You are on page 1of 14

Experimental Neurology 275 (2016) 436–449

Contents lists available at ScienceDirect

Experimental Neurology
journal homepage: www.elsevier.com/locate/yexnr

Repetitive mild traumatic brain injury with impact acceleration in the


mouse: Multifocal axonopathy, neuroinflammation, and
neurodegeneration in the visual system
Leyan Xu a,⁎, Judy V. Nguyen b, Mohamed Lehar c, Adarsh Menon a, Elizabeth Rha a, John Arena a, Jiwon Ryu a,
Nicholas Marsh-Armstrong b,d, Christina R. Marmarou e, Vassilis E. Koliatsos a,f,g
a
Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
b
Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
c
Department of Otolaryngology-HNS, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
d
Hugo W. Moser Research Institute at Kennedy Krieger, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
e
Department of Neurosurgery, Virginia Commonwealth University, Richmond, VA 23298, USA
f
Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
g
Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA

a r t i c l e i n f o a b s t r a c t

Article history: Repetitive mild traumatic brain injury (mTBI) is implicated in chronic neurological illness. The development of
Received 6 August 2014 animal models of repetitive mTBI in mice is essential for exploring mechanisms of these chronic diseases, includ-
Revised 29 October 2014 ing genetic vulnerability by using transgenic backgrounds. In this study, the rat model of impact acceleration (IA)
Accepted 4 November 2014
was redesigned for the mouse cranium and used in two clinically relevant repetitive mTBI paradigms. We first de-
Available online 20 November 2014
termined, by using increments of weight dropped from 1 m that the 40 g weight was most representative of mTBI
Keywords:
and was not associated with fractures, brain contusions, anoxic–ischemic injury, mortality, or significant neuro-
Traumatic axonal injury logical impairments. Quantitative evaluation of traumatic axonal injury (TAI) in the optic nerve/tract, cerebellum
Diffuse axonal injury and corpus callosum confirmed that weight increase produced a graded injury. We next evaluated two novel re-
Concussion petitive mTBI paradigms (1 time per day or 3 times per day at days 0, 1, 3, and 7) and compared the resulting TAI,
CTE neuronal cell death, and neuroinflammation to single hit mTBI at sub-acute (7 days) and chronic time points (10
Retinal ganglion cell weeks) post-injury. Both single and repetitive mTBI caused TAI in the optic nerve/tract, cerebellum, corticospinal
Optic nerve tract, lateral lemniscus and corpus callosum. Reactive microglia with phagocytic phenotypes were present at in-
Tau
jury sites. Severity of axonal injury corresponded to impact load and frequency in the optic nerve/tract and cere-
bellum. Both single and repeat injury protocols were associated with retinal ganglion cell loss and optic nerve
degeneration; these outcomes correlated with impact load and number/frequency. No phosphorylated tau immu-
noreactivity was detected in the brains of animals subjected to repetitive mTBI. Our findings establish a new
model of repetitive mTBI model featured by TAI in discrete CNS tracts, especially the visual system and cerebel-
lum. Injury in retina and optic nerve provides a sensitive measure of severity of mTBI, thus enabling further stud-
ies on mechanisms and experimental therapeutics. Our model can also be useful in exploring mechanisms of
chronic neurological disease caused by repetitive mTBI in wild-type and transgenic mice.
© 2014 Elsevier Inc. All rights reserved.

1. Introduction A recent trend that has attracted much attention is an increasing num-
ber of cases of progressive tauopathy in professional and amateur
There are at least 2–3 million new cases of civilian TBI in the US athletes with careers in collision sports such as football (Omalu et al.,
every year, most of them from motor vehicle accidents (MVA) or falls. 2005, 2006; McKee et al., 2009; Goldstein et al., 2012). These cases

Abbreviations: AD, Alzheimer's disease; ALS, amyotrophic lateral sclerosis; APP, amyloid precursor protein; BBB, blood–brain barrier; CCI, controlled cortical impact injury; CST, corticospinal
tract; CTE, chronic traumatic encephalopathy; CV, Cresyl violet; DAB, 3,3′-diaminobenzidine; DAPI, 4′,6-diamidino-2-phenylindole; FPI, fluid percussion injury; GOS, Glasgow Outcome Scale;
H&E, hematoxylin and eosin; IA, impact acceleration; IHC, immunohistochemistry; mTBI, mild traumatic brain injury; MVA, motor vehicle accidents; NFL, national football league; NSS,
Neurological Severity Score; ON, optic nerve; ONc, optic nerve segment proximal to chiasm; RGCs, retinal ganglion cells; ROIs, regions of interest; TAI, traumatic axonal injury.
⁎ Corresponding author at: The Johns Hopkins University School of Medicine, Division of Neuropathology, Department of Pathology, 720 Rutland Ave., Ross Research Building 558,
Baltimore, MD 21205, USA.
E-mail addresses: lxu9@jhmi.edu (L. Xu), judy.v.nguyen@gmail.com (J.V. Nguyen), mlehar@jhmi.edu (M. Lehar), amen692@gmail.com (A. Menon), lizrha@gmail.com (E. Rha),
jarena2@jhu.edu (J. Arena), jryu4@jhmi.edu (J. Ryu), Marsh-Armstrong@Kennedykrieger.Org (N. Marsh-Armstrong), crmarmar@vcu.edu (C.R. Marmarou), koliat@jhmi.edu
(V.E. Koliatsos).

http://dx.doi.org/10.1016/j.expneurol.2014.11.004
0014-4886/© 2014 Elsevier Inc. All rights reserved.
L. Xu et al. / Experimental Neurology 275 (2016) 436–449 437

have been linked to repetitive concussions and appear to be identical to


cases of dementia pugilistica (Martland, 1928; Millspaugh, 1937;
Corsellis et al., 1973), with which they are classified under the rubric
of chronic traumatic encephalopathy (CTE) (Goldstein et al., 2012).
Repetitive mild blast TBI from exposure to explosive munitions is the
signature injury in the Iraq and Afghanistan war theaters (Warden,
2006) and the Defense and Veterans Brain Injury Center has recently
developed return-to-duty guidelines to prevent further TBI incidents
in soldiers with concussive histories (Barth, 2011). The public concern
over repeat concussions is growing not only because of the risk among
NFL professionals and active-duty soldiers or veterans, but also because
of the exposure of millions of non-professional athletes playing football,
soccer and other contact sports (Langlois et al., 2006).
The development of animal models that approximate human con-
cussion scenarios can provide a much-needed proof of concept by
linking repetitive injury to adverse long-term effects, including neuro-
degeneration. Furthermore, animal models of repetitive TBI can be ap-
plied to transgenic rodents to work out molecular mechanisms that
may help identify subjects with genetic predispositions to traumatic
tauopathy. Such genetic backgrounds may include pathogenic tau mu-
tations (Ballatore et al., 2007) or H1 tau haplotypes (Ferrari et al.,
2011; Vandrovcova et al., 2010). In addition, animal models can serve
as vehicles for therapeutic targeting and experimental therapeutics.
In this paper, we modified a well-characterized rat model of closed,
non-contusive head injury, i.e. the impact acceleration (IA) model
(Marmarou et al., 1994; Beaumont et al., 1999), for repetitive use in
mice at a mild impact level. Although the IA model can produce a grad-
ed, widespread injury involving neurons, astrocytes, axons, and the mi-
crovasculature, it does not cause focal damage regardless of injury
severity. We believe that the mild, repetitive, non-contusive IA injury
by this IA mouse model that approximates conditions encountered in
repetitive concussion in contact sports, is instructive for repetitive
blast TBI occurring in combat, and may provide general insights for
situations featured by repetitive TBI such as epilepsy, self-injurious be-
haviors, and child and domestic abuse. Our findings are consistent
with the view that repetitive TBI causes cumulative axonopathy that Fig. 1. A simplified sketch of our mouse IA model that was adapted from the original rat
can lead to degeneration of axotomized CNS neurons. Marmarou model.

2. Materials and methods


All surgical procedures were carried out with gas anesthesia
2.1. Experimental animals and surgical procedures (isoflurane:oxygen:nitrous oxide = 1:33:66) and under aseptic condi-
tions. Briefly, the steel disk was glued on the saline washed, air dried
The subjects of these experiments were 5–6 weeks old C57BL6/J cranium between bregma and lambda under microscopic guidance
mice (male, n = 117; Charles River Laboratories, Wilmington, MA). (Fig. 1). The mouse was placed prone on the foam bed under the hollow
The animals received humane care in compliance with the Guide for Plexiglass tube, and secured with strapping tape. The injury was in-
the Care and Use of Laboratory Animals, 8th Ed. (National Academy duced by dropping the column of brass weights through the Plexiglass
Press, Washington, DC, 2011) and all animal care, surgical and post- tube from a distance of 1 m onto the disk (Fig. 1). The foam bed was
operative procedures were approved by the Animal Care and Use moved quickly right after the impact to avoid secondary rebound injury.
Committee of The Johns Hopkins Medical Institutions. The animals The mouse was then placed on warm pad for recovery. At the same
were housed in the vivarium with 12 h light/12 h dark cycles and time, self-righting time was recorded. Unlike the rat IA model, mechan-
given access to pellet food and water ad libitum. ical ventilation was not applied, in order to imitate real-life concussion
The animals were exposed to a modified version of the IA method of conditions. After righting itself, the mouse was re-anesthetized, the
TBI initially described for rat by Marmarou et al. (1994) (Fig. 1). The steel disk removed, and the skull checked under the surgical micro-
Plexiglass tube was 1 m high with an inner diameter of 10 mm, scope for skull fractures. The scalp incision was then closed with surgi-
designed to clear a column of 10 g, 3.6 mm-diameter brass cylinder cal staples, and the animals were returned to cage for full recovery.
weights. The diameter of the steel disk fixed on the mouse skull was Animals with skull fractures were excluded from further study. Sham
set at 3.2 mm, designed to ride over bregma and lambda sutures animals were subjected to the same procedures minus the weight
when affixed with cyanoacrylate to the skull, and thickness was set at drop maneuver. A list of experimental groups and numbers of animals
1 mm. As described in original rat IA model (Foda and Marmarou, is provided in Table 1.
1994; Marmarou et al., 1994), the main purpose of the stainless disk
was to prevent fracture. In this position, acceleration was confined to
the sagittal plane and caused both translational and rotational displace- 2.2. Grading severity of single TBI regimens
ment. The foam bed (4–0 spring constant foam, Foam to Size Inc.,
Ashland, VA) was used as initially described (Foda and Marmarou, To generate a range of injury severities based on lethality (n = 61),
1994; Marmarou et al., 1994), but cut at smaller dimensions of 20 × 8 the impactor height was set at 1 m. Weight was initially set at 10 g and
× 6 cm to accommodate the mouse body placed in the prone position. increased by 10 g increments. Weight increase stopped when mortality
438 L. Xu et al. / Experimental Neurology 275 (2016) 436–449

Table 1
Experimental design and groups of subjects.

