tiny volumes of brain in a flat-shaped skull are surrounded by thick muscles (arrows) that buffer lateral injury forces. The skull vertex, the site most frequently used for mouse TBI, is not covered by a thick muscle

by head acceleration–deceleration depends on the direction, amplitude, and velocity of the applied forces [82]. Mouse models of SBS have been developed in which anesthetized pups were shaken for 15 s on a horizontally rotating shaker at a frequency of 900 cycles/min [18, 19]. Hemorrhagic lesions of the brain were observed at postnatal day 13 (PN-13), with cysts developing gradually between PN-15 and 31. All shaken animals, with or without focal lesions, had thinning of the hemispheric white matter by PN-31 [18]. This model mimicked several aspects of human SBS and provided evidence that the release of glutamate played a role in its pathophysiology [18]. Repeat head shaking or flexion–extension rotational acceleration also increased levels of oxidative stress markers in the brain [156]. Acute subdural hematomas (aSDH) are common causes of death in humans following head injury, reaching 16–40 % in different series [79, 112, 176, 198]. Alesandri et al. developed a rat model of an a SDH that consisted of 300 microL blood injected into the subdural space [5]. Their study demonstrated a peak of TUNEL-positive cells indicating the presence of apoptosis processes and that caspase-dependent mechanisms play a major role in lesion development [5]. A mouse model of SDH has also been developed by the injection of nonheparinized blood into the subdural space with the magnitude of injury modulated by injecting different volumes of blood [179]. Sasaki and Dunn reported that this mouse model provided a method of investigating the effects of genotype on the brain’s response to acute SDH [179]. Epidural hematomas (EDH) may complicate acute head injury in humans. EDH is often associated with skull fractures and bleeding from meningeal arteries or venous sinuses into the epidural space. Survival depends on expeditious clot

evacuation, and mortality increases with associated intracranial pathology such as aSDH, cerebral contusion, or DAI [113]. To date, no mouse model of EDH has been developed.

Penetrating Traumatic Brain Injuries These injuries are customarily caused by high-velocity projectiles (shrapnel) or low-velocity objects (bone fragments) that impale the brain. Tissue damage and clinical manifestations depend on the anatomical path and degree of energy transfer [217]. High-speed objects such as bullets create greater tissue damage due to shock waves that destroy adjacent tissue than those caused by the direct laceration [151, 216, 217]. Open head injury models have been developed in rats to replicate tissue destruction caused by high energy penetrating objects in the brain [215, 218]. The progression of a nonfatal injury with a unilateral frontal penetrating traumatic brain injuries (pTBI), including the frontal cortex and striatum, showed that the volume of brain damage in the mouse increased over a course of 7 days [216]. It was converted from a state of intracerebral hemorrhage (0–6 h), to a core area of necrosis, infiltration of neutrophils, and macrophages, to degeneration of neurons and fiber tracts (thalamus, internal and external capsule, and cerebral peduncle) remote from the core lesion (3–7 days) [216]. Replicating heat-based TBI caused by high kinetic energy such as that produced by high-velocity penetrating objects is not feasible in mice because of the small volume of the brain. The use of slow penetrating objects caused by blast or projectiles in this review was not associated with heat-based damage of the brain. Penetrating injuries have also been reproduced in mice by inserting a sharp object into the

Mouse Rat

Rat

Rat

WDI

Temporal FPI (4.9±0.3 atm)

Parasagittal FPI (1.8–2.1 atm)

Moderate parasagittal FPI (1.14–2,18 atm) FPI (1.8–2.17 atm)

Hypotension and pyrexia

Gp I: 5 min forebrain ischemia. Gp II: 60 min forebrain ischemia. Gp III: TBI. Gp IV: TBI followed by 15 min ischemia. Alcohol consumption

Swine

Rat

Mouse Mouse Rat

Rat

Rat Rat

Mouse

FPI (3 atm)

Mild lateral CCI (3 m/s, 2.5 mm deformation)

CCI

CCI

Major TBI using the modified Richmond impact acceleration model Impact acceleration/TBI

TBI

Mild mechanical injury

TBI

Hemorrhagic hypotension (2 ml/100 g caused mBP=35–40 for 90 min. Resuscitated by (1) LR, (2) Hestend, and (3) PNPH

Forebrain ischemia

Effect of CCI model and hemorrhagic shock (90 min) CCI and hemorrhagic shock (HS)

Hemorrhagic hypotension (30 mg Hg) rescucitated to baseline for >60 mins. 2 gps studied: 1) tap water through G-tube, 2) ethanol (4 g/kg) through G-tube Bilateral CAO produced 1 h after CCI

Swine

Rat

Hemorrhagic hypotension (60 mmHg for 30 min) induced 5 min after FPI Rats subjected to FPI when anesthetized followed by 30 min normoxic or hypoxic states Rats subjected to FPI when anesthetized followed by 30 min normoxic or hypoxic states Cocaine administration IV 10 min (4 mg/kg) and 1 min (2 mg/kg) prior to injury

Hypoxia 30 min

Hypoxia (PaO2 =40 Hg for 30 min)

Effect of TBI and hypoxia (9 % oxygen)

Complication studied

Parasagittal FPI (1.94–2.18 atm) FPI (3 atm)

Rat

Animal

Type of brain injury

Table 5 Models used to study different types of brain injuries

TBI and ischemia separately produced identical ion dysfunction (increased [K+]e and decreased [Na+]e. Secondary ischemia (Gp IV) sustained ion dysfunction and increased ICP Alcohol produced a decrease in [Na+]serum 24 h postinjury producing edema Extensive EEG spike activity and delayed return of EEG+EP. Extensive bilateral CA1 and subarticular cell loss in hippocampus in combined TBI and ischemic group (but not either alone) PNPH and Hextend required less volume to resuscitate than RL. PNPH improved mBP and Hextend and LR did not. Fewer dying neurons in CA1 in mice treated with PNPH vs. LR and Hextend.

Bilateral CAO alone produced no cortical or hippocampal damage. 1–3 m/s CCI caused no detectible cortical contusion and minimal HC neuronal loss. 5 m/s caused small cortical contusion. 1, 3, and 5 m/s CCI followed by bilat CAO produced increased contusion volume an increased neuronal loss in HC Hemorrhagic shock exacerbated PI ischemia in mediating secondary injury in CCI Increased functional deficits and neuropath changes when CCI/HS combined compared to the effect on either alone No evidence of worsening of brain damage when studied pathologically at 4.5 h postinjury

Neuronal apoptosis inhibitor protein increased the vulnerability of brain cells to hypoxia Increased morbidity; weakness 71 % greater with hypoxia+TBI vs. 29 % with impact alone vs. 0 % weakness with hypoxia alone (24 h postinjury) Post-traumatic hypoxia increases contusion volume compared to normoxic rats. Post-traumatic hypothermia followed by slow rewarming (120 m) significantly decreases contusion volume Hypotension increased area and total volume of contusion compared with normotensive FPI rats Rats under hypoxic state had increased SM deficits and cognitive impairments 3 days FPI rats killed. Hypoxic rats had increased cortical contusion volumes and increased damage to HC (CA1 and CA2). No significant difference in CPP & CBF in cocaine treated vs. control groups. Nonsignificant trend to decrease ICP in group administered cocaine (1) Hemorrhage volume greater; (2) resuscitation requirements higher; (3) metabolic parameters worse; (4) survival time decreased in ethanol compared with tap water-infused group

Results

[58]

[91]

[96]

[192]

[109]

[81]

[62]

[36]

[224]

[141]

[22]

[22]

[137]

[137]

[89]

[146]

Reference

Transl. Stroke Res.

FPI fluid percussion injury, SDH subdural hematoma, CCI controlled cortical injury, HC hippocampus, PI postinjury, RL Ringer’s lactate, PNPH Polynitroxylated pegylated hemoglobin, SM sensorimotor, TBI traumatic brain injury

[180] Gp I: ASDH. Gp II: ASDH and DBI (?). Gp III: ASDH and DBI and hypoxia Rat Acute SDH and impact acceleration model

[88] Mouse TBI and intracerebral hemorrhage (ICH)

TBI

Mouse

[118]

[73]

Hypoxia after TBI produces increased inflammatory response and severity of secondary brain injury Treatment with propranolol increased CPP and decreased cerebral hypoxia Statins increase functional outcome, decrease neuronal degeneration, increase preserved neuronal density, and downregulate inflammatory gene expression Early evacuation of ASDH caused no change in edema at 5 h PI. Gp II: early evacuation caused decreased ICP and brain water content. Gp III: Progressive increased ICP, decreased CBF, and severe brain swelling. Early evacuation of ASDH increased brain edema. Hypoxic environment vs. normal O2 for 5 h after TBI Treatment with propranolol (beta blocking agent) vs. placebo after TBI TI and ICH treated with statins Mouse Blast TBI

Reference Results Complication studied Animal Type of brain injury

Table 5 (continued)

Transl. Stroke Res.

brain [76, 165]. In the latter series, the authors developed a nonfatal model of pTBI based on a modified air rifle that accelerated a pellet which impacted a small probe causing a penetrating injury of the rodent brain. They characterized brain tissue destruction and subsequent behavior models to examine the neurological function and impairment of memory function [165]. Stab wounds produced by driving a sharp instrument through a 4×2 mm midline craniotomy are unable to replicate the ballistic effects that transpire during a bTBI in rats [6, 57]. Mouse models of high-velocity missile PBI have not been developed, however, several examples of stab wounds into the cerebral cortex in mice have been reported during studies of glial scar formation [84]. Studies on the blood–brain barrier disruption and angiogenesis in urokinase-type plasminogen activators have also been conducted [97].

