You are on page 1of 8

Brain, Behavior, and Immunity 60 (2017) 304–311

Contents lists available at ScienceDirect

Brain, Behavior, and Immunity


journal homepage: www.elsevier.com/locate/ybrbi

Full-length Article

Binge ethanol in adulthood exacerbates negative outcomes following


juvenile traumatic brain injury
Kate Karelina ⇑, Kristopher R. Gaier, Maya Prabhu, Vanessa Wenger, Timothy E.D. Corrigan, Zachary M. Weil
Department of Neuroscience, Group in Behavioral Neuroendocrinology and Center for Brain and Spinal Cord Repair, Ohio State University Wexner Medical Center, Columbus,
OH 43210, USA

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

Article history: Traumatic brain injuries (TBI) are a major public health problem with enormous costs in terms of health
Received 5 August 2016 care dollars, lost productivity, and reduced quality of life. Alcohol is bidirectionally linked to TBI as many
Received in revised form 4 October 2016 TBI patients are intoxicated at the time of their injury and we recently reported that, in accordance with
Accepted 8 November 2016
human epidemiological data, animals injured during juvenile development self-administered signifi-
Available online 12 November 2016
cantly more alcohol as adults than did sham injured mice. There are also clinical data that drinking after
TBI significantly reduces the efficacy of rehabilitation and leads to poorer long-term outcomes. In order to
Keywords:
determine whether juvenile traumatic brain injury also increased the vulnerability of the brain to the
Traumatic brain injury
Alcohol
toxic effects of high dose alcohol, mice were injured at 21 days of age and then seven weeks later treated
Axon degeneration daily with binge-like levels of alcohol 5 g/kg (by oral gavage) for ten days. Binge-like alcohol produced a
Microglia greater degree of neuronal damage and neuroinflammation in mice that sustained a TBI. Further, mice
Neuroinflammation that sustained a juvenile TBI exhibited mild learning and memory impairments in adulthood following
Learning and memory binge alcohol and express a significant increase in hippocampal ectopic localization of newborn neurons.
Neurogenesis Taken together, these data provide strong evidence that a mild brain injury occurring early in life renders
the brain highly vulnerable to the consequences of binge-like alcohol consumption.
Ó 2016 Elsevier Inc. All rights reserved.

1. Introduction TBI itself could serve as an independent risk factor for substance
abuse issues, particularly when injuries occur early in development
Traumatic brain injuries (TBI) are a major public health problem (Bjork and Grant, 2009; Corrigan et al., 2013; Weil et al., 2016a).
with millions of injuries and tens of thousands of deaths occurring Most epidemiological studies have reported that although there
annually in the US alone (Langlois et al., 2006). Further, the long- is a population of patients that abstain from alcohol after TBI there
term health consequences and economic costs in terms of health is also a substantial number that resume drinking in the months
spending and loss of productivity are staggering (Coronado et al., after injury (Bombardier et al., 2003; Kreutzer and Harris, 1990;
2012). Ponsford et al., 2007). There is some limited, though controversial,
Alcohol use is tightly linked to traumatic brain injuries as alco- evidence that intoxication at the time of TBI can be neuroprotec-
hol intoxication is linked to, by some estimates, more than half of tive (Berry et al., 2011; Chen et al., 2012; Opreanu et al., 2010;
all traumatic brain injuries (Tagliaferri et al., 2006). Much of the Pandit et al., 2014), but alcohol abuse following TBI is clearly prob-
research into this relationship therefore has focused on the role lematic as it produces significantly poorer outcomes including:
of alcohol as a risk factor for TBI, and intoxication at injury as a impairing the efficacy of rehabilitation programs; reducing voca-
variable in functional outcomes and recovery (Berry et al., 2011; tional opportunities; and substantially increasing the risk for psy-
Chen et al., 2012; Opreanu et al., 2010; Pandit et al., 2014). How- chiatric disorders, post-traumatic seizures, and additional injuries
ever, there is also some experimental and clinical evidence that (Corrigan, 1995; Corrigan et al., 2014; Jorge et al., 2005; Vaaramo
et al., 2014a,b).
Abbreviations: TBI, traumatic brain injury; DCX, doublecortin; Iba-1, ionized
Recently, we reported that female, but not male, mice that
calcium binding adaptor molecule 1; TLR4, toll like receptor 4; IL-1b, interleukin 1 received a single mild traumatic brain injury at 21 days of age
beta. self-administered significantly more alcohol in adulthood than
⇑ Corresponding author at: Biomedical Research Tower #618, 460 West 12th did sham-injured mice or those injured in adulthood (Weil et al.,
Ave., Columbus, OH 43210, USA.
2016b). These data revealed that injuries occurring at a critical
E-mail address: Ekaterina.karelina@osumc.edu (K. Karelina).

http://dx.doi.org/10.1016/j.bbi.2016.11.009
0889-1591/Ó 2016 Elsevier Inc. All rights reserved.
K. Karelina et al. / Brain, Behavior, and Immunity 60 (2017) 304–311 305