Main experiments Survival time Measures (n = animal number)

A. Grading severity of single IA regimen to define mTBI parameters N/A for mortality, 7 days for TAI Mortality rate, n = 61 total
sham, 10 g, 20 g and 30 g, n = 4
40 g, n = 17
50 g and 60 g, n = 14
TAI severity in ON, corpus callosum and cerebellum, n = 16 total
sham, 20 g, 40 g and 60 g, n = 4 each
B. Ensuring animal wellbeing after each IA hit using parameters N/A Body weight, n = 46 total
established in (A) NSS, n = 20 total
C. Repetitive IA using mTBI parameters derived from (A)—brain 7 days TAI in OT, corpus callosum, cerebellum, n = 17 total
experiments
D. Repetitive IA using mTBI parameters derived from (A)—eye 7 days and 10 weeks Axon counts in ON, n = 16 total
and ON experiments RGC counts, n = 16 total
Microglial activation in retina, n = 16 total

The table shows experiments in which quantitative analysis was performed. Some studies, for example: general neuropathology for the assessment of contusions and hippocampal or
Purkinje cell death; establishment of site of indirect impact to the nervous system with BBB studies; neuroinflammation in brain based on IBA1, CD68, and GFAP IHC; and tauopathy
based on phosphorylated tau IHC were not included. All repetitive mTBI experiments in the brain and eye-ON contained 4 groups, i.e., sham, 1 × 10 g, 10 × 40 g and 12 × 40 g
(n = 4–5 each). The same animals were used for multiple experiments.
Abbreviations: mTBI, mild traumatic brain injury; TAI, traumatic axonal injury; NSS, Neurological Severity Score; BBB, blood–brain barrier; RGC, retinal ganglion cell; ON, optic nerve; OT,
optic tract.

reached 50%. The highest weight associated with zero mortality (40 g) with biotin (1:200; Jackson ImmunoResearch, West Grove, PA), sections
was used for all subsequent single and repeat injury regimens. were developed with a 3,3′-diaminobenzidine (DAB) kit (Vectastain
Elite ABC Kit; Vector Laboratories Inc., Burlingame, CA). For immunoflu-
2.3. Repetitive mTBI regimens orescence staining, after incubation in secondary antibodies conjugated
with Cy3 or Cy2 (1:200; Jackson ImmunoResearch, West Grove, PA)
Using the 40 g− 1 m IA setting associated with zero mortality, we for 2–4 h at room temperature, the sections were counterstained with
generated two novel repetitive mild TBI regimens (n = 56). The first the fluorescent DNA dye 4′, 6-diamidino-2-phenylindole (DAPI)
regimen involved 4 hits in one week at days 0, 1, 3 and 7; the second and coverslipped with DPX mounting media. Primary antibodies includ-
regimen included 12 hits at days 0, 1, 3 and 7 (3 hits on each of these ed: rabbit anti-GFAP (1:400; Dako, Carpinteria, CA); rat anti-CD68
days). In the second regimen, to ensure that the animals had enough (1:500; ABD Serotec, Raleigh, NC) and rabbit anti-IBA1 (1:500; Dako,
time to recover, time intervals were kept at 2 h between first and second Carpinteria, CA); mouse anti-AT8 directed against tau pS202 (1:200;
hits and 3 h between second and third hits. A single injury regimen was Thermo Scientific Inc., Rockford, IL); mouse anti-PHF1 directed against
used for comparison. In the repetitive regimens, at the date of injury, tau pS396 and pS404 (1:200); mouse anti-CP13 directed against tau
neurological deficits were assessed with an abbreviated NSS (Neurolog- pS202 and pT205 (1:200); rabbit anti-pS422 directed against phosphor-
ical Severity Score) system tailored for rodents exactly as described ylated Ser-422 tau (1:600; Genetex, Irvine, CA). Antibodies PHF1 and
without any modification (Flierl et al., 2009). In order to evaluate sub- CP13 were provided by Dr. Peter Davies (Albert Einstein College of
acute and chronic impacts from repeat injury, the animals were eutha- Medicine, Bronx, NY). For combinations of Fluoro-Jade C with IHC,
nized at day 7 or week 10 after single (for the single TBI regimen) or brain sections were first stained with IHC as described above using a sin-
final injury (for the repeat TBI regimens) and tissues were processed gle primary antibody. After incubation in the secondary antibody linked
as reported in the section below. with Cy3, sections were then processed for Fluoro-Jade C (Millipore,
Billerica, MA) staining (Schmued et al., 1997; Bian et al., 2007).
2.4. Histology, histochemistry, immunohistochemistry and microscopy Whole flat-mounted retinas were used to explore axonal injury,
survival of retinal ganglion cells (RGCs) and neuroinflammation. Retinas
Seven days after final injury, the animals were perfused with freshly were dissected, rinsed with phosphate buffered saline and then dually
depolymerized, 4% neutral-buffered paraformaldehyde. The brain and labeled with γ-synuclein (1:600; Genetex, Irvine, CA), to identify RGC
eyes with associated optic nerves (ONs) were dissected and immersed cell bodies, and the activated microglia marker CD68.
in the same fixative overnight at 4 °C. All tissues were cryoprotected Stained sections were studied on a Zeiss Axiophot microscope
and stored at − 80 °C for further processing. Sagittal brain sections equipped for epifluorescence (Diagnostic Instruments Inc., Sterling
(40 μm) were prepared in series for: general histochemistry, Heights, MI) or Zeiss LSM 510 inverted confocal microscope (Carl
i.e., hematoxylin and eosin (H&E) and Cresyl violet (CV); Gallyas silver Zeiss Inc., Oberkochen, Germany). Confocal microscopic images were
for axonal injury; immunohistochemistry (IHC) for neuroinflammation captured with pinhole set at 0.8 μm. Three-dimensional reconstruction
markers, i.e. GFAP, CD68 and IBA1 and for phosphorylated tau markers, by Z-stack scanning through regions of interest (ROIs) was acquired
i.e. AT8, PHF1, CP13 and pS422; and Fluoro-Jade C staining combined with LSM software. Adobe Photoshop 7.0 software (Adobe Systems,
with IHC for axonal injury and neuroinflammation. Fluoro-Jade C, a San Jose, CA) was used for montaging and image processing.
marker of neuronal degeneration (Schmued et al., 2005; Chidlow
et al., 2009; Pohl et al., 2011), was used to localize axonal injury and 2.5. Analysis of blood–brain barrier (BBB) disruption
neuronal cell death. For Gallyas silver staining of injured/degenerating
axons and terminals, sections were processed with a commercially Location and severity of primary traumatic impact to ON were ex-
available kit (Neurosilver kit II; FD Neurotechnologies, Ellicott City, plored with methods revealing BBB disruption as previously defined
MD) as described (Koliatsos et al., 2011). (Wang et al., 2011). Briefly, animals subjected to single injury or two re-
IHC utilized immunoperoxidase-DAB or dual-label immunofluores- petitive mild TBI regimens were euthanized 4 or 24 h after the sole or
cence methods and was performed as previously described (Koliatsos final injury event (n = 3 for each time point). ONs were dissected
et al., 2011; Xu et al., 2009). Briefly, sections were first incubated in away from the eyes and brains, cryoprotected and then sectioned in
the primary antibody overnight at 4 °C; for immunoperoxidase staining, the sagittal plane. Serial sections (10 μm) were mounted on gelatin-
after incubation with peroxidase-conjugated secondary antibody linked coated slides and dually immunolabeled for mouse IgG (serving as
L. Xu et al. / Experimental Neurology 275 (2016) 436–449 439