Combined Traumatic Brain Injuries TBI models that combine multiple types of head injuries and associated comorbidities (hemorrhage, hypoxia, multiple organ trauma) replicate the complex clinical picture of several pathological events such as DAI, cerebral contusion, and intracerebral hemorrhage observed in humans. Use of such a variety of models will increase our understanding of numerous deleterious factors that contribute to the clinical course following TBI. Most combination TBI models were developed in rats and swine (Table 5) [22, 36, 70, 89, 91, 96, 109, 137, 141, 180, 184, 192, 224]. Many behavioral, neuropathological, and immunological studies have been performed using CCI, WDI, and FPI models combined with known damaging effects following trauma (Table 5).

Limitations of Mouse Models in Replicating Clinical TBI Human TBI has been replicated in several mouse models using multiple forces including shock waves, FP, or cortical contusion. Even under forces of similar parameters, the type of brain injuries in humans and mice differ in several respects. Differences in Size and Shape of the Brain Between Humans and Mice Although mice and humans share many similarities in brain structure and function, differences in size and shape of the skull result in significant obstacles in creating mice models that mimic human TBI [208]. The mouse brain lies within an oblong skull and has inherent anatomical differences from the human that causes brain injuries in different locations. TBI in humans is usually caused by forces striking the frontal, occipital, or temporal areas of the skull. Contrarily, forces producing experimental TBI in mice are usually applied to the skull

Transl. Stroke Res.

vertex. Coup and contrecoup injuries occur frequently in humans following head injuries in which a cerebral contusion occurs directly beneath the impact site as well as contralaterally (Fig. 2). Most mice models of TBI do not reproduce this type of injury. Kilbourne et al. created an injury model in rats by creating a frontal impact injury with lateral rotation that did not produce skull fractures, prolonged apnea, or death [100]. However, petechial hemorrhage and DAI affected the orbitofrontal cortex (coup), corpus callosum, caudate, putamen, thalamus, cerebellum, and brainstem. Neurobehavioral dysfunction lasted for more than 1 week. They concluded that this frontal sagittal rotation acceleration injury produces diffuse injuries that may be relevant to human brain injury [100]. Difficulty in Comparing the Behavior Status in Mice and Humans Quantitative evaluation of neurological and behavioral deficits following clinical TBI is crucial in assessing the severity of brain injury, clinical recovery, and the effect of putative therapies. Functional deficits following human TBI are assessed by the evaluation of cognition and speech, alteration in social skills, depression, anxiety, personality changes, and motor/sensory abnormalities. Humans who sustain TBI may demonstrate measurable long-term neurological disabilities that impair daily activities [28]. These disabilities may include deficits in visual selective attention and decreased attention

function that may recover beyond 1 year postinjury [13]. Deficits in attention, nonverbal fluency, and verbal memory as well as slower visual and tactile reaction times may occur [136]. Evaluation of these psychological and neurological parameters depends on the patient’s verbal communication skills, which are unable to be performed in mice. Studies in rats have also demonstrated that multiple subacute mild head injuries using a FP device cause immediate transient impairment of spatial recognition. As the number of concussions increases from 1 to 3, a greater number of trials are required to return to baseline function using the Morris water maze [49]. By contrast, impairment is less obvious, and neurological recovery is more rapid and complete in mice. The open-field locomotor test using the Basso Mouse Scale measures hindlimb function following SCI but is not a sensitive motor behavioral assay following TBI in mice [12]. Evaluation of gait in mice following CCI-TBI has been performed using a computer-assisted automated gait analysis system. Neumann reported that the area of paw contact was decreased and that relative paw placement between fore- and hindpaw was altered, suggesting that TBI affected sensorimotor function and reduced interlimb coordination [154]. Sensitive evaluation tools or challenge tests are used to assess neurological dysfunction in rodents. The Rotarod or beam-walking tests detect motor dysfunction, and the whisker nuisance task detects somatosensory impairment [110, 144]. Cognitive skills in mice may be assessed using attention deficit testing, reduction in novelty exploration, and observation of hyperactivity/

Table 6 Rodent assessment tools used to mimic human cognitive, sensorimotor, and psychiatric function

Cognitive and sensorimotor function

Humans

Mice

Shortened attention span Memory difficulties Problem solving difficulties Learning difficulties Poor judgment Language deficits Inability to understand abstract concepts Loss of reading and writing skills Motor/Sensory dysfunction

Morris water maze; swimming T-maze/passive avoidance test; multivariate concentric square field test; forced swimming test [23, 38, 55, 64, 77, 123, 130, 148, 202, 205, 225]

Post-traumatic epilepsy

Psychiatric deficits

Changes in sexual function Sleep problems Inability to empathize with others Tendency to be more self-centered Inability to control emotions; increased irritability, frustration, aggressive behavior, depression, and mood swings

N/A N/A N/A Rotarod test, adhesive removal test, motor recovery score, wire grip score (grip strength), footprint, cylinder test, beam-walking task [20, 38, 63, 127, 140, 159, 163, 219, 228], whisker nuisance task [110, 144, 154] Electrically induced convulsions, seizure activity assessed by EEG [37, 80, 99, 104] N/A N/A N/A N/A Forced swimming [182, 226], tail suspension test, elevated plus-maze test [43, 164, 182], and ultrasonic vocalization [128]

Transl. Stroke Res.

hypoactivity (Table 6). A unique composite behavioral score index from individual complimentary behavioral tests in mice provided excellent discrimination across mild, moderate, and severe injuries compared to individual tests [223]. Nevertheless, without effective verbal communication, it is difficult to measure high-level cerebral function in mice [164]. Behavior and affective functioning may be assessed using ultrasonic vocalization in rats. Vocalization in ultrasonic frequencies expresses emotional states with high frequency calls (50 kHz) associated with a positive effect and low frequency calls (22 kHz) representing a negative emotional state [128]. The functional status of mice after TBI may be evaluated using the Neurological Severity Score which includes ten tasks that predict motor dysfunction, cognitive damage, and neurobehavioral function [205]. However, mice that were exposed to a very low intensity blast-induced brain injury demonstrate cognitive, behavioral, and MRI changes compatible with diffuse mild brain injury [175].

Conclusions Current mouse models replicate several biomechanical forces seen in human TBI and produce several pathological changes such as hemorrhage, necrosis, axon damage, scar formation, and alterations in behavior. There is a need to develop more models of mouse TBI that more effectively mimic human TBI. The use of mouse models is highly desirable because of the availability of genetically modified strains. Loss-offunction or gain-of-function mutations will provide the ability to create inexpensive models to purchase and maintain and, most importantly, will provide insight into mechanisms of TBI. Gene modifications may affect injury outcomes including fatality rates. The selection of injury models/injury parameters using transgenic mice must be carefully chosen to meet desired behavioral or pathological outcomes. However, animal models, including those in mice, will not completely replicate TBI in humans because trauma is produced by many biomechanical forces that are not easily replicated in animals. Animals are able to receive biomechanically controlled TBI in the laboratory whereas humans are not [158]. Furthermore, effective treatments for TBI in one animal model does not guarantee efficacy in another model due to variable responses in different species, sexes, ages, and weights [133, 139].

Conflict of Interest Yi Ping Zhang declares that he has no conflict of interest. Jun Cai declares that he has no conflict of interest. Lisa B.E. Shields declares that she has no conflict of interest. Naikui Liu declares that he has no conflict of interest. Xiao-Ming Xu declares that he has no conflict of interest. Christopher B. Shields declares that he has ownership in the Louisville Impactor System, Inc.

