You are on page 1of 11

Brain, Behavior, and Immunity 26 (2012) 1191–1201

Contents lists available at SciVerse ScienceDirect

Brain, Behavior, and Immunity


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

Invited Review

Neuroinflammation after traumatic brain injury: Opportunities


for therapeutic intervention
Alok Kumar, David J. Loane ⇑
Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS,
University of Maryland School of Medicine, Baltimore, MD, United States

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

Article history: Traumatic brain injury (TBI) remains one of the leading causes of mortality and morbidity worldwide, yet
Received 5 April 2012 despite extensive efforts to develop neuroprotective therapies for this devastating disorder there have
Received in revised form 27 May 2012 been no successful outcomes in human clinical trials to date. Following the primary mechanical insult
Accepted 14 June 2012
TBI results in delayed secondary injury events due to neurochemical, metabolic and cellular changes that
Available online 21 June 2012
account for many of the neurological deficits observed after TBI. The development of secondary injury
represents a window of opportunity for therapeutic intervention to prevent progressive tissue damage
Keywords:
and loss of function after injury. To establish effective neuroprotective treatments for TBI it is essential
Traumatic brain injury
Neuroinflammation
to fully understand the complex cellular and molecular events that contribute to secondary injury. Neur-
Neuroprotection oinflammation is well established as a key secondary injury mechanism after TBI, and it has been long
Microglia considered to contribute to the damage sustained following brain injury. However, experimental and
Chronic neurodegeneration clinical research indicates that neuroinflammation after TBI can have both detrimental and beneficial
effects, and these likely differ in the acute and delayed phases after injury. The key to developing future
anti-inflammatory based neuroprotective treatments for TBI is to minimize the detrimental and neuro-
toxic effects of neuroinflammation while promoting the beneficial and neurotrophic effects, thereby cre-
ating optimal conditions for regeneration and repair after injury. This review outlines how post-traumatic
neuroinflammation contributes to secondary injury after TBI, and discusses the complex and varied
responses of the primary innate immune cells of the brain, microglia, to injury. In addition, emerging
experimental anti-inflammatory and multipotential drug treatment strategies for TBI are discussed, as
well as some of the challenges faced by the research community to translate promising neuroprotective
drug treatments to the clinic.
Ó 2012 Elsevier Inc. All rights reserved.

1. Introduction increase as a result of widespread motor vehicle use (Maas et al.,


2008).
Traumatic brain injury (TBI) is associated with significant mor- TBI is a highly complex disorder that is caused by both primary
bidity and mortality; this devastating disorder has substantial di- and secondary injury mechanisms (Loane and Faden, 2010;
rect, and indirect, costs to society. The Center for Disease Control McIntosh et al., 1996). Primary injury mechanisms result from
and Prevention (CDC) estimate that more than 1.7 million individ- the mechanical damage that occurs at the time of trauma to neu-
uals in the United States suffer a TBI annually (Faul et al., 2010). rons, axons, glia and blood vessels as a result of shearing, tearing
These numbers, however, greatly underestimate the real incidence, or stretching. Collectively, these effects induce secondary injury
and costs, of TBI as the CDC data does not include reports of sports- mechanisms that evolve over minutes to days and even months
related concussions or repeated mild TBI from military conflict after the initial traumatic insult and result from delayed neuro-
zones. Globally, the incidence of TBI is also increasing, particularly chemical, metabolic and cellular changes. These secondary injury
in developing countries where road traffic accidents are on the events are thought to account for the development of many of
the neurological deficits observed after TBI (McIntosh et al.,
1996), and their delayed nature suggests that there is a window
for therapeutic intervention (pharmacological or other) to prevent
⇑ Corresponding author. Address: Department of Anesthesiology and Center for progressive tissue damage and improve functional recovery after
Shock, Trauma & Anesthesiology Research (STAR), University of Maryland School of injury.
Medicine, 655, West Baltimore Street, Suite #6-011, Baltimore, MD 21201, United Secondary injury mechanisms include a wide variety of
States. Tel.: +1 410 706 5188.
processes such as depolarizations and disturbances of ionic
E-mail address: dloane@anes.umm.edu (D.J. Loane).