BBB leakage marker) with donkey anti-mouse antibody linked with Cy3 2.9. Statistical methods
(1:200; Jackson ImmunoResearch, West Grove, PA) and for amyloid
precursor protein (APP) with rabbit anti-APP IgG against the carboxy- One-way analysis of variance (STATISTICA 8.0; StatSoft, Tulsa, OK)
terminus of APP (1:200; Zymed, Camarillo, CA), as per previous Section. was used to compare among groups of animals based on silver staining,
ON axon count and RGC count data. Comparisons were performed
across single impact and sham groups in assessing the role of cylinder
2.6. Quantitation of degenerating axons in the ON weight on injury severity and across repeat injury groups and sham
animals in all repeat-injury experiments. Tukey's post hoc test was
ONs were dissected away from the eyes and brains and then applied to reveal the significant main effects or interactions.
processed for toluidine blue staining on semithin sections. Briefly,
ONs were treated with a solution containing 4% paraformaldehyde 3. Results
and 0.2% glutaraldehyde for 24 h. After rinsing in 0.1 M phosphate
buffer (pH 7.3) for 3–10 min, tissues were immersed in 1% osmium te- 3.1. General characterization of mouse IA model with single and repetitive
troxide for 15 min and stained en bloc with 1% uranyl acetate for 1 h. mild impacts
Stained tissues were dehydrated in graded concentrations of ethanol,
embedded in Poly/Bed 812 (Polysciences Inc., Warrington, PA) in As laid out in Materials and methods, the weight drop height was set
BEEM® capsules and polymerized at 60 °C for 72 h. Semithin sections at 1 m. Weight of impactor was tested starting at 10 g and further loaded
(1 μm) were cut transversely from segments of ONs proximal to the at 10 g increments. There was no fatality until weight reached 50 g. With
optic chiasm. Next, sections were stained with 1% toluidine blue. the 50 g− 1 m impact, mortality was 18.2% and with the 60 g− 1 m
Myelinated axons were counted under 100× magnification by investi- impact, mortality was increased up to 40%. Based on these initial mortality
gators blinded to experimental groups with stereological methods data, the 40 g−1 m impact was used in all subsequent repetitive injury
(the optical fractionator probe) using Stereo Investigator® software paradigms.
(Microbrightfield Inc., Williston, VT) as described (Turgut et al., 2010). Change of body weight was used as an index to determine the opti-
The counting frame was set at 7 × 7 μm and sampling grid was mal between-impact time intervals in the repetitive injury experiments.
60 × 60 μm. The final number of axons was automatically estimated In an initial pilot experiment using a consecutive 4-day schedule, the
by the software and used for analysis. 40 g− 1 m impact one or three times per day (12× or 4 × altogether)
arrested the physiological increase in body weight after the 2nd day
and/or 3rd day injury, respectively (Fig. S1). When the injury schedule
2.7. Quantitation of silver staining included time intervals between impacts, i.e. impacts at days 0, 1, 3
and 7, no significant disruption of body weight curve or enduring neu-
The intensity of silver labeling of injured axons within regions of in- rological deficits were observed irrespective of impact burden (Fig. 2).
terest (ROIs) in the brain and ON was studied with unbiased stereolog- No NSS increase was found on any scheduled day in the two repetitive
ical methodologies, i.e. area fraction fractionator (Hartman et al., 2005; injury regimens (4× and 12×).
Jefferson et al., 2011) using Stereo Investigator® software. This method Our single or repetitive IA model did not cause focal contusive injury
obviates irrelevant variance caused by some technical factors present in under the cranial disk or in other brain locations; in addition, our model
densitometric analysis such as overstaining, granular-particle back- did not cause anoxic/ischemic injury in hippocampal CA1 or overt
ground and image processing. The result is presented as area fraction reduction in numbers of cerebellar Purkinje cells (Fig. 3A–C). Fluoro-
of silver (+) linear configurations in counted frames through the ROI. Jade C staining was negative for cell bodies in all these locations in single
In the present experiments, ROIs included optic tract (next to optic or repetitive regimens (data not shown). Based on the absence of frac-
chiasm), corpus callosum (same sections as optic tract) and cerebellar tures, zero mortality, no significant changes in body weight and no
lobule 9 (starting from section proximal to the lateral edge of lobule major neurological deficits as well as rapid neurological recovery after
9). Four serial sagittal sections (every 8 in series of 40 μm thickness each injury event, the single 40 g− 1 m impact was designated here
transverse sections) were selected through the ROIs and subjected to as “mild” injury. This designation conforms to the original IA model
stereological analysis. Counting frame was set at 50 × 50 μm and sam- description in which 450 g− 1 m was used to generate “mild” injury
pling grid and grid spacing were 200 × 200 μm and 10 μm, respectively. (Foda and Marmarou, 1994; Marmarou et al., 1994; Beaumont et al.,
The final number of axons was automatically estimated by the software 1999). As shown in the following sections, this setting (40 g− 1 m)
and entered for analysis. causes traumatic axonal injury (TAI) in multiple locations in mice, but
this pattern is similar to that in other mild TBI mouse models including

2.8. Retinal ganglion cell counts in whole-mount retina

Whole flat-mounted retinas dually immunolabeled for γ-synuclein


and CD68 were imaged with a 0.4 numerical aperture Zeiss 20× objec-
tive on a Zeiss 200 M inverted microscope (Carl Zeiss, Oberkochen,
Germany) equipped with a Linear Encoder Flat Top Zeiss 200 Ludl mo-
torized stage (Ludl, Hawthorne, NY), Roper Scientific Coolsnap FX digi-
tal camera (Photometrics, Tucson, AZ), and Excite fluorescence source
(EXFO Burleigh, Victor, NY) (Soto et al., 2008). After acquired images
were adjusted for right contrast and brightness using Adobe Photoshop
7.0, ImageJ (National Institutes of Health, Bethesda, MD) was used for
counting surviving γ-synuclein (+) RGCs and CD68 (+) microglia.
Cells were counted in 100 × 100 μm fields evenly assigned in the
peripheral, intermediate and central zone of retina. At least 10 fields
in the peripheral and intermediate zones and 8 fields in the central Fig. 2. Trends in body weight of animals included in the present study. Body weight curves
zone were utilized. Cell density was then calculated by dividing total with single (1 × 40 g) and repetitive mild (4 × 40 g and 12 × 40 g) injuries illustrate that
cell numbers with total area surveyed. animals gain weight at a rate comparable to that of sham subjects.
440 L. Xu et al. / Experimental Neurology 275 (2016) 436–449

corticospinal tract were especially prominent. TAI in cerebellum was


restricted to lower folia close to the medulla.
Intensity of axonal degeneration depended on severity of injury as
determined by the weight of the impactor (in single hit scenarios)
(Fig. 4E and F) or number of hits (in multiple-hit scenarios) (Fig. 5).
The severity of TAI at various levels of impact burden in single-hit regi-
mens (sham, 20 g, 40 g and 60 g) and repeat-hit regimens (4 × 40 g
and 12 × 40 g) was assessed using Gallyas silver staining in three
regions, i.e. optic tract, the white matter of cerebellar lobule 9 and
corpus callosum (Fig. 4A–D). These regions are representative of severe
(optic tract), intermediate (lobule 9) and mild (corpus callosum) TAI
due to IA (Figs. 4 and 5). In the case of optic tract, the severity of TAI
appears to worsen as impactor weight increases from 20 g to 60 g in
the single hit group (Fig. 4E). Area fraction fractionator data confirm
this trend (Fig. 4F, left panel), although differences between the 40 g
and 60 g animals are not statistically significant. Severity of TAI in cere-
bellar lobule 9 demonstrates a similar trend, but the significant differ-
ences in this case are between sham and both 40 g and 60 g animals
(Fig. 4F, center panel). In the case of corpus callosum, only the 60 g im-
pact causes significant TAI (Fig. 4F, right panel). Besides showing a rela-
tionship between IA burden and TAI in our mouse model, the above
trends also indicate the differential vulnerability of axons in various
white matter tracts/regions.
In the case of mild repetitive TBI, both the low- and high-frequency
regimens (4 × 40 g and 12 × 40 g, respectively) produced significant TAI
in optic tract compared with the single injury regimen (1 × 40 g) and
sham, although there was no difference in the TAI outcome between
the two repetitive injury groups (Fig. 5A). The same pattern was seen
in the corpus callosum (Fig. 5C). In cerebellar lobule 9, both repetitive
injury regimens cause TAI, but they are not different when compared
with the single injury group (1 × 40 g) (Fig. 5B). These results imply
that, at some level, at least using the regimens and histochemical proce-
dures of this paper and in certain white matter regions, the cumulative
effect of repetitive injury becomes stabilized.

3.3. Neuroinflammatory changes in sites of TAI in the mouse repetitive mild


IA model

With repetitive mild IA injury, we found extensive microglial infil-


tration/activation in TAI sites, especially optic tract including superior
colliculus, corticospinal tract, corpus callosum and part of cerebellum
white matter (Fig. 6). Microglial activation was ascertained by the pres-
ence of deramified, large, often clustered IBA1 (+) cells, many of which
expressed the lysosomal phagocytic marker CD68 (Fig. 6A and B).
Astrocytic response is also increased based on the density of GFAP (+)
Fig. 3. Absence of focal contusion or anoxic/ischemic injury in the brain of mice subjected
processes (Fig. 6C and D). In several TAI sites including ON/optic tract,
to mild IA injury. CV staining 7 days after repetitive mild injury (4 × 40 g) does not show
focal contusion underneath the cranial disk (A) or significant neuronal death in Ammon's corticospinal tract and cerebellum areas, there was co-distribution of in-
horn (B) or cell death of Purkinje cells (C). Mol, molecular layer; P, Purkinje cell layer; jured Fluoro-Jade C (+) axons and phagocytic CD68 (+) microglia
Gran, granular layer. Scale bars: A, 500 μm; B and C, 100 μm. (Fig. 6E–H).

fluid percussion injury (FPI) (Greer et al., 2011; Wang et al., 2011), blast 3.4. Degenerative changes in the visual system with single or repetitive mild
injury (Koliatsos et al., 2011), electromagnetic coil-based impact TBI
(Mouzon et al., 2012) and controlled cortical impact injury (CCI)
(Shitaka et al., 2011). The initial impact of TBI to the ON was assessed at 4 or 24 h post-IA
injury with mouse IgG and APP immunofluorescence on same sections.
Four hours after a single impact (1 × 40 g), we found that a sizeable seg-
3.2. TAI in the brains of mice subjected to IA with single and repetitive mild ment of ON closer to the eye was labeled by anti-mouse IgG antibody, a
impacts sign of BBB disruption due to the impact; this segment also showed
dense axonal swellings and bulbs stained with APP antibodies
Axonal injury in the brain of mice subjected to IA with single and re- (Fig. 7A). With a repetitive injury regimen (4 × 40 g), IgG immunoreac-
petitive mild impacts was investigated with Gallyas silver and Fluoro- tivity was weaker than the single injury and so was the presence of APP
Jade C staining. With these methods, our mouse IA model showed selec- (+) axonal swellings or bulbs (Fig. 7C). Compared to 4 h, ONs at 24 h
tive TAI in the ON/optic tract (Fig. 4A), lower corticospinal tract (Fig. 4B) post-single injury or after the last hit of repetitive injury regimens
and cerebellar white matter/cerebellar peduncles (Fig. 4C); fiber abnor- showed restricted IgG immunoreactivity and fewer axonal abnormalities
malities were also noted in the corpus callosum (Fig. 4D), internal cap- (Fig. 7B and D). Furthermore, in animals with repetitive injury regimens,
sule and lateral and medial lemnisci. Injuries in ON/optic tract and lower axon bulbs presented mainly at the periphery of ONs (Fig. 7C and D).
L. Xu et al. / Experimental Neurology 275 (2016) 436–449 441

Fig. 4. Selective axonal injury in the white matter of single and repetitive mild injured mice. Gallyas silver staining shows axonal injury (black lines or trails of dots) in select locations
including optic tract (OT, A, arrow heads), cerebellar lobule 9 (Cb9, B), corticospinal tract (CST, C, arrow heads) and corpus callosum (cc, D, arrow heads) after low-frequency repetitive
injury (4 × 40 g). Higher magnification images (insets) are enlarged from corresponding frames in main panels to show details. In the corticospinal tract (C), inset showcases a typical axon
bulb (arrow). Severity of TAI in the optic tract worsens when impactor weight increases from 20 g to 60 g (E). There is no TAI in the optic tract of sham animals (E, upper panel). Panel F
shows the results of quantitation of silver staining in the optic tract (left panel), cerebellar lobule 9 (center panel) and corpus callosum (right panel) of sham and single injured animals
with impactor weights at 20, 40 and 60 g. One-way ANOVA is used for analysis. * p b 0.05. Scale bars: A, 100 μm; B–D, 50 μm.