References 1. Adams JH, Graham DI, Murray LS, Scott G. Diffuse axonal injury due to nonmissile head injury in humans: an analysis of 45 cases. Ann Neurol. 1982;12(6):557–63. 2. Ahmad A, Crupi R, Campolo M, Genovese T, Esposito E, Cuzzocrea S. Absence of TLR4 reduces neurovascular unit and secondary inflammatory process after traumatic brain injury in mice. PLoS One. 2013;8(3):e57208. 3. Albert-Weissenberger C, Stetter C, Meuth SG, Gobel K, Bader M, Siren AL, et al. Blocking of bradykinin receptor B1 protects from focal closed head injury in mice by reducing axonal damage and astroglia activation. J Cereb Blood Flow Metab. 2012;32(9):1747– 56. 4. Alder J, Fujioka W, Lifshitz J, Crockett DP, Thakker-Varia S. Lateral fluid percussion: model of traumatic brain injury in mice. J Vis Exp. 2011;(54):3063. 5. Alessandri B, Nishioka T, Heimann A, Bullock RM, Kempski O. Caspase-dependent cell death involved in brain damage after acute subdural hematoma in rats. Brain Res. 2006;1111(1):196–202. 6. Amat JA, Ishiguro H, Nakamura K, Norton WT. Phenotypic diversity and kinetics of proliferating microglia and astrocytes following cortical stab wounds. Glia. 1996;16(4):368–82. 7. Armonda RA, Bell RS, Vo AH, Ling G, DeGraba TJ, Crandall B, et al. Wartime traumatic cerebral vasospasm: recent review of combat casualties. Neurosurgery. 2006;59(6):1215–25. 8. Arun P, bu-Taleb R, Oguntayo S, Wang Y, Valiyaveettil M, Long J, et al. Acute mitochondrial dysfunction after blast exposure: potential role of mitochondrial glutamate oxaloacetate transaminase. J Neurotrauma. 2014; (in press) 9. Arun P, Oguntayo S, Alamneh Y, Honnold C, Wang Y, Valiyaveettil M, et al. Rapid release of tissue enzymes into blood after blast exposure: potential use as biological dosimeters. PLoS One. 2012;7(4):e33798. 10. Bachstetter AD, Rowe RK, Kaneko M, Goulding D, Lifshitz J, Van Eldik LJ. The p38alpha MAPK regulates microglial responsiveness to diffuse traumatic brain injury. J Neurosci. 2013;33(14):6143–53. 11. Baranova AI, Whiting MD, Hamm RJ. Delayed, post-injury treatment with aniracetam improves cognitive performance after traumatic brain injury in rats. J Neurotrauma. 2006;23(8):1233–40. 12. Basso DM, Fisher LC, Anderson AJ, Jakeman LB, McTigue DM, Popovich PG. Basso mouse scale for locomotion detects differences in recovery after spinal cord injury in five common mouse strains. J Neurotrauma. 2006;23(5):635–59. 13. Bate AJ, Mathias JL, Crawford JR. Performance on the test of everyday attention and standard tests of attention following severe traumatic brain injury. Clin Neuropsychol. 2001;15(3):405–22. 14. Bauman RA, Ling G, Tong L, Januszkiewicz A, Agoston D, Delanerolle N, et al. An introductory characterization of a combatcasualty-care relevant swine model of closed head injury resulting from exposure to explosive blast. J Neurotrauma. 2009;26(6):841– 60. 15. Benson MD, Romero MI, Lush ME, Lu QR, Henkemeyer M, Parada LF. Ephrin-B3 is a myelin-based inhibitor of neurite outgrowth. Proc Natl Acad Sci U S A. 2005;102(30):10694–9. 16. Benson RR, Gattu R, Sewick B, Kou Z, Zakariah N, Cavanaugh JM, et al. Detection of hemorrhagic and axonal pathology in mild traumatic brain injury using advanced MRI: implications for neurorehabilitation. NeuroRehabilitation. 2012;31(3):261–79. 17. Bolkvadze T, Pitkanen A. Development of post-traumatic epilepsy after controlled cortical impact and lateral fluid-percussion-induced brain injury in the mouse. J Neurotrauma. 2012;29(5):789–812. 18. Bonnier C, Mesples B, Carpentier S, Henin D, Gressens P. Delayed white matter injury in a murine model of shaken baby syndrome. Brain Pathol. 2002;12(3):320–8.

Transl. Stroke Res. 19. Bonnier C, Mesples B, Gressens P. Animal models of shaken baby syndrome: revisiting the pathophysiology of this devastating injury. Pediatr Rehabil. 2004;7(3):165–71. 20. Bouet V, Boulouard M, Toutain J, Divoux D, Bernaudin M, Schumann-Bard P, et al. The adhesive removal test: a sensitive method to assess sensorimotor deficits in mice. Nat Protoc. 2009;4(10):1560–4. 21. Boulet T, Kelso ML, Othman SF. Long term in vivo imaging of viscoelastic properties of the mouse brain following controlled cortical impact. J Neurotrauma. 2014; (in press) 22. Bramlett HM, Dietrich WD, Green EJ. Secondary hypoxia following moderate fluid percussion brain injury in rats exacerbates sensorimotor and cognitive deficits. J Neurotrauma. 1999;16(11): 1035–47. 23. Brody DL, Holtzman DM. Morris water maze search strategy analysis in PDAPP mice before and after experimental traumatic brain injury. Exp Neurol. 2006;197(2):330–40. 24. Buckingham ET, Daniolos P. Longitudinal outcomes for victims of child abuse. Curr Psychiatr Rep. 2013;15(2):342. 25. Cai W, Carlson SW, Brelsfoard JM, Mannon CE, Moncman CL, Saatman KE, et al. Rit GTPase signaling promotes immature hippocampal neuronal survival. J Neurosci. 2012;32(29):9887–97. 26. Cantu RC, Gean AD. Second-impact syndrome and a small subdural hematoma: an uncommon catastrophic result of repetitive head injury with a characteristic imaging appearance. J Neurotrauma. 2010;27(9):1557–64. 27. Carbonell WS, Maris DO, McCall T, Grady MS. Adaptation of the fluid percussion injury model to the mouse. J Neurotrauma. 1998;15(3):217–29. 28. Carroll CP, Cochran JA, Price JP, Guse CE, Wang MC. The AIS2005 revision in severe traumatic brain injury: mission accomplished or problems for future research? Ann Adv Automot Med. 2010;54:233–8. 29. Cernak I, Merkle AC, Koliatsos VE, Bilik JM, Luong QT, Mahota TM, et al. The pathobiology of blast injuries and blast-induced neurotrauma as identified using a new experimental model of injury in mice. Neurobiol Dis. 2011;41(2):538–51. 30. Chavko M, Watanabe T, Adeeb S, Lankasky J, Ahlers ST, McCarron RM. Relationship between orientation to a blast and pressure wave propagation inside the rat brain. J Neurosci Methods. 2011;195(1):61–6. 31. Chen SF, Tsai HJ, Hung TH, Chen CC, Lee CY, Wu CH, et al. Salidroside improves behavioral and histological outcomes and reduces apoptosis via PI3K/Akt signaling after experimental traumatic brain injury. PLoS One. 2012;7(9):e45763. 32. Chen Y, Constantini S, Trembovler V, Weinstock M, Shohami E. An experimental model of closed head injury in mice: pathophysiology, histopathology, and cognitive deficits. J Neurotrauma. 1996;13(10): 557–68. 33. Chen Y, Huang W. Non-impact, blast-induced mild TBI and PTSD: concepts and caveats. Brain Inj. 2011;25(7–8):641–50. 34. Chen Z, Leung LY, Mountney A, Liao Z, Yang W, Lu XC, et al. A novel animal model of closed-head concussive-induced mild traumatic brain injury: development, implementation, and characterization. J Neurotrauma. 2012;29(2):268–80. 35. Cheng ZG, Zhang GD, Shi PQ, Du BS. Expression and antioxidation of Nrf2/ARE pathway in traumatic brain injury. Asian Pac J Trop Med. 2013;6(4):305–10. 36. Cherian L, Robertson CS, Goodman JC. Secondary insults increase injury after controlled cortical impact in rats. J Neurotrauma. 1996;13(7):371–83. 37. Chrzaszcz M, Venkatesan C, Dragisic T, Watterson DM, Wainwright MS. Minozac treatment prevents increased seizure susceptibility in a mouse “two-hit” model of closed skull traumatic brain injury and electroconvulsive shock-induced seizures. J Neurotrauma. 2010;27(7):1283–95.