developmental time point in pre-adolescent mice resulted in long- 2.3. Alcohol administration
lasting consequences that contributed to voluntary alcohol abuse
in adulthood. Beginning seven weeks following TBI or sham injury, all mice
Although it is clear that drinking after traumatic brain injury were administered 5 g/kg of a 31.5% solution of ethanol or water
reduces the effectiveness of rehabilitation therapy and produces control (dosing based on (Bertola et al., 2013)). Ethanol or water
poorer outcomes, the precise mechanisms that link drinking to was administered via daily oral gavage for 10 consecutive days.
impaired neuronal recovery and function in this clinical population Mice were weighed daily to account for body mass fluctuations
are not fully understood. However, there is mounting evidence in the dosing. This paradigm resulted in 4 groups (sham/water,
from both the experimental and clinical literature that heavy or TBI/water, sham/ethanol, TBI/ethanol), which were divided into 3
binge-like drinking, even in the absence of other insults, induces separate cohorts. One cohort was used for assessment of patho-
neuroinflammation and degeneration (Alfonso-Loeches et al., physiology only (n = 8–10 per group), one for microglial extraction
2010; Crews et al., 2011; Hayes et al., 2013; Qin and Crews, (n = 6–8 per group), and another cohort underwent behavioral
2012; Vetreno and Crews, 2012). Further, TBI itself induces long testing followed by histological assessment (n = 9–12 per group).
lasting impairments in neuronal structure and function and also See Fig. 1 for experimental timeline.
induces neuroinflammatory responses. Additionally, inflammatory
events early in life appear to prime the neuroimmune system to 2.4. Blood ethanol concentration
exhibit greater inflammatory responses to other stimuli, such as
high-dose alcohol, later in life (Bilbo et al., 2010). Thus, heavy Blood ethanol concentration (BEC) was assessed via a colori-
drinking by TBI survivors may impair functional outcomes, at least metric 96-well plate assay (modified from Prencipe et al., 1987).
in part, by inducing inflammatory responses and further impairing Blood was drawn from the retro-orbital sinus 90 min after the final
already dysfunctional neuronal circuits. oral gavage. Blood samples were centrifuged and serum stored at
Our previous study using the self-administration paradigm 80 °C until use. A BEC assay buffer (100 mM KH2PO4; 100 mM
allowed us to investigate injury-induced alterations in alcohol K2HPO4; 0.7 mM 4-aminoantopyrine; 1.7 mM chromotropic acid
reward processing, but it did not allow us to investigate the func- disodium salt; 50 mg/L EDTA; 50 mL/L triton X-100) was used to
tional consequences of alcohol consumption after injury because dilute ethanol standards and serum and all wells were assessed
sham-injured mice did not voluntarily self-administer high doses in duplicate. A reaction mixture of alcohol oxidase (Sigma A2404)
of ethanol (Weil et al., 2016a). Thus, in order to determine whether and peroxidase from horseradish (Sigma P8375) diluted in BEC
heavy drinking after TBI exacerbates the behavioral, neuropatho- assay buffer was added to all wells and the plate was read at
logical, and inflammatory consequences of juvenile injuries in 600 nm.
female mice, we administered ten daily binge-like doses of ethanol
via oral gavage to adult females that had sustained a TBI or sham 2.5. Histology
injury in early life. The goals of this study were to 1) determine
whether juvenile TBI increased the functional deficits and tissue Axon damage was assessed via silver staining using the
damage induced by binge-like levels of alcohol and 2) to begin to NeuroSilver kit (FD Neurotechnologies, PK301) per manufacturer’s
uncover the mechanisms that link TBI, binge-like alcohol, and tis- instructions. Mice were overdosed with sodium pentobarbital
sue damage. We hypothesized that binge-like exposure to alcohol (200 mg/kg) and transcardially perfused with 4% paraformalde-
would exacerbate tissue damage and functional deficits in previ- hyde. Following an overnight postfix and cryopreservation, brain
ously injured female mice in part by inducing neuroinflammation sections throughout the forebrain were sliced on a cryostat,
and impairing neuroplasticity. (40 lm) and collected into a 24-well culture plate for free-
floating immunohistochemistry and silver staining. For immuno-
2. Methods histochemistry, tissue was washed with 0.1 M phosphate-
buffered saline, quenched in hydrogen peroxide, and incubated in
2.1. Animals primary antibody overnight (rabbit anti-Iba1, Wako 019-19741;
goat anti-doublecortin, Santa Cruz sc-8006). Biotinylated sec-
All procedures were conducted on female Swiss-Webster mice ondary antibodies (goat anti-rabbit and donkey anti-goat, Vector
derived from breeders purchased from Charles River (Wilmington, Laboratories BA-1000 and BA-5000) were applied the following
MA) and bred at the Ohio State University. Pups were weaned at day, and staining was visualized using the avidin biotin-
21 days of age into a standard mouse cage (32  16  12 cm) with diaminobenzidene staining method (Vector Laboratories PK-6100
ad libitum access to food and filtered tap water. All animals were and SK-4100).
housed in a 14:10 light-dark cycle. All procedures were approved
by the OSU Institutional Animal Care and Use Committee, and were 2.6. Histological analysis
conducted in accordance with the National Institute of Health
guidelines. Axonal degeneration (silver staining) was assessed qualitatively
as previously reported (Weil et al., 2014). Briefly, representative
2.2. Traumatic brain injury tissue throughout the entire forebrain were assessed and scored
on a 4-point scale (0 = few axonal profiles, 3 = dense axonal degen-
On the day of weaning, 21 day-old female mice sustained a mild eration throughout white matter tracts bilaterally).
closed-head injury or a sham injury as described previously (Weil Microglial cells (Iba1-positive staining) were assessed in the
et al., 2016b). All mice were anesthetized with inhaled isoflurane, prefrontal cortex. Morphological analysis was conducted using
secured in a stereotaxic frame, and a round 2 mm impactor (Impact Neurolucida software (MicroBrightfield). Briefly, 2–3 cells per
One device, Leica Biosystems, Richmond IL) was placed on the sur- hemisphere from each brain were selected using strict criteria as
face of the exposed skull ( 1 mm AP, 1 mm ML). TBI mice under- previously reported (Karelina et al., 2016) and traced for subse-
went an impact at 3 mm/s (dwell time 30 ms) to a depth of 1 mm, quent sholl analysis. Using NeuroExplorer (MicroBrightfield) con-
while sham mice were exposed to an equivalent amount of anes- centric sholl rings spaced 5 lm apart were centered on the cell
thesia in the absence of impact. The skin was closed with nylon body, and the number of intersections, total process length, and
suture and mice were returned to their home cages. cell body surface area were obtained for each cell. Total microglial
306 K. Karelina et al. / Brain, Behavior, and Immunity 60 (2017) 304–311