As mentioned in previous Sections, IA causes widespread axonal in- sections through ON segments proximal to chiasm (ONc) after toluidine
jury and intense microglial response in the mouse visual pathway in- blue staining. In ONc, surviving axon numbers after single and repetitive
cluding ON, optic tract and superior colliculus. Retinal ganglion cells injury were significantly lower than after single or sham injury. Fifty to
(RGCs), i.e. the main projection neurons in ON/optic tract were further eighty percent of axons were lost after the repetitive injury regimens
examined for evidence of neuronal degeneration in the animals subject- (Fig. 9E). The area affected the most was the center of the nerve that
ed to mild single and repetitive IA injury. RGC loss after mild single and contained large spaces devoid of axons (Fig. 9A–D). Mice with high-
repetitive injuries in whole-mount retinas was obvious at 10 weeks frequency repetitive injury (12 ×) lost significant number of axons
after final injury (Fig. 8A–D). The density of surviving γ-synuclein (+) than low-frequency repetitive injury group (4×) in this area. Some de-
RGCs in whole-mount retinas was already decreased in mice subjected gree of ONc axonal degeneration was also seen with single mild injury
to high-frequency repetitive injury (12×) at 7 days after injury (Fig. 8E), (Fig. 9B).
whereas RGCs in single and low-frequency repetitive (4×) injury regi- In the RGC layer of injured mice, there was intense microglial activa-
mens did not show significant cell loss at this early time point tion. Using whole-mount retinal preparations stained with markers of
(Fig. 8E). At 10 weeks, RGC density was significantly reduced in mice in- activated microglia, we found that numbers of CD68 (+) microglial
jured with either single or both repetitive injury regimens (Fig. 8F). cells were increased 7 days after injury in all injured groups (Fig. 10D).
Mice injured with repetitive mild regimens had significantly lower Confocal 3D reconstruction of CD68 and γ-synuclein stained retina im-
RGC densities than mice subjected to single injury. Although mice ages demonstrates that the RGC layer was populated by activated mi-
injured with the high-frequency (12 ×) repetitive regimen had lower croglia (Fig. 10A-C). Cell counts of CD68 (+) microglia 7 days after
RGC densities than animals injured with the low-frequency (4 ×) injury confirmed that both single and repetitive injuries caused a signif-
regimen, there was no significant difference between the two repetitive icant increase in the number of activated microglial cells. Although there
injury groups. RGC loss in the mild single injury group was significant at were no significant differences among injury groups, these results show
10 weeks but not 7 days post final injury (p b 0.05), a difference suggest- that there is a positive correlation between injury burden and microglia
ing that RGC death begins to take place after 7 days. activation (Fig. 10D). At 10 weeks post injury, there were no significant
Axonal counts in the ON were performed at 10 weeks post-injury to differences in numbers of activated microglial cells among sham, single
confirm the degeneration of RGCs. Counts were performed on semithin and repetitive injury groups (Fig. 10E), a pattern suggesting that the
442 L. Xu et al. / Experimental Neurology 275 (2016) 436–449

or anoxic–ischemic injury in the mouse brain. Rather, our model pro-


duces selective TAI in certain white matter tracts; this effect is coupled
with local neuroinflammatory responses and is positively associated
with TBI severity. A remarkable finding is the consistent involvement
of the visual system, with degeneration of axons in both ON and optic
tract and loss of RGCs. Visual involvement may be associated with direct
injury related to the concussion. The effect of repetitive mild TBI at sites
of TAI is cumulative but eventually reaches a plateau, although it is un-
known whether the effect is saturated at that level. In summary, here
we have developed and characterized a mouse model of repeat mild
TBI featured by TAI and neuroinflammatory components that are TBI
dose-dependent and accumulate with each repeat TBI event. As such,
this model lends itself to further investigations that have translational
value for sports and military injuries, including CTE.
Animal models are powerful tools to explore cause-and-effect rela-
tionships between traumatic insults and progressive neurodegenera-
tion and, eventually, to serve as vehicles for therapeutic targeting and
experimental therapeutics or for biomarker discovery and validation.
A distinct advantage of mouse TBI models in particular is that they can
be used to study the role of generic vulnerability in the outcome of
TBI. For example, transgenic mice harboring dementia-associated iso-
forms of ApoE, especially ApoE4, or pro-aggregation forms of tau may
be extremely useful to disclose the role of genetic vulnerability to CTE
after exposure to mild repetitive TBI (Strittmatter et al., 1993; Bu,
2009; McKee et al., 2009; Wang et al., 2007).
Some TBI animal models have been used in mild repetitive scenarios,
for example a modified CCI model (Laurer et al., 2001; Uryu et al., 2002;
Conte et al., 2004; Longhi et al., 2005; Shitaka et al., 2011), or the weight
drop model of Hall (Deford et al., 2002; Creeley et al., 2004). Another
mild repetitive TBI model is an electromagnetic coil-based impact
model modified from the electromagnetic version of the CCI paradigm
(Brody et al., 2007; Mouzon et al., 2012). Blast injury models have also
been scaled down to generate mild-moderate TBI, but these models
have not been used in repetitive injury scenarios because of potential le-
thal injury to vital organs, such as the lung (Cernak et al., 2011; Koliatsos
et al., 2011). Compared to the IA model used here, these mild repetitive
injury scenarios cause contusions or involve minimal to no head move-
ment. Although the IA model has been used to inflict mild injury in the
rat (Fujita et al., 2012; Marmarou et al., 1994), it has not been
established as a model of repetitive TBI in the rat or mouse.
As indicated elsewhere, the mouse model used in this paper was de-
rived from the rat IA model by modifying some key parameters. An im-
portant parameter is the weight–height configuration of the impactor.
Assuming a steady height of 1 m, weights of 50 g or above were associ-
ated with high mortality because of apnea. The 40 g− 1 m setting did
Fig. 5. Quantitation of Gallyas silver signal. This figure shows quantitation of Gallyas silver not have any fatalities, as is almost universally the case with human
(+) degenerating axonal profiles in the optic tract (OT, A), cerebellar lobule 9 (Cb9, concussions; therefore, this setting was used in all single and repetitive
B) and corpus callosum (cc, C) of sham, single injured (1 × 40 g) and repetitive mild in- mild TBI regimens employed here. Another important variable is the
jured (4 × 40 g and 12 × 40 g) animals. One-way ANOVA with Tukey's post hoc test was
size of the stainless steel disk used to prevent fractures. Because of the
used to assess significance of variance. * p b 0.05.
thinness of the mouse cranium, unless the disk overlapped the bregma
and lambda sutures, i.e. cut at a diameter of 3.2 mm, it could not prevent
microglial neuroinflammatory response is an early event in the neuro- fractures. The young age of mice used (5–6 weeks old) was chosen in
degenerative process after the injury. order to recapitulate the age of athletes suffering mild TBI from contact
We also explored the issue of tau hyperphosphorylation in the brains sports (Ojo et al., 2013).
of mice 10 weeks after high-frequency (12×) repetitive injury with an In our repetitive mild TBI regimens, we used the “subacute” injury
extensive panel of phosphorylated tau antibodies. In representative sag- schedule, i.e., injury at days 0, 1, 3 and 7. On each one of these days,
ittal brain sections, we found no phosphorylated tau immunoreactivity the animals were exposed to single or three hits (for a total of 4 or 12
in cortical or subcortical neurons. Our antibodies did stain neurons in TBI events, respectively). The use of three hits in a single day was
the brains of tau P301S mice that served as positive controls (Fig. S2). meant to replicate the pattern of multiple concussions in a single athlet-
ic encounter that is common in the world of collision sports. The use of
4. Discussion consecutive-day schedules was discouraged by our initial finding that
such a regimen interfered with normal weight gain, evidence of feeding
4.1. General or metabolic problems, or both. The “recovery” time intervals used in
the subacute repetitive TBI schedule were 1 day, 2 days and 4 days for
Our findings indicate that our mild repetitive TBI mouse model, a the second, third and fourth hits, respectively. Animals injured in such
modification of the rat IA paradigm, does not cause either focal contusive a fashion did not show evidence weight loss or neurological deficits
L. Xu et al. / Experimental Neurology 275 (2016) 436–449 443

Fig. 6. Neuroinflammation in white matter tracts undergoing TAI after IA injury. Neuroinflammation is found in the optic nerve (ON)/optic tract (OT), corticospinal tract (CST) and cere-
bellar lobule (Cb) of repetitive mild injured (4 × 40 g) animals 7 days after final injury and is evidenced by IBA1 (A and B) or GFAP (C and D) staining or dual staining with CD68 and Fluoro-
Jade C (E–H). In the optic tract, repetitive injured (4 × 40 g) animals (B) have more IBA1 (+) microglia than sham animals (A). Some reactive microglial cells acquire phagocytic cytologies
(asterisks in B). Inset in (B) shows an activated microglial cell with phagocytic appearance (arrow) and one resting microglial cell (arrowhead). GFAP staining (C and D) in the optic tract
shows that animals exposed to repetitive mild IA injury generate a strong astrocytic response. CD68 (+) reactive microglia colocalizes with Fluoro-Jade C (+) injured axons in the ON (ON,
E), optic tract (OT, F), corticospinal tract (CST, G) and cerebellar lobule (cb, H) of animals exposed to repetitive mild injury at 7 days post final injury. Reactive microglia is labeled in red and
injured axons in green. Scale bars: A, B and E, 50 μm; C, D, F, G and H, 100 μm.