38. Clausen F, Hanell A, Bjork M, Hillered L, Mir AK, Gram H, et al. Neutralization of interleukin-1beta modifies the inflammatory response and improves histological and cognitive outcome following traumatic brain injury in mice. Eur J Neurosci. 2009;30(3):385–96. 39. Conte V, Uryu K, Fujimoto S, Yao Y, Rokach J, Longhi L, et al. Vitamin E reduces amyloidosis and improves cognitive function in Tg2576 mice following repetitive concussive brain injury. J Neurochem. 2004;90(3):758–64. 40. Corrigan JD, Selassie AW, Orman JA. The epidemiology of traumatic brain injury. J Head Trauma Rehabil. 2010;25(2):72–80. 41. Courtney MW, Courtney AC. Working toward exposure thresholds for blast-induced traumatic brain injury: thoracic and acceleration mechanisms. Neuroimage. 2011;54 Suppl 1:S55–61. 42. Creeley CE, Wozniak DF, Bayly PV, Olney JW, Lewis LM. Multiple episodes of mild traumatic brain injury result in impaired cognitive performance in mice. Acad Emerg Med. 2004;11(8):809– 19. 43. Cunha MP, Machado DG, Bettio LE, Capra JC, Rodrigues AL. Interaction of zinc with antidepressants in the tail suspension test. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(8):1913–20. 44. Dapul HR, Park J, Zhang J, Lee C, DanEshmand A, Lok J, et al. Concussive injury before or after controlled cortical impact exacerbates histopathology and functional outcome in a mixed traumatic brain injury model in mice. J Neurotrauma. 2013;30(5):382–91. 45. Davidsson J, Risling M. A new model to produce sagittal plane rotational induced diffuse axonal injuries. Front Neurol. 2011;2:41. 46. DeFord SM, Wilson MS, Rice AC, Clausen T, Rice LK, Barabnova A, et al. Repeated mild brain injuries result in cognitive impairment in B6C3F1 mice. J Neurotrauma. 2002;19(4):427–38. 47. Dennis AM, Haselkorn ML, Vagni VA, Garman RH, JaneskoFeldman K, Bayir H, et al. Hemorrhagic shock after experimental traumatic brain injury in mice: effect on neuronal death. J Neurotrauma. 2009;26(6):889–99. 48. DePalma RG, Burris DG, Champion HR, Hodgson MJ. Blast injuries. N Engl J Med. 2005;352(13):1335–42. 49. DeRoss AL, Adams JE, Vane DW, Russell SJ, Terella AM, Wald SL. Multiple head injuries in rats: effects on behavior. J Trauma. 2002;52(4):708–14. 50. Dikranian K, Cohen R, Mac DC, Pan Y, Brakefield D, Bayly P, et al. Mild traumatic brain injury to the infant mouse causes robust white matter axonal degeneration which precedes apoptotic death of cortical and thalamic neurons. Exp Neurol. 2008;211(2):551–60. 51. Dixon CE, Clifton GL, Lighthall JW, Yaghmai AA, Hayes RL. A controlled cortical impact model of traumatic brain injury in the rat. J Neurosci Methods. 1991;39(3):253–62. 52. Dixon CE, Lighthall JW, Anderson TE. Physiologic, histopathologic, and cineradiographic characterization of a new fluid-percussion model of experimental brain injury in the rat. J Neurotrauma. 1988;5(2):91–104. 53. Doering P, Stoltenberg M, Penkowa M, Rungby J, Larsen A, Danscher G. Chemical blocking of zinc ions in CNS increases neuronal damage following traumatic brain injury (TBI) in mice. PLoS One. 2010;5(4):e10131. 54. Dohi K, Ohtaki H, Nakamachi T, Yofu S, Satoh K, Miyamoto K, et al. Gp91phox (NOX2) in classically activated microglia exacerbates traumatic brain injury. J Neuroinflammation. 2010;7:41. 55. Ekmark-Lewen S, Lewen A, Meyerson BJ, Hillered L. The multivariate concentric square field test reveals behavioral profiles of risk taking, exploration, and cognitive impairment in mice subjected to traumatic brain injury. J Neurotrauma. 2010;27(9):1643–55. 56. Elder GA, Dorr NP, De GR, Gama Sosa MA, Shaughness MC, Maudlin-Jeronimo E, et al. Blast exposure induces post-traumatic stress disorder-related traits in a rat model of mild traumatic brain injury. J Neurotrauma. 2012;29(16):2564–75. 57. Eugenin EA, Eckardt D, Theis M, Willecke K, Bennett MV, Saez JC. Microglia at brain stab wounds express connexin 43 and in vitro

Transl. Stroke Res. form functional gap junctions after treatment with interferongamma and tumor necrosis factor-alpha. Proc Natl Acad Sci U S A. 2001;98(7):4190–5. 58. Exo JL, Shellington DK, Bayir H, Vagni VA, Janesco-Feldman K, Ma L, et al. Resuscitation of traumatic brain injury and hemorrhagic shock with polynitroxylated albumin, hextend, hypertonic saline, and lactated Ringer’s: effects on acute hemodynamics, survival, and neuronal death in mice. J Neurotrauma. 2009;26(12):2403–8. 59. Feng JF, Gurkoff GG, Van KC, Song M, Lowe DA, Zhou J, et al. NAAG peptidase inhibitor reduces cellular damage in a model of TBI with secondary hypoxia. Brain Res. 2012;1469:144–52. 60. Flierl MA, Stahel PF, Beauchamp KM, Morgan SJ, Smith WR, Shohami E. Mouse closed head injury model induced by a weightdrop device. Nat Protoc. 2009;4(9):1328–37. 61. Foda MA, Marmarou A. A new model of diffuse brain injury in rats. Part II: morphological characterization. J Neurosurg. 1994;80(2): 301–13. 62. Foley LM, Iqbal O'Meara AM, Wisniewski SR, Kevin HT, Melick JA, Ho C, et al. MRI assessment of cerebral blood flow after experimental traumatic brain injury combined with hemorrhagic shock in mice. J Cereb Blood Flow Metab. 2013;33(1):129–36. 63. Freret T, Bouet V, Leconte C, Roussel S, Chazalviel L, Divoux D, et al. Behavioral deficits after distal focal cerebral ischemia in mice: usefulness of adhesive removal test. Behav Neurosci. 2009;123(1): 224–30. 64. Fujimoto ST, Longhi L, Saatman KE, Conte V, Stocchetti N, McIntosh TK. Motor and cognitive function evaluation following experimental traumatic brain injury. Neurosci Biobehav Rev. 2004;28(4):365–78. 65. Garner J, Brett SJ. Mechanisms of injury by explosive devices. Anesthesiol Clin. 2007;25(1):147–60. x. 66. Gennarelli TA. Animate models of human head injury. J Neurotrauma. 1994;11(4):357–68. 67. Ghajar J. Traumatic brain injury. Lancet. 2000;356(9233):923–9. 68. Gilad R, Boaz M, Sadeh M, Eilam A, Dabby R, Lampl Y. Seizures after very mild head or spine trauma. J Neurotrauma. 2013;30(6): 469–72. 69. Gilmer LK, Roberts KN, Scheff SW. Efficacy of progesterone following a moderate unilateral cortical contusion injury. J Neurotrauma. 2008;25(6):593–602. 70. Giri BK, Krishnappa IK, Bryan Jr RM, Robertson C, Watson J. Regional cerebral blood flow after cortical impact injury complicated by a secondary insult in rats. Stroke. 2000;31(4):961–7. 71. Goldstein LE, Fisher AM, Tagge CA, Zhang XL, Velisek L, Sullivan JA, et al. Chronic traumatic encephalopathy in blastexposed military veterans and a blast neurotrauma mouse model. Sci Transl Med. 2012;4(134):134ra60. 72. Gomes PS, Fernandes MH. Rodent models in bone-related research: the relevance of calvarial defects in the assessment of bone regeneration strategies. Lab Anim. 2011;45(1):14–24. 73. Goodman MD, Makley AT, Huber NL, Clarke CN, Friend LA, Schuster RM, et al. Hypobaric hypoxia exacerbates the neuroinflammatory response to traumatic brain injury. J Surg Res. 2011;165(1):30–7. 74. Hall ED, Bryant YD, Cho W, Sullivan PG. Evolution of posttraumatic neurodegeneration after controlled cortical impact traumatic brain injury in mice and rats as assessed by the de Olmos silver and fluorojade staining methods. J Neurotrauma. 2008;25(3): 235–47. 75. Hamberger A, Viano DC, Saljo A, Bolouri H. Concussion in professional football: morphology of brain injuries in the NFL concussion model—part 16. Neurosurgery. 2009;64(6):1174–82. 76. Hampton DW, Asher RA, Kondo T, Steeves JD, Ramer MS, Fawcett JW. A potential role for bone morphogenetic protein signalling in glial cell fate determination following adult central nervous system injury in vivo. Eur J Neurosci. 2007;26(11):3024–35.