0889-1591/$ - see front matter Ó 2012 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.bbi.2012.06.008
1192 A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201

homeostasis (Gentile and McIntosh, 1993), release of neurotrans- Trauma to the brain results in rupture of the BBB, enabling
mitters (e.g. glutamate excitotoxicity) (Faden et al., 1989), mito- recruitment of circulating neutrophils, macrophages and lympho-
chondrial dysfunction (Xiong et al., 1997), neuronal apoptosis cytes to the injured site. The accumulation of blood-born immune
(Yakovlev et al., 1997), lipid degradation (Hall et al., 2004), and ini- cells within the brain parenchyma has been reported in human TBI
tiation of inflammatory and immune responses (Morganti-Koss- as well as animal models of brain trauma (Morganti-Kossmann
mann et al., 2007), among others. These neurochemical events et al., 2001). These cells release inflammatory mediators that mobi-
generate a host of toxic and pro-inflammatory molecules such as lize glia and immune cells to the site of injury. In addition to the
prostaglandins, oxidative metabolites, chemokines and pro-inflam- infiltration of immune cells, the activation of resident microglia
matory cytokines, which lead to lipid peroxidation, blood–brain play a major role in the response to brain injury (Loane and Byrnes,
barrier (BBB) disruption and the development of cerebral edema. 2010). Elegant in vivo two-photon microscopy imaging studies of
The associated increase in intracranial pressure can contribute to fluorescently labeled microglia following a laser-induced injury
local hypoxia and ischemia, secondary hemorrhage and herniation demonstrated rapid proliferation and movement of ramified
and additional neuronal cell death via necrotic and apoptotic microglial cells to the site of injury in response to extracellular
mechanisms (McIntosh et al., 1996). Although each secondary in- ATP released by the injured tissue (Davalos et al., 2005; Haynes
jury mechanism is often considered to be a distinct event, many et al., 2006). The microglial processes then fused to form an area
are highly interactive and may occur in parallel. of containment between healthy and injured tissues, suggesting
Considerable research efforts have sought to elucidate second- that microglia may represent the first line of defense following
ary injury mechanisms in order to develop neuroprotective treat- traumatic injury (Davalos et al., 2005). However, when microglia
ments. Although preclinical studies have suggested many become over-activated or reactive they can induce detrimental
promising pharmacological treatments, more than 30 phase III pro- neurotoxic effects by releasing multiple cytotoxic substances,
spective clinical trials have failed to show significance for their pri- including pro-inflammatory cytokines (e.g. interleukin (IL)-1b, tu-
mary endpoint (Maas et al., 2010). Most of these trials targeted mor necrosis factor-a (TNFa), and interferon-c (IFNc)) and oxida-
single secondary injury mechanisms, but given the multifactorial tive metabolites (e.g. nitric oxide, reactive oxygen and nitrogen
nature of the secondary injury process targeting a single factor will species) (Block and Hong, 2005). Further, the release of pro-inflam-
unlikely result in significant improvements in outcome. The com- matory cytokines and other soluble factors by activated microglia
plexity and diversity of secondary injury mechanisms have led to can significantly influence the subsequent activation of astrocytes
calls to target multiple delayed secondary injury mechanisms, and glial scar formation under pathological conditions including
either by combining agents that have complementary effects or central nervous system (CNS) injury (Zhang et al., 2010).
by using multipotential drugs that modulate multiple injury mech- Astrocyte activation (astrogliosis) is characterized by the in-
anisms (Loane and Faden, 2010; Margulies and Hicks, 2009; Vink crease of intermediate filaments (vimentin and GFAP), increased
and Nimmo, 2009). This recognition has led to the recent emphasis cell proliferation and an accompanying cellular hypertrophy
on multipotential drug treatments, several of which are now in (Herrmann et al., 2008). Similar to microglia, reactive astrocytes
clinical trials for human head injury (Vink and Nimmo, 2009). His- can have detrimental and/or beneficial roles following CNS injury.
torically neuroprotection treatments for TBI have been dominated Upon activation, astrocytes upregulate a number of neurotrophic
by a neuronocentric view, in which modification of neuronal based factors [e.g. brain-derived neurotrophic factor (BDNF)] that sup-
injury mechanisms is the primary or even exclusive focus of the port and protect against injury-induced cell death (Zhao et al.,
neuroprotective strategy. However, it is well established that neur- 2004). In addition, astrocytes play a crucial role in regulating extra-
oinflammation represents a key pathological response to brain in- cellular glutamate levels, which can reduce glutamate excitotoxic-
jury, and the important role that non-neuronal cells, such as ity to neurons and other cells (Schousboe and Waagepetersen,
endothelial cells, astrocytes, microglia, oligodendrocytes, play in 2005). Notably, impaired astrocyte performance exacerbates neu-
secondary injury-mediated responses is becoming increasingly ronal dysfunction following brain injury and transgenic ablation
recognized (Floyd and Lyeth, 2007; Loane and Byrnes, 2010; of reactive astrocytes increases neuronal cell death and promotes
Simard et al., 2010; Ziebell and Morganti-Kossmann, 2010). worse outcome after TBI (Myer et al., 2006); this may in part reflect
In this review we will discuss neuroinflammation as a key sec- the loss of ability to limit the influx of inflammatory cells. Follow-
ondary injury mechanism in TBI before focusing on the complex ing injury, hypertrophic astrocytes surround the lesion site and de-
and varied responses of microglia in terms of their detrimental posit an inhibitory extracellular matrix including chondroitin
and beneficial effects after injury. In addition, we will describe sulfate proteoglycans that contributes to the glial scar. This dense
emerging experimental anti-inflammatory and multipotential drug physical and chemical barrier inhibits axonal regeneration and pre-
treatment strategies that show considerable promise for the treat- vents functional connections required for axonal growth and repair
ment of human TBI, and we will discuss some of the challenges fac- (Cafferty et al., 2007). On the one hand, astrocytes provide neuro-
ing basic and clinical researchers in translating novel drug trophic support and guidance for axonal growth following CNS in-
treatment strategies from the bench to the bedside. jury, while on the other, prolonged astrogliosis inhibits axon
regeneration and hinders functional recovery. In fact, improved ax-
onal growth and repair following experimental brain and spinal
2. The neuroinflammatory response to traumatic brain injury cord injury has been demonstrated in transgenic mice deficient
in both vimentin and GFAP (Menet et al., 2003; Wilhelmsson
Neuroinflammation is an important secondary injury mecha- et al., 2004). Therefore, limiting prolonged astrogliosis may be
nism that contributes to on-going neurodegeneration and neuro- important for axon regeneration after traumatic CNS injury.
logical impairments associated with TBI. Post-traumatic
neuroinflammation is characterized by glial cell activation, leuko-
cyte recruitment, and upregulation of inflammatory mediators 3. Microglia – mediators of the innate immune response to CNS
(Morganti-Kossmann et al., 2007). Although much research has injury
focused on the detrimental effects of neuroinflammation on the
injured brain, clear beneficial effects can be achieved if neuroin- Microglia are the primary innate immune cells in the CNS. Un-
flammation is controlled in a regulated manner and for defined der normal physiological conditions these highly dynamic and mo-
periods of time. tile cells are spread throughout the brain parenchyma and
A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201 1193