with abbreviated NSS testing. NSS used in our subjects is a sensitive, deficits were detected with the Morris Water Maze (Beaumont et al.,
specific and very popular motor and cognitive scale used for the evalu- 1999).
ation of TBI severity and recovery in mice (Flierl et al., 2009). The fact
that NSS does not detect motor or behavioral deficits in our mice is con- 4.2. Traumatic axonal injury
sistent with the idea that our regimen causes mild TBI. However, it is
possible that subtle impairments may require more specialized testing; Gallyas silver and Fluoro-Jade C staining indicate the presence of se-
for example, in the rat IA model, mild spatial learning and memory lective axonal injury whose severity is related to the IA burden as
444 L. Xu et al. / Experimental Neurology 275 (2016) 436–449

4.3. Neuroinflammatory changes

TAI sites in our mild single or repetitive IA model show intense


neuroinflammatory responses involving activated microglia and
astrocytes. Activated microglia are deramified or present with round
cytologies that are typical of phagocytes and express the macrophage/
lysosomal marker CD68. This microglial response is consistent with ob-
servations in other animal models of TBI (Chen et al., 2003; Kelley et al.,
2007; Shitaka et al., 2011) and is also seen in the brains of patients with
TBI histories (Johnson et al., 2013; Smith et al., 2013; Gentleman et al.,
2004). Microglial activation in TBI can be detrimental or beneficial
(Morganti-Kossmann et al., 2002). The first corresponds to the classical
neurotoxic pathway (M1 cells); the latter involves activation in the al-
ternative neuroprotective pathway (M2 phenotype) (Martinez et al.,
2008; Loane and Byrnes, 2010). With progressive injury beyond two
weeks, microglia may transform from M2 to M1 (Hu et al., 2012). In
the early stage of TBI, M2 microglia clears debris from dead neurons
and protects vulnerable neurons by releasing trophic factors or cyto-
kines (Neumann et al., 2009). In some cases, microglial activation after
TBI persists for months or even years (Gentleman et al., 2004; Johnson
et al., 2013), a pattern indicating chronic neuroinflammation. Although
the role of this chronic neuroinflammation in TBI scenarios is not entire-
ly clear, in classical neurodegenerative diseases such amyotrophic later-
al sclerosis (ALS) and Alzheimer's disease (AD) activated (transformed)
microglia is presumed to promote further degeneration (Akiyama et al.,
2000; Liao et al., 2012). Similarly, in the hypoglossal axotomy model,
activated microglia becomes neurotoxic 14 days after injury, a transfor-
mation suppressible by minocycline (Yamada and Jinno, 2013). In
Fig. 7. Disruption of blood brain- barrier (BBB) in ON of IA-injured animals. BBB disruption addition, suppression of microglial activation by minocycline has
and primary TAI in the ON with single (A and B) and repetitive mild injury (C and D) was
shown benefits in the animals subjected to weight drop TBI (Siopi
examined with double immunofluorescence for mouse IgG (red) and APP (green). Eyeball
is to the left of panel, optic chiasm on the right (asterisk). Four hours after single mild in- et al., 2011, 2012a,b; Homsi et al., 2010).
jury (1 × 40 g) (A), there is disruption of BBB over a large segment of ON close to the eye The role of microglia in axonal regeneration is complex. Both bene-
(red, arrowhead). There are also axonal swellings and bulbs on the ocular (left) side of the ficial and detrimental effects have been reported (McPhail et al., 2004;
disrupted BBB front, evidence of primary TAI at the BBB disruption site. Inset illustrates de- Prewitt et al., 1997; Donnelly and Popovich, 2008). In one study, activat-
tails of a typical axonal swelling. Twenty four hours after single injury (B), IgG immunore-
ed microglia was shown to promote the retraction of injured axons
activity is diminished (red, arrowhead) and abnormal axon signal is reduced as well
(green, arrows). In the case of repetitive injury (4 × 40 g) (C and D), there is little BBB dis- (Horn et al., 2008). While the role of activated microglia in the outcome
ruption (red, arrowhead) and only few retraction balls form at the ocular aspect of seg- of TAI will need to be clarified with further research, the specific
ment with BBB disruption (green, arrows). Scale bars: A–D, 500 μm. association of neuroinflammation with TAI sites may serve as the basis
of neuroimaging for the diagnosis of concussion. Based on previous
pathological evidence showing inflammation in the brain of patients
with concussion (Oppenheimer, 1968) and our present findings in ani-
measured by impactor weight and number of IA events (Figs. 4 and 6). mals, PET ligands marking activated microglia such as [11C](R)-PK11195
Although the size and positioning of the metal disk could contribute to or even astrocytes such as [11C]-DED can be used to localize areas of
this distribution of impact forces and therefore influence the regional axonal injury in patients with concussion (Banati, 2002; Carter et al.,
pattern of TAI observed here, similar patterns have been observed 2012).
with other blunt (Wang et al., 2011) and blast (Koliatsos et al., 2011)
TBI models. It is possible that additional factors germane to the involved 4.4. Degeneration of the visual system
pathways may determine the regional intensity of TAI.
With the single-injury regimen, our findings indicate a correlation The visual system was severely and consistently impacted in our
between severity of axonal injury and weight of impactor, especially model and the visual pathway was the principal site of TAI along with
evident in optic tract and cerebellar white matter (area 9Cb) (Fig. 5). the corticospinal tract (Fig. 4). Injury to the visual system include the
In the corpus callosum, single impacts below 50 g− 1 m do not retina, ON, optic tract, the lateral geniculate nucleus and superficial
cause significant axonal injury; TAI is evident at the level of 60 g−1 m layers of the superior colliculus and is associated with an early primary
(severe injury). With the repetitive mild injury regimens (4 × 40 g effect of IA to the ON. The intense BBB disruption at 4 h after single inju-
and 12 × 40 g), we found a cumulative effect of IA in the optic tract ry that subsides at 24 h is consistent with an early effect at the ON level.
and corpus callosum (Fig. 5). However, with the silver staining used A corresponding trend in APP (+) axon abnormalities is also supportive
here, we did not see a difference in TAI in the optic tract, cerebellum of this idea. In the rat, BBB is known to open quickly in less than 4 h and
and corpus callosum between the two regimens, a pattern suggesting then close after IA injury using parameters corresponding to the ones
that TAI may plateau at a level between 4 × 40 g and 12 × 40 g. Although used here (Beaumont et al., 2000). The presence of early axonal lesions
TAI in corticospinal tract (CST) was not quantified, severity of impact in ON segments overlapping these with BBB disruption indicates the site
(as represented by impactor weight) and number of hits (the cumulative of impact and is consistent with findings in the fluid percussion model
factor) seem to also correlate with TAI severity (L. Xu, personal of TBI (Wang et al., 2011,). The peak of APP immunoreactivity in TAI
observations). All these findings indicate a selective vulnerability to in- varies from location to location (Bramlett et al., 1997; Spain et al.,
jury within white matter tracts in our mouse IA model and also demon- 2010). The presence of fewer APP (+) axonal abnormalities in the ON
strate some correlation of TAI severity in these locations with impact with repetitive injury suggests that the primary traumatic effect of IA
burden and number of IA hits. in the ON may have reached a plateau prior to the final hit or that
L. Xu et al. / Experimental Neurology 275 (2016) 436–449 445

Fig. 8. Retinal ganglion cell (RGC) loss after single and repetitive mild injury. In whole-mount retinas stained with the RGC marker, γ-synuclein (Sncg), repetitive mild injury (4 × 40 g and
12 × 40 g) causes significant RGC loss (C and D) at 10 weeks. Even single injured mice (1 × 40 g) have lower RGCs (B) than sham mice (A). Insets are enlargements of frames in main panels
to show more detail. Densities of surviving RGCs in the retinas of sham, single and repetitive mild injured animals are shown in (E) and (F), 7 days and 10 weeks post final injury,
respectively. Data were analyzed with one-way ANOVA followed by Tukey's post hoc test. * p b 0.05. Scale bars: A–D, 100 μm.

ongoing axonal degeneration leaves fewer residual axons exposed to in- (and RGC degeneration) may continue, to some extent, even beyond
jury with every subsequent hit. this time point.
In the retina, no RGC loss is seen after single mild injury at 7 days, but This paper is a detailed report of RGC degeneration in rodents after
cell death is significant at 10 weeks and involves more than a third of repetitive mild blunt TBI. We have previously presented evidence of
RGCs. Based on the known outcomes of ON axotomy, RGC degeneration RGC degeneration associated with TAI in the ON-optic tract after
is most likely secondary to ON injury (Dratviman-Storobinsky et al., mild–moderate blast injury in mice (Koliatsos et al., 2011). ON injury
2008; Leung et al., 2008). In the case of repetitive injury, RGC loss is sig- was also reported in a rat model of central fluid percussion injury
nificant already at 7 days and approaches 40% of RGCs in the high- (Wang et al., 2011), although a subsequent study from this group did
frequency repetitive regimen (12 × 40 g). At 10 weeks post injury, not demonstrate RGC loss (Wang et al., 2013). In another repetitive
more than 50% of RGCs degenerate with the 4 × 40 g regimen and 70% mild TBI model with minimal, if any, head movement (Mouzon et al.,
of RGCs die with the 12 × 40 g regimen. In the case of repetitive injury, 2012), five consecutive insults caused significant loss of brain-specific
severity of RGC degeneration corresponds to severity of TAI in the ON. homeobox/POU domain protein 3A (+) RGCs, thinning of the inner
Neuroinflammatory response in retinas, primarily evidenced by an in- retina, and decreased photopic negative response at 10–13 weeks
creased number of activated microglial cells, is significant at 7 days post (Tzekov et al., 2014). These results are consistent with our findings
injury but returns to control levels at 10 weeks in all experimental groups using the IA model and suggest that injury to visual system is not un-
exposed to either single or repetitive injury. These findings suggest that common in rodent models of mild TBI. Concussion patients frequently
the bulk of neuroinflammatory response in retinas has ended at 10 report blurred or double vision, problems with accommodation and
weeks, although it is conceivable that low-grade neuroinflammation other visual symptoms (Rees, 2003; Bowen, 2003; Erlanger et al.,
446 L. Xu et al. / Experimental Neurology 275 (2016) 436–449

Fig. 9. Axonal degeneration in the ON of IA-injured animals. Axonal pathology in the ON of mice exposed to single and repetitive mild injury was examined with toluidine blue staining of
semithin sections from ON segments proximal to chiasm 10 weeks post-injury. Panels A–D illustrate type and distribution of axonal pathology. Panel E is a bar diagram plotting numbers of
surviving axons in animals subjected to sham, single and repetitive mild injury. In A–D, stained semithin sections depict increasing severity of axonal degeneration from single (B) to
repetitive (C) injury and from low-frequency (C) to high-frequency (D) repetitive injury. Panel A illustrates a sham control for the purpose of comparison. Degenerating axonal profiles
(arrows) are especially abundant at the center of the nerve, whereas the periphery is relatively spared with higher numbers of surviving axons (arrowheads) at each level of injury severity.
Axonal loss leaves spaces and cavities, especially at the center of the nerve (asterisks). Panel E illustrates the progressive nature of degeneration based on injury burden. Axons were
grouped per experimental history; averages were analyzed with one-way ANOVA followed by Tukey's post hoc testing. * p b 0.05. Scale bars: A–D, 10 μm.