Fig. 1. Experimental timeline. All animals underwent TBI or sham injury at 21 days of age and were returned to their home cages for seven weeks. In adulthood, all mice were
administered ethanol or water via oral gavage daily for ten consecutive days. These studies were conducted on three cohorts, cohort 1 was perfused for tissue collection
immediately after the final gavage, cohort 2 was used for microglia isolation, and cohort 3 underwent behavioral testing before tissue collection. Passive avoidance testing
was begun twenty-four hours following the final gavage and brain tissue collection was conducted the day after testing was completed.

numbers were counted in a 0.26 mm2 region of interest using Ima- stepped into the dark chamber. Once the mouse entered the dark
geJ software (Abràmoff et al., 2004) and converted to cell number/ chamber, the door closed, a brief shock (1 mA, 2 s) was delivered
mm2. to the grid floor, and the mouse was immediately replaced into
Doublecortin-positive neurons were counted in the hippocam- its home cage. Twenty-four hours following the training, each
pus dentate gyrus, sections spanning approximately 1.94 mm mouse was again individually placed into the illuminated chamber
through 2.18 mm from bregma. Total cell counts were collected for 1 min. Once the guillotine door was open, latency to enter the
in the granule cell layer in both the top and bottom blades of the dark chamber was recorded (with an upper limit of 300 s). No
dentate, as well as the molecular layer and dentate hilus. shock was administered on testing day.

2.7. Microglial isolation 2.10. Statistical analysis

Microglia were extracted from bilateral frontal lobes of sham Qualitative analysis of silver staining was assessed via the non-
and injured mice 24 h following their final oral gavage. Fresh parametric Mann-Whitney U and Kruskall-Wallis tests. Significant
brains were collected, olfactory bulbs were removed and tissue overall Kruskall-Wallis results were followed up by pairwise com-
was dissected (approximately bregma 0.56 mm through parisons. Microglial sholl analysis was conducted using a repeated
3.56 mm) and homogenized in Dulbecco’s phosphate buffer solu- measures ANOVA. All histology analyses (cell counts, process
tion (DPBS). The homogenate was centrifuged and the pellet resus- length, cell body area), behavioral data, and gene expression data
pended in 70% isotonic Percoll (GE Healthcare Life Sciences #17- were assessed via a 2-way ANOVA (injury  ethanol administra-
0891-01). A discontinuous Percoll gradient was created by layering tion). All statistics were analyzed using the IBM SPSS V.23 soft-
50% and 35% Percoll and topping off with (DPBS). The gradient was ware. All significant overall ANOVA results were followed by the
then centrifuged and the microglia-containing layer in the 50%/70% Least Significant Differences posthoc test. Data were considered
interface was collected and washed. significant for p-values <0.05.

2.8. qPCR 3. Results

Extracted microglia were lysed and cDNA synthesized using the 3.1. Binge-like alcohol administration exacerbates TBI-associated
SuperScript III CellsDirect cDNA synthesis kit (Invitrogen pathology
#11739010 and #18080200) according to the manufacturer’s pro-
tocol. A TaqMan 18S ribosomal RNA primer/probe set (Applied Blood ethanol concentrations were assessed on the final day of
Biosystems #4319413E) was used as an internal control. Primer/ oral gavage (90 min following the gavage) and indicate a high level
probe sets for the target genes were also purchased at Applied of ethanol in the blood (0.4 g/dL) which did not differ between
Biosystems (TLR4: Mm00445273_m1; IL1b: Mm00434228_m1) sham and TBI mice (Fig. 2A). In order to determine the pathological
and amplification was performed on an ABI 7500 Sequencing Fast impact of high-dose alcohol exposure, we examined whether
System using the TaqMan Fast Advanced Master Mix (#4444963). binge-like alcohol levels further exacerbate axonal degeneration
The universal two-step RT-PCR cycling conditions used were: 50 °C in previously injured mice (Fig. 2B–C). As expected, TBI sustained
for 2 min, 95 °C for 20 s, followed by 40 cycles of 95 °C for 3 s and in pre-adolescence led to a significant degree of axonal degenera-
60 °C for 30 s. Gene expression was analyzed relative to a standard tion (U = 14.0, p < 0.05). However, a 10-day period of binge-like
curve and normalized to the sham/water group. alcohol administration in adulthood significantly exacerbated axon
damage in mice that had sustained a TBI as juveniles (H3 = 20.009,
2.9. Passive avoidance p < 0.05). The overall significant Kruskall-Wallis test was followed
up by pairwise comparisons which indicated significant differences
Learning and memory was evaluated via the passive avoidance between the sham/water and TBI/water groups, as well a signifi-
task. The task consisted of a 2-chambered testing environment cant increase in degeneration in the TBI/ethanol mice compared
(one chamber was illuminated and the other was dark) with a grid to TBI/water (all p < 0.05).
floor and guillotine door separating the chambers. On training day, We further examined microglial morphology as a measure of
mice were placed individually into the illuminated chamber for neuroinflammation. Brain tissue from all four groups was
1 min; the door was then lifted and remained open until the mouse processed for Iba1 immunohistochemistry. Sholl analysis of
K. Karelina et al. / Brain, Behavior, and Immunity 60 (2017) 304–311 307

Latency to enter the dark chamber on the training day was


recorded and indicates no significant difference between groups
(Fig. 5A). However, analysis of the latency to enter the dark cham-
ber (in which each mouse had previously endured a painful shock)
revealed an interaction of injury history and ethanol treatment
(F1,34 = 5.247, p < 0.05). These data indicate that the TBI/ethanol
group exhibited a significantly shorter latency to enter the
shock-associated chamber compared to the TBI/water group.
Importantly, this difference is not likely to be a result of increased
anxiety in the TBI/ethanol group as all mice had similar latencies to
escape the light chamber on the training day.