constantly survey their microenvironment for noxious agents and complicated by the influence of infiltrating blood-borne macro-
injurious processes (Nimmerjahn et al., 2005). They respond to phages of M1 or M2 phenotype and other infiltrating cells follow-
extracellular signals and are responsible for clearing cellular debris ing CNS injury (Kigerl et al., 2009). However, understanding the
and toxic substances by phagocytosis, thereby maintaining normal molecular mechanisms that polarize and differentiate resident
cellular homeostasis in the CNS (Hanisch and Kettenmann, 2007). microglia or infiltrating macrophages towards an M2 phenotype,
Therefore, under non-pathological conditions there is continuing thereby promoting CNS repair while limiting inflammatory-medi-
low-level microglial activity in the CNS which is primarily involved ated secondary injury cascades, may provide an opportunity for
in activity-dependent synaptic pruning and in repair processes therapeutic intervention after CNS injury.
(Tremblay et al., 2011). However, in response to insult, infection
or injury, microglia become dysregulated and highly activated
which results in dramatic changes in cell morphology and behav- 4. Involvement of pro- and anti-inflammatory cytokines and
ior. Similar to peripheral innate immune cells microglia express chemokines in traumatic brain injury
pathogen recognition receptors (PRRs) such as toll-like receptors
(TLRs) and NOD-like receptors (NLRs), and therefore respond to The neuroinflammatory cascade activated in response to TBI is
pathogen-associated molecular patterns (PAMPs) and endoge- mediated by the release of pro- and anti-inflammatory cytokines
nously produced danger-associated molecular patterns (DAMPs), and chemokines, and microglia are the primary source of these
which are secreted by damaged neurons and others cells of the inflammatory mediators in the brain. Gene profiling studies in
CNS (Hanisch and Kettenmann, 2007). They also express receptors experimental models of TBI have shown that genes related to neur-
for a number of other factors that are released by damaged neu- oinflammation are strongly up-regulated in the acute phase after
rons, including ATP, glutamate, growth factors and cytokines. injury (Kobori et al., 2002; Natale et al., 2003; Raghavendra Rao
Microglia respond to an array of molecules by secreting chemo- et al., 2003). Additional studies focused on microglial related genes
kines, pro- and anti-inflammatory cytokines, neurotrophins and and their temporospatial localization after TBI demonstrated that
oxidative metabolites. Furthermore, microglia are antigen present- markers of activation (e.g. CD68, MHCII), stress responses (e.g.
ing cells, and upon activation they up-regulate the expression of p22phox, heme oxygenase 1) and chemokine expression (e.g.
cell surface markers such as MHC class II and CD86 among others, CXCL10, CXCL6) were markedly increased after TBI (Israelsson
as well as adhesion molecules and complement receptors (Lynch, et al., 2008).
2009). A key to maintaining microglia in a quiescent state is the Consistent with the early activation profile of microglia follow-
immunosuppressive potential of the brain microenvironment. In ing injury, there is rapid elevation of pro-inflammatory IL-1b with-
the healthy brain neurons express a number of immunosuppres- in hours of TBI in both humans and rodents (Fan et al., 1995;
sive proteins such as CD200, CD47 and fractalkine, which interact Winter et al., 2002; Woodroofe et al., 1991). The damaging effects
with their receptors on microglia to maintain the microglia in a of IL-1b are mediated through interleukin 1 receptor type 1 (IL-
quiescent/non-activated state (Harrison et al., 1998; Hoek et al., 1RI), which is expressed on microglia and neurons (Lu et al.,
2000; Lynch, 2009). In addition, soluble factors such as neurotro- 2005b; Pinteaux et al., 2002). This damage is not as a result of
phins, anti-inflammatory cytokines and prostaglandins released lo- the cytokine itself, but rather its affect on activating other pro-