77. Hanell A, Clausen F, Bjork M, Jansson K, Philipson O, Nilsson LN, et al. Genetic deletion and pharmacological inhibition of Nogo-66 receptor impairs cognitive outcome after traumatic brain injury in mice. J Neurotrauma. 2010;27(7):1297–309. 78. Hanell A, Clausen F, Djupsjo A, Vallstedt A, Patra K, Israelsson C, et al. Functional and histological outcome after focal traumatic brain injury is not improved in conditional EphA4 knockout mice. J Neurotrauma. 2012;29(17):2660–71. 79. Hanif S, Abodunde O, Ali Z, Pidgeon C. Age related outcome in acute subdural haematoma following traumatic head injury. Ir Med J. 2009;102(8):255–7. 80. Haselkorn ML, Shellington DK, Jackson EK, Vagni VA, JaneskoFeldman K, Dubey RK, et al. Adenosine A1 receptor activation as a brake on the microglial response after experimental traumatic brain injury in mice. J Neurotrauma. 2010;27(5):901–10. 81. Hemerka JN, Wu X, Dixon CE, Garman RH, Exo JL, Shellington DK, et al. Severe brief pressure-controlled hemorrhagic shock after traumatic brain injury exacerbates functional deficits and long-term neuropathological damage in mice. J Neurotrauma. 2012;29(12): 2192–208. 82. Higgens LS, Unterharnscheidt F. Pathomorphology of experimental head injury due to rotational acceleration. Acta Neuropathol. 1969;12(2):200–4. 83. Hong Y, Yan W, Chen S, Sun CR, Zhang JM. The role of Nrf2 signaling in the regulation of antioxidants and detoxifying enzymes after traumatic brain injury in rats and mice. Acta Pharmacol Sin. 2010;31(11):1421–30. 84. Horvat A, Schwaiger F, Hager G, Brocker F, Streif R, Knyazev P, et al. A novel role for protein tyrosine phosphatase shp1 in controlling glial activation in the normal and injured nervous system. J Neurosci. 2001;21(3):865–74. 85. Huang WC, Kuo WC, Cherng JH, Hsu SH, Chen PR, Huang SH, et al. Chondroitinase ABC promotes axonal re-growth and behavior recovery in spinal cord injury. Biochem Biophys Res Commun. 2006;349(3):963–8. 86. Hunt RF, Scheff SW, Smith BN. Posttraumatic epilepsy after controlled cortical impact injury in mice. Exp Neurol. 2009;215(2): 243–52. 87. Hylin MJ, Orsi SA, Rozas NS, Hill JL, Zhao J, Redell JB, et al. Repeated mild closed head injury impairs short-term visuospatial memory and complex learning. J Neurotrauma. 2013;30(9):716–26. 88. Indraswari F, Wang H, Lei B, James ML, Kernagis D, Warner DS, et al. Statins improve outcome in murine models of intracranial hemorrhage and traumatic brain injury: a translational approach. J Neurotrauma. 2012;29(7):1388–400. 89. Ishige N, Pitts LH, Berry I, Carlson SG, Nishimura MC, Moseley ME, et al. The effect of hypoxia on traumatic head injury in rats: alterations in neurologic function, brain edema, and cerebral blood flow. J Cereb Blood Flow Metab. 1987;7(6):759–67. 90. James ML, Wang H, Venkatraman T, Song P, Lascola CD, Laskowitz DT. Brain natriuretic peptide improves long-term functional recovery after acute CNS injury in mice. J Neurotrauma. 2010;27(1):217–28. 91. Jenkins LW, Moszynski K, Lyeth BG, Lewelt W, DeWitt DS, Allen A, et al. Increased vulnerability of the mildly traumatized rat brain to cerebral ischemia: the use of controlled secondary ischemia as a research tool to identify common or different mechanisms contributing to mechanical and ischemic brain injury. Brain Res. 1989;477(1–2):211–24. 92. Jordan BD. Brain injury in boxing. Clin Sports Med. 2009;28(4): 561–78. vi. 93. Kabadi SV, Stoica BA, Loane DJ, Byrnes KR, Hanscom M, Cabatbat RM, et al. Cyclin D1 gene ablation confers neuroprotection in traumatic brain injury. J Neurotrauma. 2012;29(5):813–27. 94. Kamnaksh A, Kwon SK, Kovesdi E, Ahmed F, Barry ES, Grunberg NE, et al. Neurobehavioral, cellular, and molecular consequences of

Transl. Stroke Res. single and multiple mild blast exposure. Electrophoresis. 2012;33(24):3680–92. 95. Kane MJ, ngoa-Perez M, Briggs DI, Viano DC, Kreipke CW, Kuhn DM. A mouse model of human repetitive mild traumatic brain injury. J Neurosci Methods. 2012;203(1):41–9. 96. Katada R, Nishitani Y, Honmou O, Mizuo K, Okazaki S, Tateda K, et al. Expression of aquaporin-4 augments cytotoxic brain edema after traumatic brain injury during acute ethanol exposure. Am J Pathol. 2012;180(1):17–23. 97. Kataoka K, Asai T, Taneda M, Ueshima S, Matsuo O, Kuroda R, et al. Roles of urokinase type plasminogen activator in a brain stab wound. Brain Res. 2000;887(1):187–90. 98. Kelso ML, Scheff SW, Pauly JR, Loftin CD. Effects of genetic deficiency of cyclooxygenase-1 or cyclooxygenase-2 on functional and histological outcomes following traumatic brain injury in mice. BMC Neurosci. 2009;10:108. 99. Kharatishvili I, Pitkanen A. Posttraumatic epilepsy. Curr Opin Neurol. 2010;23(2):183–8. 100. Kilbourne M, Kuehn R, Tosun C, Caridi J, Keledjian K, Bochicchio G, et al. Novel model of frontal impact closed head injury in the rat. J Neurotrauma. 2009;26(12):2233–43. 101. Kim JY, Kim N, Zheng Z, Lee JE, Yenari MA. The 70 kDa heat shock protein protects against experimental traumatic brain injury. Neurobiol Dis. 2013;58:289–95. 102. Kimbler DE, Shields J, Yanasak N, Vender JR, Dhandapani KM. Activation of P2X7 promotes cerebral edema and neurological injury after traumatic brain injury in mice. PLoS One. 2012;7(7): e41229. 103. Kipnis J, Nevo U, Panikashvili D, Alexandrovich A, Yoles E, Akselrod S, et al. Therapeutic vaccination for closed head injury. J Neurotrauma. 2003;20(6):559–69. 104. Kochanek PM, Vagni VA, Janesko KL, Washington CB, Crumrine PK, Garman RH, et al. Adenosine A1 receptor knockout mice develop lethal status epilepticus after experimental traumatic brain injury. J Cereb Blood Flow Metab. 2006;26(4):565–75. 105. Koliatsos VE, Cernak I, Xu L, Song Y, Savonenko A, Crain BJ, et al. A mouse model of blast injury to brain: initial pathological, neuropathological, and behavioral characterization. J Neuropathol Exp Neurol. 2011;70(5):399–416. 106. Kotapka MJ, Gennarelli TA, Graham DI, Adams JH, Thibault LE, Ross DT, et al. Selective vulnerability of hippocampal neurons in acceleration-induced experimental head injury. J Neurotrauma. 1991;8(4):247–58. 107. Krajewska M, You Z, Rong J, Kress C, Huang X, Yang J, et al. Neuronal deletion of caspase 8 protects against brain injury in mouse models of controlled cortical impact and kainic acidinduced excitotoxicity. PLoS One. 2011;6(9):e24341. 108. Kuehn R, Simard PF, Driscoll I, Keledjian K, Ivanova S, Tosun C, et al. Rodent model of direct cranial blast injury. J Neurotrauma. 2011;28(10):2155–69. 109. Lammie GA, Piper IR, Thomson D, Brannan F. Neuropathologic characterization of a rodent model of closed head injury– addition of clinically relevant secondary insults does not significantly potentiate brain damage. J Neurotrauma. 1999;16(7): 603–15. 110. Learoyd AE, Lifshitz J. Comparison of rat sensory behavioral tasks to detect somatosensory morbidity after diffuse brain-injury. Behav Brain Res. 2012;226(1):197–204. 111. Leinhase I, Rozanski M, Harhausen D, Thurman JM, Schmidt OI, Hossini AM, et al. Inhibition of the alternative complement activation pathway in traumatic brain injury by a monoclonal anti-factor B antibody: a randomized placebo-controlled study in mice. J Neuroinflammation. 2007;4:13. 112. Leitgeb J, Mauritz W, Brazinova A, Janciak I, Majdan M, Wilbacher I, et al. Outcome after severe brain trauma due to acute subdural hematoma. J Neurosurg. 2012;117(2):324–33.