3.4. Binge alcohol and TBI interact to induce ectopic expression of


doublecortin-positive cells in the hippocampus

Given the learning/memory impairment observed in the TBI/


ethanol mice, we conducted an immunohistochemical analysis of
doublecortin in the hippocampus. Doublecortin (DCX) is tran-
siently expressed in progenitor cells committed to the neuronal
lineage and serves as an indirect marker of hippocampal neuroge-
nesis. DCX is expressed for approximately 2–3 weeks in newborn
neurons before it is replaced by mature neuronal markers
Fig. 2. Binge alcohol administration exacerbates TBI-associated pathology. A) Blood
(Brown et al., 2003), thus brain tissue fixed 13 days after the start
ethanol concentrations in sham and TBI mice 90 min after a gavage. B) Analysis of of the ethanol administration paradigm is likely to contain a sub-
silver staining in the forebrain white matter reveals that juvenile TBI-induced axon stantial subset of neurons developing during our experimental
degeneration is further exacerbated by repeated high dose administration of manipulation. Here, we assessed DCX-positive cells in the dentate
alcohol in adulthood. C) Representative staining in the genu of the corpus callosum
gyrus (Fig. 6). The total number of DCX-positive cells in the dentate
(bregma 0.50 mm). Scale bar = 50 lm, an asterisk (⁄) indicates statistically signif-
icant differences between indicated groups (p < 0.05). gyrus granule cell layer did not differ significantly by injury or
ethanol manipulations. However, we identified a significant
representative microglia in the prefrontal cortex revealed a increase in the ectopic expression of DCX-positive neurons in
significant effect of injury history on microglial morphology ethanol-administered TBI mice. Specifically, the TBI/ethanol group
(F1,141 = 194.942, p < 0.01) such that mice that had sustained a had significantly more DCX-positive neurons in the dentate hilus
TBI exhibited microglia with fewer sholl ring intersections (F1,33 = 7.129, p < 0.05) than any other group. Moreover, among
(reduced process complexity), indicative of an activated state mice that sustained a TBI, binge ethanol significantly increased
(Fig. 3A). TBI also significantly reduced overall microglial process DCX expression in the molecular layer (T14 = 2.420, p < 0.03).
length compared to sham injury (F3,145 = 3.346, p < 0.05; Fig. 3C).
An analysis of the total number of microglia in the prefrontal cor-
4. Discussion
tex revealed main effects of both injury (F1,29 = 6.294, p < 0.05) and
ethanol treatment (F1,29 = 80,959, p < 0.01), as well as an interac-
Binge-like levels of alcohol produce significant neuropathology,
tion (F1,29 = 8.382, p < 0.01) such that the ethanol-administered
neuroinflammation, and functional deficits in mice that had expe-
TBI mice exhibited the greatest number of cortical microglia
rienced a traumatic brain injury during juvenile development.
(Fig. 3D). Finally, a measure of microglial cell body area revealed
Specifically, alcohol administration led to greater axonal degener-
a main effect of injury (F1,27 = 8.879, p < 0.001), and an interaction
ation and microglial reactivity in mice that had been previously
(F1,27 = 5.427, p < 0.05) such that ethanol significantly increased
injured. Further, microglia isolated from previously injured ani-
cell body area in TBI mice compared to all other groups (Fig. 3E).
mals exhibited significantly increased gene expression for both
TLR4 and IL-1b following alcohol administration. The combination
3.2. Microglia from injured mice treated with alcohol show increased of binge-like levels of alcohol and TBI also impaired normal neuro-
pro-inflammatory gene expression genesis, resulting in increased ectopic expression of DCX-positive
cells in the hippocampus. Finally, the combination of high-dose
Given that both TBI and alcohol resulted in activated microglia, alcohol and juvenile TBI produced impairments in learning and
and the increased total number of microglia in the TBI/ethanol memory as assessed in the passive avoidance paradigm. Taken
group, we examined the microglia directly in order to determine together, these data indicate that a history of traumatic brain
whether the activational state corresponds to increased neuroin- injury significantly alters the neural and functional responses to
flammation. Both Toll-like receptor 4 (F3,16 = 3.516, p < 0.05; Fig. 4A) binge-like levels of alcohol and that the deleterious consequences
and interleukin-1 beta (F3,19 = 3.311, p < 0.05) gene expression were of high levels of alcohol in human populations may be related to
significantly increased in the TBI/ethanol group relative to all other inflammatory responses and impairments in neural plasticity.
groups assessed. Imaging and post-mortem studies have revealed pronounced
neuroinflammation and degeneration, particularly of white matter,
3.3. High doses of alcohol impair learning and memory in previously in the brains of alcoholics. Specifically, there is evidence of ongoing
injured mice inflammation, microglial activation, and cortical and subcortical
atrophy (Harper et al., 2003; He and Crews, 2008; Ikegami et al.,
Learning and memory was assessed via the passive avoidance 2003). The neurotoxicity of alcohol varies across the lifespan, but
test in all four groups beginning 24 h after the final ethanol (or is most prominent in cases of long term heavy drinking. Similarly,
water) administration day. This test takes advantage of the fear- axonal degeneration is a key pathological marker of TBI, however
motivated tendency of mice to avoid brightly illuminated areas. this pathology resolves over time following a mild injury. Here
308 K. Karelina et al. / Brain, Behavior, and Immunity 60 (2017) 304–311

Fig. 3. Binge alcohol and TBI alter microglial morphology. A) Sholl analysis in the prefrontal cortex reveals that both alcohol and TBI reduce microglial process complexity,
indicative of an activated state, as seen in B) representative images, scale bar = 50 lm. C) TBI also significantly reduced process length. D) The total number of microglia were
significantly increased in ethanol-treated mice that sustained a juvenile TBI. E) The cell body area of cortical microglia were also significantly increased in ethanol-treated TBI
mice. An asterisk (⁄) indicates statistically significant differences between the indicated groups (p < 0.05).