cally in the brain by neurons, astrocytes and microglia inflammatory pathways such as TNFa (Rothwell, 2003). Inhibiting
downregulate the immune response thereby maintaining microg- IL-1b in experimental models of TBI has been shown to be neuro-
lia in a quiescent form (Lynch, 2009). protective and improve functional recovery (Basu et al., 2002; Lu
Microglia, like macrophages, have multiple activation pheno- et al., 2005a,b; Tehranian et al., 2002; Toulmond and Rothwell,
types (Gordon, 2003; Mantovani et al., 2004). Dependent on the 1995). The potent neurotoxic effects of IL-1b have been shown to
stimuli in their local microenvironment microglia can be polarized be synergistically enhanced in the presence of TNFa, suggesting
to have distinct molecular phenotypes and effector functions that these important cytokines mediate post-traumatic neuroin-
(Colton, 2009). For example, lipopolysaccharide (LPS) and the flammation and brain damage (Chao et al., 1995). Although IL-1b
pro-inflammatory cytokine IFNc promote a ‘classically activated’ and TNFa interact with distinct receptors that are structurally
M1 phenotype, which produces high levels of pro-inflammatory un-related, both cytokines share significant down-stream signaling
cytokines and oxidative metabolites that are essential for host de- pathways that may promote synergy of action. TNFa levels rise
fense and phagocytic activity, but that also cause damage to within 1 h of TBI, peak between 3 and 8 h, and return to normal
healthy cells and tissue. Conversely, activating microglia in the by 24 h (Fan et al., 1996; Shohami et al., 1994; Stover et al.,
presence of cytokines such as IL-4 or IL-13 promote an ‘alterna- 2000). Consistently, TNFa levels are elevated early after injury in
tively activated’ M2 phenotype (Colton et al., 2006; Ponomarev the serum and cerebrospinal fluid (CSF) of severely injured TBI pa-
et al., 2007). Although there is limited data on resident M2 microg- tients (Goodman et al., 1990; Ross et al., 1994). However, the role
lia in the brain, it is thought that much like the M2 macrophages, of TNFa in the pathogenesis of TBI is somewhat controversial and
these cells can promote angiogenesis, wound healing and tissue re- complex in nature with different functional outcomes in the acute
pair, extracellular matrix remodeling and suppress destructive im- and delayed phases after TBI (Scherbel et al., 1999; Sullivan et al.,
mune responses (Colton, 2009). M2 microglia express specific 1999). Initial pre-clinical neuroprotection studies targeting TNFa
antigens such as arginase 1 (Arg1), mannose receptor (MRC), found showed considerable promise, with three different compounds
in inflammatory zone 1 (FIZZ1) and chitinase 3-like 3 (Ym1) (Col- (dexanabinol {HU-211}, TNF-binding protein, pentoxifylline
ton et al., 2006), among others, and cultured microglia exposed to {PTX}) showing significant improvements in neurological out-
IL-4 or IL-13 develop the M2 phenotype, which can result in exten- comes and reducing post-traumatic BBB disruption and brain ede-
sive neurite elongation and outgrowth across inhibitory surfaces in ma (Shohami et al., 1997). However, deletion of TNFa in knockout
in vitro co-culture systems (Butovsky et al., 2005; Kigerl et al., mice attenuated deficits in the acute phase following TBI, but lim-
2009). Following an insult or injury to the brain it is likely that ited neurological improvements in the long term (Scherbel et al.,
M1 and M2 microglial exist in a state of dynamic equilibrium with- 1999). Another study demonstrated that knockout mice for the
in the lesion microenvironment. Whether these cells differentiate TNFa receptor had exacerbated tissue and BBB damage and im-
into an M1 phenotype that exacerbates tissue injury or into an paired neurological recovery after TBI (Sullivan et al., 1999), sug-
M2 phenotype that promotes CNS repair likely depends on the gesting that endogenous TNFa may be neuroprotective. These
local signals in the lesion microenvironment. This is further genetic studies indicate that the function of TNFa differs in the
1194 A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201