113. Leitgeb J, Mauritz W, Brazinova A, Majdan M, Wilbacher I. Outcome after severe brain trauma associated with epidural hematoma. Arch Orthop Trauma Surg. 2013;133(2):199–207. 114. Levin HS, Robertson CS. Mild traumatic brain injury in translation. J Neurotrauma. 2013;30(8):610–7. 115. Lew HL, Poole JH, Alvarez S, Moore W. Soldiers with occult traumatic brain injury. Am J Phys Med Rehabil. 2005;84(6):393–8. 116. Ley EJ, Clond MA, Bukur M, Park R, Chervonski M, Dagliyan G, et al. beta-adrenergic receptor inhibition affects cerebral glucose metabolism, motor performance, and inflammatory response after traumatic brain injury. J Trauma Acute Care Surg. 2012;73(1):33–40. 117. Ley EJ, Clond MA, Singer MB, Shouhed D, Salim A. IL6 deficiency affects function after traumatic brain injury. J Surg Res. 2011;170(2):253–6. 118. Ley EJ, Scehnet J, Park R, Schroff S, Dagliyan G, Conti PS, et al. The in vivo effect of propranolol on cerebral perfusion and hypoxia after traumatic brain injury. J Trauma. 2009;66(1):154–9. 119. Lifshitz J, Witgen BM, Grady MS. Acute cognitive impairment after lateral fluid percussion brain injury recovers by 1 month: evaluation by conditioned fear response. Behav Brain Res. 2007;177(2):347– 57. 120. Lighthall JW, Dixon CE, Anderson TE. Experimental models of brain injury. J Neurotrauma. 1989;6(2):83–97. 121. Liu NK, Zhang YP, O’Connor J, Gianaris A, Oakes E, Lu QB, et al. A bilateral head injury that shows graded brain damage and behavioral deficits in adult mice. Brain Res. 2013;1499:121–8. 122. Long JB, Bentley TL, Wessner KA, Cerone C, Sweeney S, Bauman RA. Blast overpressure in rats: recreating a battlefield injury in the laboratory. J Neurotrauma. 2009;26(6):827–40. 123. Longhi L, Ortolano F, Zanier ER, Perego C, Stocchetti N, De Simoni MG. Effect of traumatic brain injury on cognitive function in mice lacking p55 and p75 tumor necrosis factor receptors. Acta Neurochir Suppl. 2008;102:409–13. 124. Longhi L, Perego C, Ortolano F, Aresi S, Fumagalli S, Zanier ER, et al. Tumor necrosis factor in traumatic brain injury: effects of genetic deletion of p55 or p75 receptor. J Cereb Blood Flow Metab. 2013;33(8):1182–9. 125. Longhi L, Saatman KE, Fujimoto S, Raghupathi R, Meaney DF, Davis J, et al. Temporal window of vulnerability to repetitive experimental concussive brain injury. Neurosurgery. 2005;56(2): 364–74. 126. Longhi L, Saatman KE, Raghupathi R, Laurer HL, Lenzlinger PM, Riess P, et al. A review and rationale for the use of genetically engineered animals in the study of traumatic brain injury. J Cereb Blood Flow Metab. 2001;21(11):1241–58. 127. Longhi L, Watson DJ, Saatman KE, Thompson HJ, Zhang C, Fujimoto S, et al. Ex vivo gene therapy using targeted engraftment of NGF-expressing human NT2N neurons attenuates cognitive deficits following traumatic brain injury in mice. J Neurotrauma. 2004;21(12):1723–36. 128. Maier EY, Ma ST, Ahrens A, Schallert TJ, Duvauchelle CL. Assessment of ultrasonic vocalizations during drug selfadministration in rats. J Vis Exp. 2010;(41):2014. 129. Mannix RC, Zhang J, Park J, Lee C, Whalen MJ. Detrimental effect of genetic inhibition of B-site APP-cleaving enzyme 1 on functional outcome after controlled cortical impact in young adult mice. J Neurotrauma. 2011;28(9):1855–61. 130. Manvelyan H. Contemporary experimental models of traumatic brain injury. Georgian Med News. 2006;140:13–6. 131. Marciano D, Shohami E, Kloog Y, Alexandrovitch A, Brandeis R, Goelman G. Neuroprotective effects of the Ras inhibitor S-transtrans-farnesylthiosalicylic acid, measured by diffusion-weighted imaging after traumatic brain injury in rats. J Neurotrauma. 2007;24(8):1378–86. 132. Margulies SS, Thibault LE, Gennarelli TA. Physical model simulations of brain injury in the primate. J Biomech. 1990;23(8):823–36.

Transl. Stroke Res. 133. Marklund N, Hillered L. Animal modelling of traumatic brain injury in preclinical drug development: where do we go from here? Br J Pharmacol. 2011;164(4):1207–29. 134. Marmarou A, Foda MA, van den Brink W, Campbell J, Kita H, Demetriadou K. A new model of diffuse brain injury in rats. Part I: pathophysiology and biomechanics. J Neurosurg. 1994;80(2):291– 300. 135. Mascia L. Acute lung injury in patients with severe brain injury: a double hit model. Neurocrit Care. 2009;11(3):417–26. 136. Mathias JL, Beall JA, Bigler ED. Neuropsychological and information processing deficits following mild traumatic brain injury. J Int Neuropsychol Soc. 2004;10(2):286–97. 137. Matsushita Y, Bramlett HM, Alonso O, Dietrich WD. Posttraumatic hypothermia is neuroprotective in a model of traumatic brain injury complicated by a secondary hypoxic insult. Crit Care Med. 2001;29(11):2060–6. 138. Mayer SA, Badjatia N. Head injury. In: Rowland LP, Pedley TA, editors. Merritt’s neurology. Philadelphia: Wolters Kluwer Lippincott Williams & Wilkins; 2010. p. 479–94. 139. Mayoral SR, Omar G, Penn AA. Sex differences in a hypoxia model of preterm brain damage. Pediatr Res. 2009;66(3):248–53. 140. Mbye LH, Singh IN, Carrico KM, Saatman KE, Hall ED. Comparative neuroprotective effects of cyclosporin A and NIM811, a nonimmunosuppressive cyclosporin A analog, following traumatic brain injury. J Cereb Blood Flow Metab. 2009;29(1): 87–97. 141. McBeth BD, Stern SA, Wang X, Mertz M, Zink BJ. Effects of cocaine in an experimental model of traumatic brain injury. Acad Emerg Med. 2005;12(6):483–90. 142. McCrory P, Davis G, Makdissi M. Second impact syndrome or cerebral swelling after sporting head injury. Curr Sports Med Rep. 2012;11(1):21–3. 143. McKee AC, Stein TD, Nowinski CJ, Stern RA, Daneshvar DH, Alvarez VE, et al. The spectrum of disease in chronic traumatic encephalopathy. Brain. 2013;136(Pt 1):43–64. 144. McNamara KC, Lisembee AM, Lifshitz J. The whisker nuisance task identifies a late-onset, persistent sensory sensitivity in diffuse brain-injured rats. J Neurotrauma. 2010;27(4):695–706. 145. Meehan III WP, Zhang J, Mannix R, Whalen MJ. Increasing recovery time between injuries improves cognitive outcome after repetitive mild concussive brain injuries in mice. Neurosurgery. 2012;71(4):885–91. 146. Mikrogianakis A, Shaye RE, Griffin P, Kawesa S, Lockwood J, Gendron NH, et al. Hypoxia alters the expression of inhibitor of apoptosis proteins after brain trauma in the mouse. J Neurotrauma. 2007;24(2):338–53. 147. Miller G. Neuropathology. Blast injuries linked to neurodegeneration in veterans. Science. 2012;336(6083):790–1. 148. Milman A, Rosenberg A, Weizman R, Pick CG. Mild traumatic brain injury induces persistent cognitive deficits and behavioral disturbances in mice. J Neurotrauma. 2005;22(9):1003–10. 149. Milman A, Zohar O, Maayan R, Weizman R, Pick CG. DHEAS repeated treatment improves cognitive and behavioral deficits after mild traumatic brain injury. Eur Neuropsychopharmacol. 2008;18(3):181–7. 150. Mirzayan MJ, Probst C, Samii M, Krettek C, Gharabaghi A, Pape HC, et al. Histopathological features of the brain, liver, kidney and spleen following an innovative polytrauma model of the mouse. Exp Toxicol Pathol. 2012;64(3):133–9. 151. Mountney A, Leung LY, Pedersen R, Shear D, Tortella F. Longitudinal assessment of gait abnormalities following penetrating ballistic-like brain injury in rats. J Neurosci Methods. 2012;212(1):1–16. 152. Nadler Y, Alexandrovich A, Grigoriadis N, Hartmann T, Rao KS, Shohami E, et al. Increased expression of the gamma-secretase components presenilin-1 and nicastrin in activated astrocytes and