Fig. 4. TBI and alcohol administration increase proinflammatory gene expression. Relative mRNA gene expression is shown relative to the sham/water group and indicates
that A) TLR4 and B) IL-1b is exacerbated by alcohol in mice that underwent juvenile TBI. An asterisk (⁄) indicates statistically significant differences between the indicated
groups (p < 0.05).

we report that chronic exposure to binge-like levels of alcohol con- particularly when it occurs during development, has been shown
tribute to sustained axonal damage even when the drinking occurs to prime or sensitize the neuroimmune system to later inflamma-
many weeks after the actual injury. Importantly, the binge-like tory events. For instance, neonatal E. coli infection enhances the
alcohol administration paradigm used in this study was not suffi- cognitive deficits and microglial responses to lipopolysaccharide-
cient to induce axonal degeneration in mice that had not experi- induced inflammation later in life (Bilbo et al., 2008, 2010). Fur-
enced a prior TBI; rather TBI renders axons vulnerable to the ther, TBI itself can act as a priming stimulus as microglial reactivity
toxic effects of high dose alcohol. and behavioral depression induced by experimental inflammation
Mild traumatic brain injury, such as that used in the current is enhanced in mice with a history of mild traumatic brain injury
study, is characterized by a significant but transient inflammatory (Fenn et al., 2014). A primary goal of the work presented here
response and a number of cognitive and emotional disruptions that was to determine whether an early life brain injury could similarly
occur in the acute phase following a TBI typically improve as the potentiate the consequences of repeated high dose alcohol expo-
pathophysiology resolves. However, even mild neuroinflammation, sure in adulthood.
K. Karelina et al. / Brain, Behavior, and Immunity 60 (2017) 304–311 309

Fig. 5. Learning and memory are mildly impaired in alcohol-treated mice that sustained a juvenile TBI. A) All mice exhibit a similar latency to enter the dark side of the
passive avoidance chamber during training. B) However, the latency to re-enter the dark, shock-associated, side 24 h later was considerably longer, but was significantly
reduced in alcohol-treated mice that sustained a juvenile TBI. An asterisk (⁄) indicates statistically significant differences between the indicated groups (p < 0.05).

Fig. 6. Binge alcohol increases the number of ectopic doublecortin-positive cells in the hippocampus of mice that sustained a juvenile TBI. A) While the number of total DCX-
positive cells did not differ in the granule cell layer, high doses of alcohol significantly increased the number of ectopic newborn neurons in the B) hilus and C) molecular cell
layers of the hippocampus (white arrows). D) Representative images of the dentate gyrus, scale bar = 100 lm. An asterisk (⁄) indicates statistically significant differences
between the indicated groups (p < 0.05). ML = molecular layer, GCL = granule cell layer.
310 K. Karelina et al. / Brain, Behavior, and Immunity 60 (2017) 304–311