acute and delayed phase after TBI; immediately after injury TNFa both functional deficits and histological outcomes through
seems to act as a potent inflammatory mediator, but later it is a 4 months post-injury (Byrnes et al., 2012).
neurotrophic factor that is required for neuroprotection and repair. Aging also influences microglial activation (Conde and Streit,
Interestingly, anti-inflammatory cytokine levels are also chan- 2006b; Lucin and Wyss-Coray, 2009), and exacerbated microglial
ged after TBI. In humans, IL-10 and transforming growth factor- activation and astroglial responses to injury likely contribute to
b1 (TGFb1) levels are elevated acutely after injury (Csuka et al., the enhanced susceptibility to and poor recovery from TBI in el-
1999; Morganti-Kossmann et al., 1999), and experimental studies derly patients (Galbraith, 1987; Pennings et al., 1993). In fact,
have shown that IL-10 has beneficial effects following trauma experimental studies demonstrated that microglial activation
(Knoblach and Faden, 1998). For example, intravenous administra- was exaggerated and prolonged in aged mice when compared to
tion of IL-10 after experimental TBI in rats improved neurological adult mice (Sandhir et al., 2008), and these observations are consis-
recovery and significantly reduced TNFa and IL-1b expression in tent with reports of elevated microglial activation in the aged brain
the traumatized cortex and hippocampus. These neuroprotective following injuries such as facial nerve axotomy (Conde and Streit,
effects may be as a result of suppressed microglial activation, as 2006a) and cerebral ischemia (Popa-Wagner et al., 2007). It has
IL-10 treatment has been shown to decrease production of pro- been proposed that microglia in the aged brain are ‘primed’ to re-
inflammatory cytokines (Kremlev and Palmer, 2005). Furthermore, spond more rapidly, produce more pronounced inflammatory re-
treatment with the anti-inflammatory cytokine TGFb1 reduced le- sponses and proliferate more vigorously than microglia in the
sion size and improved neurological function after injury in rodent young brain following TBI (Conde and Streit, 2006b; Godbout
models (Tyor et al., 2002). Notably, the levels of IL-1 receptor et al., 2005). Therefore, a hyperactivated and dysfunctional microg-
antagonist (IL-1ra) are also elevated following TBI, and IL-1ra has lial response in the aged hippocampus may contribute to enhanced
significant anti-inflammatory properties. IL-1ra plays a major role neuronal loss and worse cognitive outcomes in the elderly follow-
in counteracting the biological effects of IL-1b, owing to its ability ing brain trauma.
to bind to IL-1RI without initiating signal transduction (Allan et al., There is growing awareness of the epidemiological association
2005). In experimental models of TBI, IL-1ra or IL-1b neutralization between a history of TBI and the development of Alzheimer’s dis-
resulted in attenuated pro-inflammatory cytokine and chemokine ease (AD) later in life (Graves et al., 1990; Mortimer et al., 1985;
production, reduced hippocampal damage and improved neurolog- van Duijn et al., 1992). This link is supported by the identification
ical behavior after injury (Basu et al., 2002; Lu et al., 2005a,b; of acute and chronic AD-like pathologies in the brains of TBI pa-
Tehranian et al., 2002; Toulmond and Rothwell, 1995). tients and in animal models of TBI. Amyloid-b (Ab) plaques, a hall-
mark of AD, may be found in TBI patients within hours of injury
(Ikonomovic et al., 2004; Roberts et al., 1991, 1994), and there
5. Chronic microglial activation and neurodegeneration after are several pathophysiological mechanisms linking TBI and AD,
traumatic brain injury such as the accumulation and clearance of Ab peptides after TBI
(Johnson et al., 2010). However, chronic neuroinflammation is a
Chronic microglial activation is considered to be the most dam- common neuropathological feature of TBI and AD, and chronic
aging response of microglia to injury (Block et al., 2007). DAMPs re- microglial activation may be a key causative factor. Ab is impli-
leased by injured neurons after TBI interact with TLRs and other cated in the pathology of AD, both through direct toxicity to neu-
PRRs on activated microglia and trigger a vicious self-perpetuating rons (Yankner et al., 1989), and by potentiating neuronal damage
cycle of damaging events that lead to prolonged and dysregulated by microglial activation (Combs et al., 2000). In AD patients acti-
microglial activation that drives pathogenic processes and neuro- vated microglia cluster at sites of aggregated Ab and penetrate
degeneration (Block et al., 2007; Loane and Byrnes, 2010). Human the neuritic plaques (Cagnin et al., 2001; McGeer et al., 1987). Fur-
and animal studies indicate that microglia are chronically activated thermore, Ab is a potent pro-inflammatory stimulator of microglia
for weeks, months and even years after the initial brain trauma, (Ii et al., 1996; Qin et al., 2002). Interestingly, proinflammatory
and may contribute to chronic neurodegeneration and related neu- cytokines expressed by microglia, such as IL-1b, TNFa and IFNc,
rological deficits following injury (Bendlin et al., 2008; Bramlett can specifically stimulate c-secretase activity resulting in in-
and Dietrich, 2002; Maxwell et al., 2006; Smith et al., 1997). Persis- creased production of Ab and the intracellular domain of APP
tent long-term microglial activation has been demonstrated in ani- (AICD) (Liao et al., 2004). Moreover, TBI induces the expression of
mal models of TBI and is associated with increased expression of the c-secretase complex proteins in microglia and astrocytes
pro-inflammatory cytokines (e.g. IL-1b, TNFa) (Holmin and (Nadler et al., 2008), thereby suggesting a role for glial cells in post-
Mathiesen, 1999). Notably, a recent clinical study utilizing the pos- traumatic accumulation of Ab. It is evident that the interactions
itron emission tomography ligand [11C] (R)PK11195 to assess between Ab accumulation/clearance and chronic microglial activa-
chronic microglial activation in 10 patients studied more than tion after TBI are complex and poorly understood. Further detailed
11 months after moderate to severe TBI reported significantly in- mechanistic studies on these molecular interactions after brain in-
creased binding bilaterally at sites distant from areas of focal in- jury are needed because targeting these pathways may provide no-
jury, such as thalamus, and correlated these changes with several vel therapeutic opportunities for the treatment of TBI.
measures of cognitive dysfunction (Ramlackhansingh et al.,
2011). Furthermore, post-mortem studies have also demonstrated
increased microglial activation in the white matter of head-injury 6. Challenges translating promising experimental
survivors up to 16 years after TBI (Gentleman et al., 2004). The neuroprotection strategies to the clinic
experimental and clinical evidence now suggest that TBI should
not be viewed as a static, acute neurodegenerative disorder. In- Neuroprotective treatments that limit secondary injury mecha-
stead, TBI initiates chronic biochemical processes leading to pro- nisms and/or improve behavioral outcome have been well estab-
longed neuroinflammation and microglial activation, and as such lished in multiple animal models of TBI. However, translation of
there may be a longer therapeutic window for the treatment of promising experimental neuroprotective treatments to human in-
head injury than traditionally accepted. In fact, a recent experi- jury have been very disappointing, with none of the pharmacolog-
mental study which used a pharmacological treatment (mGluR5 ical treatments resulting in any consistent improvements in
agonist) to inhibit chronic microglial activation demonstrated that outcome in the clinic (Maas et al., 2010). Both conceptual issues
delayed treatment at 1 month post-injury significantly reduced and methodological differences between preclinical research and
A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201 1195