1999). In addition, patients with severe TBI including blast injury have used to explore the role of mild repetitive TBI in causing tauopathy in a
marked visual deficits (Brahm et al., 2009; Hellerstein et al., 1995; localized region of the nervous system that would be more straightfor-
Summers, 2006). Such symptoms probably have complex origin and ward to study. IHC for multiple phosphorylated tau epitopes failed to
there is no evidence that, in humans, there is TAI in visual pathways demonstrate tau hyperphosphorylation in the brains of wild-type
after concussion. The involvement of the visual system in rodent models mice injured as per protocols used here. In another mouse TBI model
of TBI including concussion bypasses the controversy of the presence or based on mild blast exposure in a shock tube, a single blast event was
not of authentic primary axonal injury in rodent species. Axonal injury reported to have caused tauopathy (Goldstein et al., 2012). Reasons
is certainly accentuated in gyrencephalic brains because of the abun- causing this variability may include differences in injury mechanisms
dance of white matter tracts (Smith et al., 1997, 2003; Browne et al., between our model and theirs. The issue of phosphorylation of wild
2011). Although the rodent brain as a whole is not as vulnerable, the vi- tau in rodent species remains controversial and other investigators
sual system and perhaps also some cerebellar tracts appear to be sus- have also reported negative results in blast injured rat (Gama Sosa
ceptible. Reasons for this phenomenon may be the relative abundance et al., 2014; Sosa et al., 2013) and wild type mouse (Xu et al., 2014).
of white matter in these systems or biomechanical peculiarities related Another advantage of our model is that it offers great opportunities to
to distribution of shearing forces. Whatever the cause, the visual system explore experimental therapies for TAI. Neurons of interest in visual
may be the ideal site to study primary TAI in rodent species. TAI, i.e. RGCs, are exposed in the vitreous chamber and thus eminently
The preferential injury to the visual system in our mouse model of accessible for direct therapeutic manipulations with intravitreous injec-
repetitive mild TBI, including degeneration of RGCs, may also have im- tions of therapeutic compounds or grafts of cell preparations including
plications for research on CTE. Compared with the corticospinal tract, stem cells (Xin et al., 2013; Bharti et al., 2014).
i.e. another system implicated in our model with a rather wide distribu- In summary, the rat IA model can be adjusted for use in mild repet-
tion of nerve cells of origin, the population of affected nerve cells in the itive TBI protocols in mice. These protocols do not cause fractures or
visual system is concentrated in the retina. Therefore, our model can be focal contusions and also do not cause significant anoxic–ischemic
L. Xu et al. / Experimental Neurology 275 (2016) 436–449 447

Fig. 10. Neuroinflammation in the retinal ganglion cell (RGC) layer after single and repetitive mild injury. Repetitive mild injury (4 × 40 g) leads to a significantly higher number of CD68
(+) activated microglial cells (green) in the RGC layer compared to sham (B) 7 days after final injury. Confocal microscopy (C) verifies that CD68 (+) microglial cells (green) are localized
in the γ-synuclein (+) RGC layer (Sncg, red, arrows). Densities of CD68 (+) microglia in the retinas of sham, single and repetitive mild injured animals are shown in D and E at 7 days and
10 weeks post final injury, respectively. One-way ANOVA with Tukey's post hoc test is used for analysis. * p b 0.05. Scale bars: A and B, 20 μm; C, 50 μm.

injury. Instead, such mild repetitive protocols generate TAI in select CNS Beaumont, A., Marmarou, A., Hayasaki, K., Barzo, P., Fatouros, P., Corwin, F., Marmarou, C.,
Dunbar, J., 2000. The permissive nature of blood brain barrier (BBB) opening in
sites coupled with local microglial activation and phagocytic transfor- edema formation following traumatic brain injury. Acta Neurochir. Suppl. 76,
mation. The visual system is particularly vulnerable in these protocols. 125–129.
Our model of mild repetitive injury and the involvement of the visual Bharti, K., Rao, M., Hull, S.C., Stroncek, D., Brooks, B.P., Feigal, E., van Meurs, J.C., Huang,
C.A., Miller, S.S., 2014. Developing cellular therapies for retinal degenerative diseases.
system in particular support the notion that it is possible to generate Invest. Ophthalmol. Vis. Sci. 55, 1191–1202.
consistent primary TAI in the rodent brain. Moreover, this model can Bian, G.L., Wei, L.C., Shi, M., Wang, Y.Q., Cao, R., Chen, L.W., 2007. Fluoro-Jade C can
serve for further investigations into mechanisms of mild repetitive TBI, specifically stain the degenerative neurons in the substantia nigra of the 1-
methyl-4-phenyl-1,2,3,6-tetrahydro pyridine-treated C57BL/6 mice. Brain Res.
including tau-related neurodegenerative events in genetically vulnera- 1150, 55–61.
ble subjects, and is eminently applicable to therapeutic interventions Bowen, A.P., 2003. Second impact syndrome: a rare, catastrophic, preventable complica-
targeting mild repetitive TBI and its chronic complications. tion of concussion in young athletes. J. Emerg. Nurs. 29, 287–289.
Brahm, K.D., Wilgenburg, H.M., Kirby, J., Ingalla, S., Chang, C.Y., Goodrich, G.L., 2009. Visual
Supplementary data to this article can be found online at http://dx.
impairment and dysfunction in combat-injured servicemembers with traumatic
doi.org/10.1016/j.expneurol.2014.11.004. brain injury. Optom. Vis. Sci. 86, 817–825.
Bramlett, H.M., Kraydieh, S., Green, E.J., Dietrich, W.D., 1997. Temporal and regional pat-
Conflict of interest terns of axonal damage following traumatic brain injury: a beta-amyloid precursor
protein immunocytochemical study in rats. J. Neuropathol. Exp. Neurol. 56,
1132–1141.
No competing financial interests exist. Brody, D.L., Mac, D.C., Kessens, C.C., Yuede, C., Parsadanian, M., Spinner, M., Kim, E.,
Schwetye, K.E., Holtzman, D.M., Bayly, P.V., 2007. Electromagnetic controlled cortical
impact device for precise, graded experimental traumatic brain injury. J. Neurotrauma
Acknowledgment 24, 657–673.
Browne, K.D., Chen, X.H., Meaney, D.F., Smith, D.H., 2011. Mild traumatic brain injury and
This work was supported by Maryland Technology Development diffuse axonal injury in swine. J. Neurotrauma 28, 1747–1755.
Bu, G., 2009. Apolipoprotein E and its receptors in Alzheimer's disease: pathways,
Corporation funds to VEK. pathogenesis and therapy. Nat. Rev. Neurosci. 10, 333–344.
Carter, S.F., Scholl, M., Almkvist, O., Wall, A., Engler, H., Langstrom, B., Nordberg, A., 2012.
References Evidence for astrocytosis in prodromal Alzheimer disease provided by 11C-
deuterium-L-deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh
Akiyama, H., Barger, S., Barnum, S., Bradt, B., Bauer, J., Cole, G.M., Cooper, N.R., compound B and 18F-FDG. J. Nucl. Med. 53, 37–46.
Eikelenboom, P., Emmerling, M., Fiebich, B.L., Finch, C.E., Frautschy, S., Griffin, W.S., Cernak, I., Merkle, A.C., Koliatsos, V.E., Bilik, J.M., Luong, Q.T., Mahota, T.M., Xu, L.Y., Slack,
Hampel, H., Hull, M., Landreth, G., Lue, L., Mrak, R., Mackenzie, I.R., McGeer, P.L., N., Windle, D., Ahmed, F.A., 2011. The pathobiology of blast injuries and blast-induced
O'Banion, M.K., Pachter, J., Pasinetti, G., Plata-Salaman, C., Rogers, J., Rydel, R., Shen, neurotrauma as identified using a new experimental model of injury in mice.
Y., Streit, W., Strohmeyer, R., Tooyoma, I., Van Muiswinkel, F.L., Veerhuis, R., Neurobiol. Dis. 41, 538–551.
Walker, D., Webster, S., Wegrzyniak, B., Wenk, G., Wyss-Coray, T., 2000. Inflammation Chen, S., Pickard, J.D., Harris, N.G., 2003. Time course of cellular pathology after controlled
and Alzheimer's disease. Neurobiol. Aging 21, 383–421. cortical impact injury. Exp. Neurol. 182, 87–102.
Ballatore, C., Lee, V.M.Y., Trojanowski, J.Q., 2007. Tau-mediated neurodegeneration in Chidlow, G., Wood, J.P., Sarvestani, G., Manavis, J., Casson, R.J., 2009. Evaluation of Fluoro-
Alzheimer's disease and related disorders. Nat. Rev. Neurosci. 8, 663–672. Jade C as a marker of degenerating neurons in the rat retina and optic nerve. Exp. Eye
Banati, R.B., 2002. Visualising microglial activation in vivo. GLIA 40, 206–217. Res. 88, 426–437.
Barth, J.T., 2011. Defense and Veterans Brain Injury Center. Sports Concussion and Combat Conte, V., Uryu, K., Fujimoto, S., Yao, Y., Rokach, J., Longhi, L., Trojanowski, J.Q., Lee, V.M.Y.,
Blast Injury. McIntosh, T.K., Pratico, D., 2004. Vitamin E reduces amyloidosis and improves cogni-
Beaumont, A., Marmarou, A., Czigner, A., Yamamoto, M., Demetriadou, K., Shirotani, T., tive function in Tg2576 mice following repetitive concussive brain injury. J.
Marmarou, C., Dunbar, J., 1999. The impact-acceleration model of head injury: injury Neurochem. 90, 758–764.
severity predicts motor and cognitive performance after trauma. Neurol. Res. 21, Corsellis, J.A.N., Bruton, C.J., Freeman-Browne, D., 1973. The aftermath of boxing. Psychol.
742–754. Med. 3, 270–303.
448 L. Xu et al. / Experimental Neurology 275 (2016) 436–449