Neuroinflammatory responses are bidirectionally linked to alco- indicating that any acute effects of TBI on this cell population
hol abuse as neuroinflammation can increase drinking behavior had resolved by adulthood.
(Crews et al., 2011). Conversely, chronic exposure to high levels CNS trauma often leads to an increase in the proliferation rate of
of alcohol is neuroinflammatory as alcohol can activate glial cells neuronal precursors, and this phenomenon has been explored as a
and promote the expression of cytokine and chemokine molecules potential replacement source for cells lost to trauma (Dash et al.,
(He and Crews, 2008). Although the mechanisms of alcohol- 2001; Kernie and Parent, 2010). However, this strategy is compli-
induced neuroinflammation are incompletely understood, alcohol cated by reactive and aberrant neurogenesis that can occur after
does appear to signal in part through toll-like receptors (Alfonso- injury, seizure, or following withdrawal from heavy drinking which
Loeches et al., 2010; Fernandez-Lizarbe et al., 2009). For instance, can render the brain more vulnerable to cognitive dysfunction,
the long term behavioral and neurochemical effects of adolescent excitotoxicity, and additional seizures (Parent et al., 2006). Ectopic
alcohol exposure require TLR4 (Montesinos et al., 2016). granule cells that have been identified in the brains of epileptic
Microglia isolated from the injured frontal lobes following the animals and humans exhibit abnormal morphology, connectivity
binge-like administration of alcohol exhibited marked upregula- and electrophysiological properties that contribute to hyperex-
tion of mRNAs encoding the genes toll-like receptor 4 and IL-1b. citability (Parent et al., 2006; Scharfman et al., 2000). Additionally,
Critically, this increase in expression of TLR4 and IL-1b only alcohol (and alcohol withdrawal) even in the absence of trauma
occurred in mice that had been previously injured and treated with modulates neurotransmission and generally biases towards excita-
binge-like levels of ethanol in adulthood. We have hypothesized tory neurotransmission via changes in neuronal and glial neuro-
that TBI sets up a vicious cycle wherein TBI during development transmitter metabolism, transport, and receptor expression
primes inflammatory responses to exhibit exaggerated inflamma- (Adermark and Bowers, In Press; Dodd et al., 2000). Clinically,
tory responses to alcohol, which in turn promotes increased drink- TBI patients at high risk for post-traumatic epilepsy are advised
ing (Weil et al., 2016a). As an extension of this theory, the to abstain from alcohol as drinking is positively correlated with
neurotoxic effects of juvenile TBI could also be exacerbated by high the occurrence of seizures (Centers for Disease Control and
doses of alcohol. The up-regulation of both TLR4, a potential medi- Prevention; Corrigan, 1995). The data collected here suggest that
ator of alcohol-induced inflammation, and IL-1b, a key proinflam- an increased propensity for alcohol-induced ectopic neurogenesis
matory cytokine, only in mice that had been injured and exposed in the previously injured brain may at least in part, underlie this
to high levels of alcohol would tend to support this hypothesis. It relationship. Further, we cannot rule out the possibility that alco-
remains unspecified why we did not detect changes in gene hol or alcohol-withdrawal induced seizures may also have
expression in microglia from sham-injured mice although this lar- occurred and underlies the ectopic neurogenesis rather than the
gely agrees with our histological evidence including reduced other way around.
microglial numbers, activated morphology and axonal The behavioral results reported here, namely relatively subtle
degeneration. deficits in simple associative learning and memory in the passive
In the current study, we report exacerbation of the TBI-induced avoidance paradigm, were conducted on animals experiencing
damage to white matter tracts by binge-like levels of alcohol in alcohol withdrawal (retention trials occurred approximately 48 h
accordance with increased numbers and activational state of after their final dose of alcohol). Importantly, although there were
microglia as well as exaggerated inflammatory gene expression statistical differences between alcohol-and water-treated injured
profiles. Given that much of the tissue damage induced by chronic mice there were not statistical differences between the sham
exposure to high levels of alcohol appears to require microglial- injured and TBI groups. In any case, we elected to test animals dur-
mediated inflammatory responses (Boyadjieva and Sarkar, 2010; ing withdrawal because we wanted to avoid any direct effects of
Chastain and Sarkar, 2014; Yang et al., 2014), we conclude that a intoxication on behavioral outcomes. Additionally, it will be
transient period of TBI-induced inflammation in juvenile develop- important in future investigations to determine whether the mild
ment is sufficient to significantly exacerbate the neurotoxic effects impairments in learning and memory persist or improve beyond
of binge alcohol in adulthood. the acute phase of alcohol withdrawal. Given the enhanced axonal
Interestingly we report a differential effect of TBI and alcohol degeneration some degree of long-term functional deficit is likely
exposure on microglial morphology, counts and gene expression and may require more extensive behavioral characterization. In
profiles. Specifically, both TBI and alcohol exposure reduced the any case, these data provide further compelling evidence that
length of microglial processes as assessed by Sholl analysis but binge-like alcohol consumption negatively impacts functional out-
only microglia from mice that were exposed to alcohol and were comes in animals that were previously injured. Importantly, over
previously injured were more numerous, had larger cell bodies the 10-day treatment paradigm, mice were administered a single
and upregulated TLR4 and IL1b. That microglial morphology does daily high dose of alcohol every 24 h, thus in effect these animals
not fully track gene expression profiles is consistent with previous experienced repeated cycles of intoxication and withdrawal over
reports (McClain et al., 2011) and suggests that both alcohol expo- a short period of time. In addition, although we did not assess
sure and TBI are independently associated with changes in micro- the estrous cycle, the prolonged alcohol exposure paradigm did
glial physiology and morphology but that can induce a fuller presumably span at least two cycles. Finally, the paradigm we uti-
activation when combined. lized produced high (but similar between sham-injured and
Moreover, exposure to binge-like levels of alcohol greatly injured mice) blood alcohol concentrations. Such a high dose of
increased the presence of immature doublecortin-positive neurons alcohol was utilized in order to evaluate whether neurotoxic doses
in the non-neurogenic molecular layer and hilus of the dentate would produce worse outcomes in the previously injured brain,
gyrus but did so only in mice that had experienced TBI. Impor- however, it remains unclear whether a longer exposure to more
tantly, the exposure to binge-like alcohol in the current study moderate levels of alcohol would also be deleterious.
occurred more than six weeks after the TBI, thus the DCX-
positive cells assessed at this time point were those cells that were 5. Conclusions
born around the time of alcohol exposure rather than at the time of
injury. Although the combination of injury and alcohol-exposure The human epidemiological literature has clearly demonstrated
increased the number of hilar DCX-positive cells, the total number that 1) the TBI patient population consists of a high proportion of
was relatively small. Further there were no differences in the num- heavy and binge drinkers, 2) some percentage of TBI patients con-
ber or localization of DCX-positive cells induced by injury alone tinue to drink at high levels after TBI, and critically 3) that drinking
K. Karelina et al. / Brain, Behavior, and Immunity 60 (2017) 304–311 311