microglia following traumatic brain injury. Glia. 2008;56(5):552– 67. 153. Nakagawa A, Manley GT, Gean AD, Ohtani K, Armonda R, Tsukamoto A, et al. Mechanisms of primary blast-induced traumatic brain injury: insights from shock-wave research. J Neurotrauma. 2011;28(6):1101–19. 154. Neumann M, Wang Y, Kim S, Hong SM, Jeng L, Bilgen M, et al. Assessing gait impairment following experimental traumatic brain injury in mice. J Neurosci Methods. 2009;176(1):34–44. 155. Ning YL, Yang N, Chen X, Xiong RP, Zhang XZ, Li P, et al. Adenosine A2A receptor deficiency alleviates blast-induced cognitive dysfunction. J Cereb Blood Flow Metab. 2013;33(11):1789– 98. 156. Oliva Jr AA, Kang Y, Sanchez-Molano J, Furones C, Atkins CM. STAT3 signaling after traumatic brain injury. J Neurochem. 2012;120(5):710–20. 157. Ommaya AK, Corrao P, Letcher FS. Head injury in the chimpanzee. 1. Biodynamics of traumatic unconsciousness. J Neurosurg. 1973;39(2):152–66. 158. Ommaya AK, Goldsmith W, Thibault L. Biomechanics and neuropathology of adult and paediatric head injury. Br J Neurosurg. 2002;16(3):220–42. 159. Oron A, Oron U, Streeter J, de Taboada L, Alexandrovich A, Trembovler V, et al. Low-level laser therapy applied transcranially to mice following traumatic brain injury significantly reduces longterm neurological deficits. J Neurotrauma. 2007;24(4):651–6. 160. Paiva WS, Soares MS, Amorim RL, de Andrade AF, Matushita H, Teixeira MJ. Traumatic brain injury and shaken baby syndrome. Acta Med Port. 2011;24(5):805–8. 161. Parkinson D. Evaluating cerebral concussion. Surg Neurol. 1996;45(5):459–62. 162. Peters M, Trembovler V, Alexandrovich A, Parnas M, Birnbaumer L, Minke B, et al. Carvacrol together with TRPC1 elimination improve functional recovery after traumatic brain injury in mice. J Neurotrauma. 2012;29(18):2831–4. 163. Petraglia AL, Marky AH, Walker C, Thiyagarajan M, Zlokovic BV. Activated protein C is neuroprotective and mediates new blood vessel formation and neurogenesis after controlled cortical impact. Neurosurgery. 2010;66(1):165–71. 164. Pinna G. In a mouse model relevant for post-traumatic stress disorder, selective brain steroidogenic stimulants (SBSS) improve behavioral deficits by normalizing allopregnanolone biosynthesis. Behav Pharmacol. 2010;21(5–6):438–50. 165. Plantman S, Ng KC, Lu J, Davidsson J, Risling M. Characterization of a novel rat model of penetrating traumatic brain injury. J Neurotrauma. 2012;29(6):1219–32. 166. Pleasant JM, Carlson SW, Mao H, Scheff SW, Yang KH, Saatman KE. Rate of neurodegeneration in the mouse controlled cortical impact model is influenced by impactor tip shape: implications for mechanistic and therapeutic studies. J Neurotrauma. 2011;28(11): 2245–62. 167. Povlishock JT. The window of risk in repeated head injury. J Neurotrauma. 2013;30(1):1. 168. Prins ML, Alexander D, Giza CC, Hovda DA. Repeated mild traumatic brain injury: mechanisms of cerebral vulnerability. J Neurotrauma. 2013;30(1):30–8. 169. Probst C, Mirzayan MJ, Mommsen P, Zeckey C, Tegeder T, Geerken L, et al. Systemic inflammatory effects of traumatic brain injury, femur fracture, and shock: an experimental murine polytrauma model. Mediat Inflamm. 2012;2012:136020. 170. Pun PB, Kan EM, Salim A, Li Z, Ng KC, Moochhala SM, et al. Low level primary blast injury in rodent brain. Front Neurol. 2011;2:19. 171. Reneer DV, Hisel RD, Hoffman JM, Kryscio RJ, Lusk BT, Geddes JW. A multi-mode shock tube for investigation of blast-induced traumatic brain injury. J Neurotrauma. 2011;28(1):95–104.

Transl. Stroke Res. 172. Reshef A, Shirvan A, Shohami E, Grimberg H, Levin G, Cohen A, et al. Targeting cell death in vivo in experimental traumatic brain injury by a novel molecular probe. J Neurotrauma. 2008;25(6):569–80. 173. Risling M, Plantman S, Angeria M, Rostami E, Bellander BM, Kirkegaard M, et al. Mechanisms of blast induced brain injuries, experimental studies in rats. Neuroimage. 2011;54 Suppl 1:S89–97. 174. Rostami E, Davidsson J, Ng KC, Lu J, Gyorgy A, Walker J, et al. A model for mild traumatic brain injury that induces limited transient memory impairment and increased levels of axon related serum biomarkers. Front Neurol. 2012;3:115. 175. Rubovitch V, Ten-Bosch M, Zohar O, Harrison CR, Tempel-Brami C, Stein E, et al. A mouse model of blast-induced mild traumatic brain injury. Exp Neurol. 2011;232(2):280–9. 176. Ryan CG, Thompson RE, Temkin NR, Crane PK, Ellenbogen RG, Elmore JG. Acute traumatic subdural hematoma: current mortality and functional outcomes in adult patients at a Level I trauma center. J Trauma Acute Care Surg. 2012;73(5):1348–54. 177. Saatman KE, Feeko KJ, Pape RL, Raghupathi R. Differential behavioral and histopathological responses to graded cortical impact injury in mice. J Neurotrauma. 2006;23(8):1241–53. 178. Salehi-Had H, Brandt JD, Rosas AJ, Rogers KK. Findings in older children with abusive head injury: does shaken-child syndrome exist? Pediatrics. 2006;117(5):e1039–44. 179. Sasaki M, Dunn L. A model of acute subdural hematoma in the mouse. J Neurotrauma. 2001;18(11):1241–6. 180. Sawauchi S, Marmarou A, Beaumont A, Signoretti S, Fukui S. Acute subdural hematoma associated with diffuse brain injury and hypoxemia in the rat: effect of surgical evacuation of the hematoma. J Neurotrauma. 2004;21(5):563–73. 181. Schumann J, Alexandrovich GA, Biegon A, Yaka R. Inhibition of NR2B phosphorylation restores alterations in NMDA receptor expression and improves functional recovery following traumatic brain injury in mice. J Neurotrauma. 2008;25(8):945–57. 182. Schwarzbold ML, Rial D, De BT, Machado DG, Cunha MP, dos Santos AA, et al. Effects of traumatic brain injury of different severities on emotional, cognitive, and oxidative stress-related parameters in mice. J Neurotrauma. 2010;27(10):1883–93. 183. Serbanescu I, Brown SM, Ramsay D, Levin AV. Natural animal shaking: a model for non-accidental head injury in children? Eye (Lond). 2008;22(5):715–7. 184. Shellington DK, Du L, Wu X, Exo J, Vagni V, Ma L, et al. Polynitroxylated pegylated hemoglobin: a novel neuroprotective hemoglobin for acute volume-limited fluid resuscitation after combined traumatic brain injury and hemorrhagic hypotension in mice. Crit Care Med. 2011;39(3):494–505. 185. Shih AY, Mateo C, Drew PJ, Tsai PS, Kleinfeld D. A polished and reinforced thinned-skull window for long-term imaging of the mouse brain. J Vis Exp. 2012;(61). 186. Shitaka Y, Tran HT, Bennett RE, Sanchez L, Levy MA, Dikranian K, et al. Repetitive closed-skull traumatic brain injury in mice causes persistent multifocal axonal injury and microglial reactivity. J Neuropathol Exp Neurol. 2011;70(7):551–67. 187. Shohami E, Yatsiv I, Alexandrovich A, Haklai R, Elad-Sfadia G, Grossman R, et al. The Ras inhibitor S-trans, transfarnesylthiosalicylic acid exerts long-lasting neuroprotection in a mouse closed head injury model. J Cereb Blood Flow Metab. 2003;23(6):728–38. 188. Shultz SR, Bao F, Omana V, Chiu C, Brown A, Cain DP. Repeated mild lateral fluid percussion brain injury in the rat causes cumulative long-term behavioral impairments, neuroinflammation, and cortical loss in an animal model of repeated concussion. J Neurotrauma. 2012;29(2):281–94. 189. Smith DH, Soares HD, Pierce JS, Perlman KG, Saatman KE, Meaney DF, et al. A model of parasagittal controlled cortical impact in the mouse: cognitive and histopathologic effects. J Neurotrauma. 1995;12(2):169–78.