Creeley, C.E., Wozniak, D.F., Bayly, P.V., Olney, J.W., Lewis, L.M., 2004. Multiple episodes of Loane, D.J., Byrnes, K.R., 2010. Role of microglia in neurotrauma. Neurotherapeutics 7,
mild traumatic brain injury result in impaired cognitive performance in mice. Acad. 366–377.
Emerg. Med. 11, 809–819. Longhi, L., Saatman, K.E., Fujimoto, S., Raghupathi, R., Meaney, D.F., Davis, J., McMillan, A.,
Deford, S.M., Wilson, M.S., Rice, A.C., Clausen, T., Rice, L.K., Barabnova, A., Bullock, R., Conte, V., Laurer, H.L., Stein, S., Stocchetti, N., McIntosh, T.K., 2005. Temporal window
Hamm, R.J., 2002. Repeated mild brain injuries result in cognitive impairment in of vulnerability to repetitive experimental concussive brain injury. Neurosurgery 56,
B6C3F1 mice. J. Neurotrauma 19, 427–438. 364–373.
Donnelly, D.J., Popovich, P.G., 2008. Inflammation and its role in neuroprotection, axonal Marmarou, A., Foda, M.A.A., Vandenbrink, W., Campbell, J., Kita, H., Demetriadou, K., 1994.
regeneration and functional recovery after spinal cord injury. Exp. Neurol. 209, A new model of diffuse brain injury in rats. 1. Pathophysiology and biomechanics. J.
378–388. Neurosurg. 80, 291–300.
Dratviman-Storobinsky, O., Hasanreisoglu, M., Offen, D., Barhum, Y., Weinberger, D., Martinez, F.O., Sica, A., Mantovani, A., Locati, M., 2008. Macrophage activation and polar-
Goldenberg-Cohen, N., 2008. Progressive damage along the optic nerve following in- ization. Front. Biosci. 13, 453–461.
duction of crush injury or rodent anterior ischemic optic neuropathy in transgenic Martland, H.S., 1928. Punch drunk. JAMA 91, 1103–1107.
mice. Mol. Vis. 14, 2171–2179. McKee, A.C., Cantu, R.C., Nowinski, C.J., Hedley-Whyte, E.T., Gavett, B.E., Budson, A.E.,
Erlanger, D.M., Kutner, K.C., Barth, J.T., Barnes, R., 1999. Neuropsychology of sports-related Santini, V.E., Lee, H.S., Kubilus, C.A., Stern, R.A., 2009. Chronic traumatic encephalop-
head injury: dementia pugilistica to post concussion syndrome. Clin. Neuropsychol. athy in athletes: progressive tauopathy after repetitive head injury. J. Neuropathol.
13, 193–209. Exp. Neurol. 68, 709–735.
Ferrari, R., Hardy, J., Momeni, P., 2011. Frontotemporal dementia: from Mendelian genet- McPhail, L.T., Stirling, D.P., Tetzlaff, W., Kwiecien, J.M., Ramer, M.S., 2004. The contribution
ics towards genome wide association studies. J. Mol. Neurosci. 45, 500–515. of activated phagocytes and myelin degeneration to axonal retraction/dieback fol-
Flierl, M.A., Stahel, P.F., Beauchamp, K.M., Morgan, S.J., Smith, W.R., Shohami, E., 2009. lowing spinal cord injury. Eur. J. Neurosci. 20, 1984–1994.
Mouse closed head injury model induced by a weight-drop device. Nat. Protoc. 4, Millspaugh, 1937. Dementia pugilistica. U.S. Naval Med. Bull. 35, 297–303.
1328–1337. Morganti-Kossmann, M.C., Rancan, M., Stahel, P.F., Kossmann, T., 2002. Inflammatory re-
Foda, M.A.A., Marmarou, A., 1994. A new model of diffuse brain injury in rats. 2. Morpho- sponse in acute traumatic brain injury: a double-edged sword. Curr. Opin. Crit. Care 8,
logical characterization. J. Neurosurg. 80, 301–313. 101–105.
Fujita, M., Wei, E.P., Povlishock, J.T., 2012. Intensity- and interval-specific repetitive trau- Mouzon, B., Chaytow, H., Crynen, G., Bachmeier, C., Stewart, J., Mullan, M., Stewart, W.,
matic brain injury can evoke both axonal and microvascular damage. J. Neurotrauma Crawford, F., 2012. Repetitive mild traumatic brain injury in a mouse model produces
29, 2172–2180. learning and memory deficits accompanied by histological changes. J. Neurotrauma
Gama Sosa, M.A., De Gasperi, R., Janssen, P.L., Yuk, F.J., Anazodo, P.C., Pricop, P.E., Paulino, 29, 2761–2773.
A.J., Wicinski, B., Shaughness, M.C., Maudlin-Jeronimo, E., Hall, A.A., Dickstein, D.L., Neumann, H., Kotter, M.R., Franklin, R.J., 2009. Debris clearance by microglia: an essential
McCarron, R.M., Chavko, M., Hof, P.R., Ahlers, S.T., Elder, G.A., 2014. Selective vulner- link between degeneration and regeneration. Brain 132, 288–295.
ability of the cerebral vasculature to blast injury in a rat model of mild traumatic Ojo, J.O., Mouzon, B., Greenberg, M.B., Bachmeier, C., Mullan, M., Crawford, F., 2013. Repet-
brain injury. Acta Neuropathol. Commun. 2, 67. itive mild traumatic brain injury augments tau pathology and glial activation in aged
Gentleman, S.M., Leclercq, P.D., Moyes, L., Graham, D.I., Smith, C., Griffin, W.S., Nicoll, J.A., hTau mice. J. Neuropathol. Exp. Neurol. 72, 137–151.
2004. Long-term intracerebral inflammatory response after traumatic brain injury. Omalu, B.I., DeKosky, S.T., Minster, R.L., Kamboh, M.I., Hamilton, R.L., Wecht, C.H., 2005.
Forensic Sci. Int. 146, 97–104. Chronic traumatic encephalopathy in a National Football League player. Neurosur-
Goldstein, L.E., Fisher, A.M., Tagge, C.A., Zhang, X.L., Velisek, L., Sullivan, J.A., Upreti, C., gery 57, 128–133.
Kracht, J.M., Ericsson, M., Wojnarowicz, M.W., Goletiani, C.J., Maglakelidze, G.M., Omalu, B.I., DeKosky, S.T., Hamilton, R.L., Minster, R.L., Kamboh, M.I., Shakir, A.M., Wecht,
Casey, N., Moncaster, J.A., Minaeva, O., Moir, R.D., Nowinski, C.J., Stern, R.A., Cantu, C.H., 2006. Chronic traumatic encephalopathy in a national football league player:
R.C., Geiling, J., Blusztajn, J.K., Wolozin, B.L., Ikezu, T., Stein, T.D., Budson, A.E., part II. Neurosurgery 59, 1086–1092.
Kowall, N.W., Chargin, D., Sharon, A., Saman, S., Hall, G.F., Moss, W.C., Cleveland, Oppenheimer, D.R., 1968. Microscopic lesions in the brain following head injury. J. Neurol.
R.O., Tanzi, R.E., Stanton, P.K., McKee, A.C., 2012. Chronic traumatic encephalopathy Neurosurg. Psychiatry 31, 299–306.
in blast-exposed military veterans and a blast neurotrauma mouse model. Sci. Transl. Pohl, H.B., Porcheri, C., Mueggler, T., Bachmann, L.C., Martino, G., Riethmacher, D.,
Med. 4, 134ra60. Franklin, R.J., Rudin, M., Suter, U., 2011. Genetically induced adult oligodendrocyte
Greer, J.E., McGinn, M.J., Povlishock, J.T., 2011. Diffuse traumatic axonal injury in the cell death is associated with poor myelin clearance, reduced remyelination, and axo-
mouse induces atrophy, c-Jun activation, and axonal outgrowth in the axotomized nal damage. J. Neurosci. 31, 1069–1080.
neuronal population. J. Neurosci. 31, 5089–5105. Prewitt, C.M., Niesman, I.R., Kane, C.J., Houle, J.D., 1997. Activated macrophage/microglial
Hartman, R.E., Izumi, Y., Bales, K.R., Paul, S.M., Wozniak, D.F., Holtzman, D.M., 2005. Treat- cells can promote the regeneration of sensory axons into the injured spinal cord. Exp.
ment with an amyloid-beta antibody ameliorates plaque load, learning deficits, and Neurol. 148, 433–443.
hippocampal long-term potentiation in a mouse model of Alzheimer's disease. J. Rees, P.M., 2003. Contemporary issues in mild traumatic brain injury. Arch. Phys. Med.
Neurosci. 25, 6213–6220. Rehabil. 84, 1885–1894.
Hellerstein, L.F., Freed, S., Maples, W.C., 1995. Vision profile of patients with mild brain in- Schmued, L.C., Albertson, C., Slikker Jr., W., 1997. Fluoro-Jade: a novel fluorochrome for
jury. J. Am. Optom. Assoc. 66, 634–639. the sensitive and reliable histochemical localization of neuronal degeneration. Brain
Homsi, S., Piaggio, T., Croci, N., Noble, F., Plotkine, M., Marchand-Leroux, C., Jafarian- Res. 751, 37–46.
Tehrani, M., 2010. Blockade of acute microglial activation by minocycline promotes Schmued, L.C., Stowers, C.C., Scallet, A.C., Xu, L., 2005. Fluoro-Jade C results in ultra
neuroprotection and reduces locomotor hyperactivity after closed head injury in high resolution and contrast labeling of degenerating neurons. Brain Res. 1035,
mice: a twelve-week follow-up study. J. Neurotrauma 27, 911–921. 24–31.
Horn, K.P., Busch, S.A., Hawthorne, A.L., van, R.N., Silver, J., 2008. Another barrier to regen- Shitaka, Y., Tran, H.T., Bennett, R.E., Sanchez, L., Levy, M.A., Dikranian, K., Brody, D.L.,
eration in the CNS: activated macrophages induce extensive retraction of dystrophic 2011. Repetitive closed-skull traumatic brain injury in mice causes persistent
axons through direct physical interactions. J. Neurosci. 28, 9330–9341. multifocal axonal injury and microglial reactivity. J. Neuropathol. Exp. Neurol.
Hu, X., Li, P., Guo, Y., Wang, H., Leak, R.K., Chen, S., Gao, Y., Chen, J., 2012. Microglia/ 70, 551–567.
macrophage polarization dynamics reveal novel mechanism of injury expansion Siopi, E., Cho, A.H., Homsi, S., Croci, N., Plotkine, M., Marchand-Leroux, C., Jafarian-Tehrani,
after focal cerebral ischemia. Stroke 43, 3063–3070. M., 2011. Minocycline restores sAPPalpha levels and reduces the late histopatholog-
Jefferson, S.C., Tester, N.J., Howland, D.R., 2011. Chondroitinase ABC promotes recovery ical consequences of traumatic brain injury in mice. J. Neurotrauma 28, 2135–2143.
of adaptive limb movements and enhances axonal growth caudal to a spinal Siopi, E., Calabria, S., Plotkine, M., Marchand-Leroux, C., Jafarian-Tehrani, M., 2012a.
hemisection. J. Neurosci. 31, 5710–5720. Minocycline restores olfactory bulb volume and olfactory behavior after traumatic
Johnson, V.E., Stewart, J.E., Begbie, F.D., Trojanowski, J.Q., Smith, D.H., Stewart, W., 2013. brain injury in mice. J. Neurotrauma 29, 354–361.
Inflammation and white matter degeneration persist for years after a single traumatic Siopi, E., Llufriu-Daben, G., Fanucchi, F., Plotkine, M., Marchand-Leroux, C., Jafarian-
brain injury. Brain 136, 28–42. Tehrani, M., 2012b. Evaluation of late cognitive impairment and anxiety states fol-
Kelley, B.J., Lifshitz, J., Povlishock, J.T., 2007. Neuroinflammatory responses after experi- lowing traumatic brain injury in mice: the effect of minocycline. Neurosci. Lett. 511,
mental diffuse traumatic brain injury. J. Neuropathol. Exp. Neurol. 66, 989–1001. 110–115.
Koliatsos, V.E., Cernak, I., Xu, L., Song, Y., Savonenko, A., Crain, B.J., Eberhart, C.G., Smith, D.H., Chen, X.H., Xu, B.N., McIntosh, T.K., Gennarelli, T.A., Meaney, D.F., 1997. Char-
Frangakis, C.E., Melnikova, T., Kim, H., Lee, D., 2011. A mouse model of blast injury acterization of diffuse axonal pathology and selective hippocampal damage following
to brain: initial pathological, neuropathological, and behavioral characterization. J. inertial brain trauma in the pig. J. Neuropathol. Exp. Neurol. 56, 822–834.
Neuropathol. Exp. Neurol. 70, 399–416. Smith, D.H., Meaney, D.F., Shull, W.H., 2003. Diffuse axonal injury in head trauma. J. Head
Langlois, J.A., Rutland-Brown, W., Wald, M.M., 2006. The epidemiology and impact of Trauma Rehabil. 18, 307–316.
traumatic brain injury: a brief overview. J. Head Trauma Rehabil. 21, 375–378. Smith, C., Gentleman, S.M., Leclercq, P.D., Murray, L.S., Griffin, W.S., Graham, D.I., Nicoll,
Laurer, H.L., Bareyre, F.M., Lee, V.M.Y.C., Trojanowski, J.Q., Longhi, L., Hoover, R., Saatman, J.A., 2013. The neuroinflammatory response in humans after traumatic brain injury.
K.E., Raghupathi, R., Hoshino, S., Grady, M.S., McIntosh, T.K., 2001. Mild head injury Neuropathol. Appl. Neurobiol. 39, 654–666.
increasing the brain's vulnerability to a second concussive impact. J. Neurosurg. 95, Sosa, M.A., de Gasperi, R., Paulino, A.J., Pricop, P.E., Shaughness, M.C., Maudlin-Jeronimo, E.,
859–870. Hall, A.A., Janssen, W.G., Yuk, F.J., Dorr, N.P., Dickstein, D.L., McCarron, R.M., Chavko, M.,
Leung, C.K., Lindsey, J.D., Crowston, J.G., Lijia, C., Chiang, S., Weinreb, R.N., 2008. Hof, P.R., Ahlers, S.T., Elder, G.A., 2013. Blast overpressure induces shear-related inju-
Longitudinal profile of retinal ganglion cell damage after optic nerve crush ries in the brain of rats exposed to a mild traumatic brain injury. Acta Neuropathol.
with blue-light confocal scanning laser ophthalmoscopy. Invest. Ophthalmol. Commun. 1, 51.
Vis. Sci. 49, 4898–4902. Soto, I., Oglesby, E., Buckingham, B.P., Son, J.L., Roberson, E.D., Steele, M.R., Inman, D.M.,
Liao, B., Zhao, W., Beers, D.R., Henkel, J.S., Appel, S.H., 2012. Transformation from a neuro- Vetter, M.L., Horner, P.J., Marsh-Armstrong, N., 2008. Retinal ganglion cells downreg-
protective to a neurotoxic microglial phenotype in a mouse model of ALS. Exp. ulate gene expression and lose their axons within the optic nerve head in a mouse
Neurol. 237, 147–152. glaucoma model. J. Neurosci. 28, 548–561.
L. Xu et al. / Experimental Neurology 275 (2016) 436–449 449