clinical injury have undoubtedly contributed to these translational drugs. Recommendations regarding preclinical experimental de-
difficulties. Apart from the potential shortcomings of the drugs sign for the evaluation of neuroprotective agents for TBI have been
themselves, a number of flaws in preclinical research and clinical made (Loane and Faden, 2010), and they build on the STAIR criteria
translation have been recognized. These include (1) inadequate for development of neuroprotective treatments for stroke/ischemia
understanding of secondary injury mechanisms; (2) insufficient (Fisher et al., 2009; STAIR, 1999), and are in line with NIH guide-
preclinical testing in multiple TBI models (e.g. lateral fluid percus- lines for effective therapies for TBI (Margulies and Hicks, 2009;
sion, controlled cortical impact, closed head injury models), strains, Saatman et al., 2008). However, the heterogeneity of clinical TBI
species (including gyrencephalic), genders and ages; (3) lack of and the complex nature of the secondary injury process represent
thorough investigation of pharmacokinetics and therapeutic brain the most significant hurdles to future clinical trial successes. It is
concentrations of drugs under investigation; (4) failure to ade- unlikely that targeting any single secondary injury factor will re-
quately examine the therapeutic window for the drug and failure sult in significant improvement in outcome after TBI. Therefore,
to use clinically relevant behavioral outcomes; (5) lack of animal simultaneous targeting of several injury factors using multipoten-
models that include secondary insults such as CNS hypoxia and tial drugs may maximize the likelihood of developing a successful
systemic injuries despite these insults being commonplace in clin- therapeutic intervention to improve outcome in TBI patients.
ical TBI (6) use of heterogeneous patient populations in clinical tri- A number of anti-inflammatory and multipotential drug treat-
als for TBI; (7) inadequate sample size (lack of power); and (8) ment strategies inhibit post-traumatic neuroinflammation and
inadequate functional outcome measurements and biomarkers microglial activation, and have shown considerable promise in pre-
(molecular and neuroimaging) for clinical TBI. Each of these factors clinical studies (Fig. 1). In the following section we will review
has contributed to the failure to translate a successful pharmaco- these neuroprotective drug treatment strategies, focusing on drugs
logical intervention to date. Nonetheless, many of these shortcom- that are currently in randomized controlled clinical trials for head
ings can be addressed with improved preclinical screening of injury, or emerging pharmacological treatments that show promise