190. Spain A, Daumas S, Lifshitz J, Rhodes J, Andrews PJ, Horsburgh K, et al. Mild fluid percussion injury in mice produces evolving selective axonal pathology and cognitive deficits relevant to human brain injury. J Neurotrauma. 2010;27(8):1429–38. 191. Squier W. The “shaken baby” syndrome: pathology and mechanisms. Acta Neuropathol. 2011;122(5):519–42. 192. Stiefel MF, Tomita Y, Marmarou A. Secondary ischemia impairing the restoration of ion homeostasis following traumatic brain injury. J Neurosurg. 2005;103(4):707–14. 193. Strauss KI, Gruzdev A, Zeldin DC. Altered behavioral phenotypes in soluble epoxide hydrolase knockout mice: effects of traumatic brain injury. Prostaglandins Lipid Mediat. 2013;104–105:18–24. 194. Sundaramurthy A, Alai A, Ganpule S, Holmberg A, Plougonven E, Chandra N. Blast-induced biomechanical loading of the rat: an experimental and anatomically accurate computational blast injury model. J Neurotrauma. 2012;29(13):2352–64. 195. Sword J, Masuda T, Croom D, Kirov SA. Evolution of neuronal and astroglial disruption in the peri-contusional cortex of mice revealed by in vivo two-photon imaging. Brain. 2013;136(Pt 5):1446–61. 196. Taber KH, Warden DL, Hurley RA. Blast-related traumatic brain injury: what is known? J Neuropsychiatry Clin Neurosci. 2006;18(2):141–5. 197. Tanaka Y, Matsuwaki T, Yamanouchi K, Nishihara M. Exacerbated inflammatory responses related to activated microglia after traumatic brain injury in progranulin-deficient mice. Neuroscience. 2013;231:49–60. 198. Taussky P, Widmer HR, Takala J, Fandino J. Outcome after acute traumatic subdural and epidural haematoma in Switzerland: a single-centre experience. Swiss Med Wkly. 2008;138(19–20): 281–5. 199. Theriault M, De BL, Tremblay S, Lassonde M, Jolicoeur P. Cumulative effects of concussions in athletes revealed by electrophysiological abnormalities on visual working memory. J Clin Exp Neuropsychol. 2011;33(1):30–41. 200. Theus MH, Ricard J, Bethea JR, Liebl DJ. EphB3 limits the expansion of neural progenitor cells in the subventricular zone by regulating p53 during homeostasis and following traumatic brain injury. Stem Cells. 2010;28(7):1231–42. 201. Thurman DJ, Alverson C, Dunn KA, Guerrero J, Sniezek JE. Traumatic brain injury in the United States: a public health perspective. J Head Trauma Rehabil. 1999;14(6):602–15. 202. Tian DS, Yu ZY, Xie MJ, Bu BT, Witte OW, Wang W. Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal cord injury rat model by the cell cycle inhibitor olomoucine. J Neurosci Res. 2006;84(5): 1053–63. 203. Tomasevic G, Laurer HL, Mattiasson G, van Steeg H, Wieloch T, McIntosh TK. Delayed neuromotor recovery and increased memory acquisition dysfunction following experimental brain trauma in mice lacking the DNA repair gene XPA. J Neurosurg. 2012;116(6):1368–78. 204. Trabold R, Eros C, Zweckberger K, Relton J, Beck H, Nussberger J, et al. The role of bradykinin B(1) and B(2) receptors for secondary brain damage after traumatic brain injury in mice. J Cereb Blood Flow Metab. 2010;30(1):130–9. 205. Tsenter J, Beni-Adani L, Assaf Y, Alexandrovich AG, Trembovler V, Shohami E. Dynamic changes in the recovery after traumatic brain injury in mice: effect of injury severity on T2-weighted MRI abnormalities, and motor and cognitive functions. J Neurotrauma. 2008;25(4):324–33. 206. Valiyaveettil M, Alamneh Y, Wang Y, Arun P, Oguntayo S, Wei Y, et al. Contribution of systemic factors in the pathophysiology of repeated blast-induced neurotrauma. Neurosci Lett. 2013;539:1–6. 207. Viano DC, Hamberger A, Bolouri H, Saljo A. Concussion in professional football: animal model of brain injury—part 15. Neurosurgery. 2009;64(6):1162–73.

Transl. Stroke Res. 208. Vinogradov AE. Human more complex than mouse at cellular level. PLoS One. 2012;7(7):e41753. 209. von Baumgarten L, Trabold R, Thal S, Back T, Plesnila N. Role of cortical spreading depressions for secondary brain damage after traumatic brain injury in mice. J Cereb Blood Flow Metab. 2008;28(7):1353–60. 210. Walker KR, Kang EL, Whalen MJ, Shen Y, Tesco G. Depletion of GGA1 and GGA3 mediates postinjury elevation of BACE1. J Neurosci. 2012;32(30):10423–37. 211. Wang HC, Duan ZX, Wu FF, Xie L, Zhang H, Ma YB. A new rat model for diffuse axonal injury using a combination of linear acceleration and angular acceleration. J Neurotrauma. 2010;27(4): 707–19. 212. Wang Y, Arun P, Wei Y, Oguntayo S, Gharavi RB, Valiyaveettil M, et al. Repeated Blast Exposures cause DNA Fragmentation in Mice. J Neurotrauma. 2014; (in press) 213. Wang Y, Wei Y, Oguntayo S, Wilkins W, Arun P, Valiyaveettil M, et al. Tightly coupled repetitive blast-induced traumatic brain injury: development and characterization in mice. J Neurotrauma. 2011;28(10):2171–83. 214. Warden D. Military TBI, during the Iraq and Afghanistan wars. J Head Trauma Rehabil. 2006;21(5):398–402. 215. Wei HH, Lu XC, Shear DA, Waghray A, Yao C, Tortella FC, et al. NNZ-2566 treatment inhibits neuroinflammation and proinflammatory cytokine expression induced by experimental penetrating ballistic-like brain injury in rats. J Neuroinflammation. 2009;6:19. 216. Williams AJ, Hartings JA, Lu XC, Rolli ML, Tortella FC. Penetrating ballistic-like brain injury in the rat: differential time courses of hemorrhage, cell death, inflammation, and remote degeneration. J Neurotrauma. 2006;23(12):1828–46. 217. Williams AJ, Ling GS, Tortella FC. Severity level and injury track determine outcome following a penetrating ballistic-like brain injury in the rat. Neurosci Lett. 2006;408(3):183–8. 218. Williams AJ, Wei HH, Dave JR, Tortella FC. Acute and delayed neuroinflammatory response following experimental penetrating ballistic brain injury in the rat. J Neuroinflammation. 2007;4:17.

219. Yoneyama-Sarnecky T, Olivas AD, Azari S, Ferriero DM, Manvelyan HM, Noble-Haeusslein LJ. Heme oxygenase-2 modulates early pathogenesis after traumatic injury to the immature brain. Dev Neurosci. 2010;32(1):81–90. 220. Yoshiyama Y, Uryu K, Higuchi M, Longhi L, Hoover R, Fujimoto S, et al. Enhanced neurofibrillary tangle formation, cerebral atrophy, and cognitive deficits induced by repetitive mild brain injury in a transgenic tauopathy mouse model. J Neurotrauma. 2005;22(10): 1134–41. 221. Yu S, Kaneko Y, Bae E, Stahl CE, Wang Y, van Loveren H, et al. Severity of controlled cortical impact traumatic brain injury in rats and mice dictates degree of behavioral deficits. Brain Res. 2009;1287:157–63. 222. Zaltzman R, Beni SM, Giladi E, Pinhasov A, Steingart RA, Romano J, et al. Injections of the neuroprotective peptide NAP to newborn mice attenuate head-injury-related dysfunction in adults. Neuroreport. 2003;14(3):481–4. 223. Zhao Z, Loane DJ, Murray MG, Stoica BA, Faden AI. Comparing the predictive value of multiple cognitive, affective, and motor tasks after rodent traumatic brain injury. J Neurotrauma. 2012;29(15):2475–89. 224. Zink BJ, Stern SA, McBeth BD, Wang X, Mertz M. Effects of ethanol on limited resuscitation in a model of traumatic brain injury and hemorrhagic shock. J Neurosurg. 2006;105(6):884–93. 225. Zohar O, Rubovitch V, Milman A, Schreiber S, Pick CG. Behavioral consequences of minimal traumatic brain injury in mice. Acta Neurobiol Exp (Wars). 2011;71(1):36–45. 226. Zomkowski AD, Hammes L, Lin J, Calixto JB, Santos AR, Rodrigues AL. Agmatine produces antidepressant-like effects in two models of depression in mice. Neuroreport. 2002;13(4):387–91. 227. Zweckberger K, Eros C, Zimmermann R, Kim SW, Engel D, Plesnila N. Effect of early and delayed decompressive craniectomy on secondary brain damage after controlled cortical impact in mice. J Neurotrauma. 2006;23(7):1083–93. 228. Zweckberger K, Stoffel M, Baethmann A, Plesnila N. Effect of decompression craniotomy on increase of contusion volume and functional outcome after controlled cortical impact in mice. J Neurotrauma. 2003;20(12):1307–14.

Traumatic brain injury using mouse models.

The use of mouse models in traumatic brain injury (TBI) has several advantages compared to other animal models including low cost of breeding, easy ma...
545KB Sizes 1 Downloads 0 Views