after TBI impairs functional and occupational recovery and puts the Crews, F.T., Zou, J., Qin, L., 2011. Induction of innate immune genes in brain create
the neurobiology of addiction. Brain Behav. Immun. 25, S4–S12.
patient at risk for serious complications including additional brain
Dash, P.K., Mach, S.A., Moore, A.N., 2001. Enhanced neurogenesis in the rodent
injuries and seizures. Here we begin to demonstrate the underlying hippocampus following traumatic brain injury. J. Neurosci. Res. 63, 313–319.
neurobiological mechanisms that underlie this phenomenon Dodd, P.R., Beckmann, A.M., Davidson, M.S., Wilce, P.A., 2000. Glutamate-mediated
including alcohol-induced axonal degeneration, ectopic neurogen- transmission, alcohol, and alcoholism. Neurochem. Int. 37, 509–533.
Fenn, A.M., Gensel, J.C., Huang, Y., Popovich, P.G., Lifshitz, J., Godbout, J.P., 2014.
esis, and enhanced neuroinflammation induced by alcohol in pre- Immune activation promotes depression 1 month after diffuse brain injury: a
viously injured mice. This vulnerability to alcohol-induced role for primed microglia. Biol. Psychiatry 76, 575–584.
histopathology and functional impairment occurred despite a long Fernandez-Lizarbe, S., Pascual, M., Guerri, C., 2009. Critical role of TLR4 response in
the activation of microglia induced by ethanol. J. Immunol. 183, 4733–4744.
delay (more than 6 weeks) between the injury and the beginning of Harper, C., Dixon, G., Sheedy, D., Garrick, T., 2003. Neuropathological alterations in
binge-alcohol administration. These data reinforce the importance alcoholic brains. Studies arising from the New South Wales Tissue Resource
of targeting substance abuse in this population and are of even Centre. Prog. Neuropsychopharmacol. Biol. Psychiatry 27, 951–961.
Hayes, D.M., Deeny, M.A., Shaner, C.A., Nixon, K., 2013. Determining the threshold
more importance given our previous observation that TBI can also for alcohol-induced brain damage: new evidence with gliosis markers. Alcohol.
increase alcohol self-administration and reward. Clin. Exp. Res. 37, 425–434.
He, J., Crews, F.T., 2008. Increased MCP-1 and microglia in various regions of the
human alcoholic brain. Exp. Neurol. 210, 349–358.
Acknowledgments Ikegami, Y., Goodenough, S., Inoue, Y., Dodd, P.R., Wilce, P.A., Matsumoto, I., 2003.
Increased TUNEL positive cells in human alcoholic brains. Neurosci. Lett. 349,
201–205.
This work was supported by an OSU Neurological Institute Pilot Jorge, R.E., Starkstein, S.E., Arndt, S., Moser, D., Crespo-Facorro, B., Robinson, R.G.,
Award and the National Institutes of Health (NS045758). The 2005. Alcohol misuse and mood disorders following traumatic brain injury.
authors gratefully acknowledge Joseph Abraham, Katie Centner, Arch. Gen. Psychiatry 62, 742–749.
Karelina, K., Sarac, B., Freeman, L.M., Gaier, K.R., Weil, Z.M., 2016. Traumatic brain
Samuel Nicholson, Benjamin Sarac, and Lauren Martin for their injury and obesity induce persistent central insulin resistance. Eur. J. Neurosci.
assistance and support on these experiments. We also thank Dr. 43, 1034–1043.
Kristina Kigerl for guidance through the microglial extraction pro- Kernie, S.G., Parent, J.M., 2010. Forebrain neurogenesis after focal Ischemic and
traumatic brain injury. Neurobiol. Dis. 37, 267–274.
tocol and Dr. Jacqueline Barker for assistance with the BEC assay. Kreutzer, J.S., Harris, J., 1990. Model systems of treatment for alcohol abuse
following traumatic brain injury. Brain Inj. 4, 1–5.
Langlois, J.A., Rutland-Brown, W., Wald, M.M., 2006. The epidemiology and impact
References of traumatic brain injury: a brief overview. J. Head Trauma Rehabil. 21, 375–
378.
Abràmoff, M.D., Magalhães, P.J., Ram, S.J., 2004. Image processing with ImageJ. McClain, J.A., Morris, S.A., Deeny, M.A., Marshall, S.A., Hayes, D.M., Kiser, Z.M., Nixon,
Biophotonics Int. 11, 36–42. K., 2011. Adolescent binge alcohol exposure induces long-lasting partial
Adermark, L., Bowers, M.S., In Press. Disentangling the Role of Astrocytes in Alcohol activation of microglia. Brain Behav. Immun. 25, S120–S128.
Use Disorder. Alcohol. Clin. Exp. Res. Montesinos, J., Pascual, M., Rodriguez-Arias, M., Minarro, J., Guerri, C., 2016.
Alfonso-Loeches, S., Pascual-Lucas, M., Blanco, A.M., Sanchez-Vera, I., Guerri, C., Involvement of TLR4 in the long-term epigenetic changes, rewarding and
2010. Pivotal role of TLR4 receptors in alcohol-induced neuroinflammation and anxiety effects induced by intermittent ethanol treatment in adolescence. Brain
brain damage. J. Neurosci. 30, 8285–8295. Behav. Immun. 53, 159–171.
Berry, C., Ley, E.J., Margulies, D.R., Mirocha, J., Bukur, M., Malinoski, D., Salim, A., Opreanu, R.C., Kuhn, D., Basson, M.D., 2010. Influence of alcohol on mortality in
2011. Correlating the blood alcohol concentration with outcome after traumatic traumatic brain injury. J. Am. Coll. Surg. 210, 997–1007.
brain injury: too much is not a bad thing. Am. Surg. 77, 1416–1419. Pandit, V., Patel, N., Rhee, P., Kulvatunyou, N., Aziz, H., Green, D.J., O’Keeffe, T.,
Bertola, A., Mathews, S., Ki, S.H., Wang, H., Gao, B., 2013. Mouse model of chronic Zangbar, B., Tang, A., Gries, L., Friese, R.S., Joseph, B., 2014. Effect of alcohol in
and binge ethanol feeding (the NIAAA model). Nat. Protoc. 8, 627–637. traumatic brain injury: is it really protective? J. Surg. Res. 190, 634–639.