Spain, A., Daumas, S., Lifshitz, J., Rhodes, J., Andrews, P.J., Horsburgh, K., Fowler, J.H., 2010. Wang, J., Hamm, R.J., Povlishock, J.T., 2011. Traumatic axonal injury in the optic nerve:
Mild fluid percussion injury in mice produces evolving selective axonal pathology evidence for axonal swelling, disconnection, dieback, and reorganization. J.
and cognitive deficits relevant to human brain injury. J. Neurotrauma 27, 1429–1438. Neurotrauma 28, 1185–1198.
Strittmatter, W.J., Saunders, A.M., Schmechel, D., Pericak-Vance, M., Enghild, J., Salvesen, Wang, J., Fox, M.A., Povlishock, J.T., 2013. Diffuse traumatic axonal injury in the optic
G.S., Roses, A.D., 1993. Apolipoprotein E: high-avidity binding to b-amyloid and in- nerve does not elicit retinal ganglion cell loss. J. Neuropathol. Exp. Neurol. 72,
creased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc. 768–781.
Natl. Acad. Sci. U. S. A. 90, 1977–1981. Warden, D., 2006. Military TBI during the Iraq and Afghanistan wars. J. Head Trauma
Summers, M.J., 2006. Increased inattentional blindness in severe traumatic brain injury: Rehabil. 21, 398–402.
evidence for reduced distractibility? Brain Inj. 20, 51–60. Xin, X., Rodrigues, M., Umapathi, M., Kashiwabuchi, F., Ma, T., Babapoor-Farrokhran, S.,
Turgut, M., Kaplan, S., Unal, B.Z., Bozkurt, M., Yuruker, S., Yenisey, C., Sahin, B., Uyanikgil, Wang, S., Hu, J., Bhutto, I., Welsbie, D.S., Duh, E.J., Handa, J.T., Eberhart, C.G., Lutty,
Y., Baka, M., 2010. Stereological analysis of sciatic nerve in chickens following neona- G., Semenza, G.L., Montaner, S., Sodhi, A., 2013. Hypoxic retinal Muller cells promote
tal pinealectomy: an experimental study. J. Brachial. Plex. Peripher. Nerve Inj. 5, 10. vascular permeability by HIF-1-dependent up-regulation of angiopoietin-like 4. Proc.
Tzekov, R., Quezada, A., Gautier, M., Biggins, D., Frances, C., Mouzon, B., Jamison, J., Mullan, Natl. Acad. Sci. U. S. A. 110, E3425–E3434.
M., Crawford, F., 2014. Repetitive mild traumatic brain injury causes optic nerve and Xu, L., Ryugo, D.K., Pongstaporn, T., Johe, K., Koliatsos, V.E., 2009. Human neural stem cell
retinal damage in a mouse model. J. Neuropathol. Exp. Neurol. 73, 345–361. grafts in the spinal cord of SOD1 transgenic rats: differentiation and structural inte-
Uryu, K., Laurer, H., McIntosh, T., Pratico, D., Martinez, D., Leight, S., Lee, V.M., Trojanowski, gration into the segmental motor circuitry. J. Comp. Neurol. 514, 297–309.
J.Q., 2002. Repetitive mild brain trauma accelerates Abeta deposition, lipid peroxida- Xu, H., Rosler, T.W., Carlsson, T., Bruch, J., de Andrade, A., Hollerhage, M., Oertel, W.H.,
tion, and cognitive impairment in a transgenic mouse model of Alzheimer amyloid- Hoglinger, G.U., 2014. Memory deficits correlate with tau and spine pathology in
osis. J. Neurosci. 22, 446–454. P301S MAPT transgenic mice. Neuropathol. Appl. Neurobiol. 40 (7), 833–843.
Vandrovcova, J., Anaya, F., Kay, V., Lees, A., Hardy, J., de Silva, R., 2010. Disentangling the Yamada, J., Jinno, S., 2013. Novel objective classification of reactive microglia following
role of the tau gene locus in sporadic tauopathies. Curr. Alzheimer Res. 7, 726–734. hypoglossal axotomy using hierarchical cluster analysis. J. Comp. Neurol. 521,
Wang, Y.P., Biernat, J., Pickhardt, M., Mandelkow, E., Mandelkow, E.M., 2007. Stepwise 1184–1201.
proteolysis liberates tau fragments that nucleate the Alzheimer-like aggregation of
full-length tau in a neuronal cell model. Proc. Natl. Acad. Sci. U. S. A. 104,
10252–10257.

You might also like