Bilbo, S.D., Barrientos, R.M., Eads, A.S., Northcutt, A., Watkins, L.R., Rudy, J.W., Maier, Parent, J.M., Elliott, R.C., Pleasure, S.J., Barbaro, N.M., Lowenstein, D.H., 2006.
S.F., 2008. Early-life infection leads to altered BDNF and IL-1beta mRNA Aberrant seizure-induced neurogenesis in experimental temporal lobe epilepsy.
expression in rat hippocampus following learning in adulthood. Brain Behav. Ann. Neurol. 59, 81–91.
Immun. 22, 451–455. Ponsford, J., Whelan-Goodinson, R., Bahar-Fuchs, A., 2007. Alcohol and drug use
Bilbo, S.D., Wieseler, J.L., Barrientos, R.M., Tsang, V., Watkins, L.R., Maier, S.F., 2010. following traumatic brain injury: a prospective study. Brain Inj. 21, 1385–1392.
Neonatal bacterial infection alters fever to live and simulated infections in Prencipe, L., Iaccheri, E., Manzati, C., 1987. Enzymic ethanol assay: a new
adulthood. Psychoneuroendocrinology 35, 369–381. colorimetric method based on measurement of hydrogen peroxide. Clin.
Bjork, J.M., Grant, S.J., 2009. Does traumatic brain injury increase risk for substance Chem. 33, 486–489.
abuse? J. Neurotrauma 26, 1077–1082. Qin, L., Crews, F.T., 2012. Chronic ethanol increases systemic TLR3 agonist-induced
Bombardier, C.H., Temkin, N.R., Machamer, J., Dikmen, S.S., 2003. The natural history neuroinflammation and neurodegeneration. J. Neuroinflammation 9, 130.
of drinking and alcohol-related problems after traumatic brain injury. Arch. Scharfman, H.E., Goodman, J.H., Sollas, A.L., 2000. Granule-like neurons at the hilar/
Phys. Med. Rehabil. 84, 185–191. CA3 border after status epilepticus and their synchrony with area CA3
Boyadjieva, N.I., Sarkar, D.K., 2010. Role of microglia in ethanol’s apoptotic action on pyramidal cells: functional implications of seizure-induced neurogenesis. J.
hypothalamic neuronal cells in primary cultures. Alcohol. Clin. Exp. Res. 34, Neurosci. 20, 6144–6158.
1835–1842. Tagliaferri, F., Compagnone, C., Korsic, M., Servadei, F., Kraus, J., 2006. A systematic
Brown, J.P., Couillard-Despres, S., Cooper-Kuhn, C.M., Winkler, J., Aigner, L., Kuhn, H. review of brain injury epidemiology in Europe. Acta Neurochir. (Wien) 148,
G., 2003. Transient expression of doublecortin during adult neurogenesis. J. 255–268. discussion 268.
Comp. Neurol. 467, 1–10. Vaaramo, K., Puljula, J., Tetri, S., Juvela, S., Hillbom, M., 2014a. Head trauma
Centers for Disease Control and Prevention, Facts about Concussion and Brain sustained under the influence of alcohol is a predictor for future traumatic brain
Injury: Where to get Help. <http://www.cdc.gov/headsup/pdfs/providers/facts_ injury: a long-term follow-up study. Eur. J. Neurol. 21, 293–298.
about_concussion_tbi-a.pdf>. Vaaramo, K., Puljula, J., Tetri, S., Juvela, S., Hillbom, M., 2014b. Predictors of new-
Chastain, L.G., Sarkar, D.K., 2014. Role of microglia in regulation of ethanol onset seizures: a 10-year follow-up of head trauma subjects with and without
neurotoxic action. Int. Rev. Neurobiol. 118, 81–103. traumatic brain injury. J. Neurol. Neurosurg. Psychiatry 85, 598–602.
Chen, C.M., Yi, H.-Y., Yoon, Y.-H., Dong, C., 2012. Alcohol use at time of injury and Vetreno, R.P., Crews, F.T., 2012. Adolescent binge drinking increases expression of
survival following traumatic brain injury: results from the National Trauma the danger signal receptor agonist HMGB1 and Toll-like receptors in the adult
Data Bank. J. Stud. Alcohol Drugs 73, 531–541. prefrontal cortex. Neuroscience 226, 475–488.
Coronado, V.G., McGuire, L.C., Sarmiento, K., Bell, J., Lionbarger, M.R., Jones, C.D., Weil, Z.M., Corrigan, J.D., Karelina, K., 2016a. Alcohol abuse after traumatic brain
Geller, A.I., Khoury, N., Xu, L., 2012. Trends in Traumatic Brain Injury in the U.S. injury: experimental and clinical evidence. Neurosci. Biobehav. Rev. 62, 89–99.
and the public health response: 1995–2009. J. Safety Res. 43, 299–307. Weil, Z.M., Gaier, K.R., Karelina, K., 2014. Injury timing alters metabolic,
Corrigan, J.D., 1995. Substance abuse as a mediating factor in outcome from inflammatory and functional outcomes following repeated mild traumatic
traumatic brain injury. Arch. Phys. Med. Rehabil. 76, 302–309. brain injury. Neurobiol. Dis. 70, 108–116.
Corrigan, J.D., Bogner, J., Mellick, D., Bushnik, T., Dams-O’Connor, K., Hammond, F. Weil, Z.M., Karelina, K., Gaier, K.R., Corrigan, T.E., Corrigan, J.D., 2016b. Juvenile
M., Hart, T., Kolakowsky-Hayner, S., 2013. Prior history of traumatic brain injury traumatic brain injury increases alcohol consumption and reward in female
among persons in the traumatic brain injury model systems national database. mice. J. Neurotrauma 33, 895–903.
Arch. Phys. Med. Rehabil. 94, 1940–1950. Yang, J.-Y., Xue, X., Tian, H., Wang, X.-X., Dong, Y.-X., Wang, F., Zhao, Y.-N., Yao, X.-C.,
Corrigan, J.D., Cuthbert, J.P., Harrison-Felix, C., Whiteneck, G.G., Bell, J.M., Miller, A. Cui, W., Wu, C.-F., 2014. Role of microglia in ethanol-induced
C., Coronado, V.G., Pretz, C.R., 2014. US population estimates of health and social neurodegenerative disease: pathological and behavioral dysfunction at
outcomes 5 years after rehabilitation for traumatic brain injury. J. Head Trauma different developmental stages. Pharmacol. Ther. 144, 321–337.
Rehabil. 29, E1–E9.

You might also like