Fig. 1. Anti-inflammatory and multipotential drug treatment strategies for TBI. Neuroinflammation and microglial activation are key secondary injury mechanisms that
contribute to chronic neurodegeneration and loss of neurological function following TBI. Preclinical studies have identified a number of anti-inflammatory and multipotential
drug treatment strategies that inhibit post-traumatic neuroinflammation and microglial activation in addition to reducing other secondary injury mechanisms such as edema
formation and neuronal apoptosis.
1196 A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201

in experimental TBI studies and warrant further investigation. stress and cerebral edema, and improved neurological function
Unfortunately due to space limitations only a small number of when administered after TBI (Besson et al., 2005; Chen et al.,
promising anti-inflammatory drug treatments can be detailed in 2007b). Similarly, the PPARc receptor agonists, pioglitazone and
this article, and readers are directed to excellent review articles rosiglitazone, reduced post-traumatic microglial activation and
on other preclinical drug treatment strategies for TBI (Shohami promoted anti-oxidant (e.g. Mn-SOD) and neuroprotective chaper-
et al., 2011; Simard et al., 2010; Ziebell and Morganti-Kossmann, one protein (e.g. HSP27/70) expression that resulted in improved
2010). functional and histological outcomes after TBI (Sauerbeck et al.,
2011; Thal et al., 2011; Yi et al., 2008). The anti-inflammatory
and neuroprotective effects of pioglitazone are likely to be
7. Anti-inflammatory treatments for traumatic brain injury
PPARc-independent as the beneficial effects of treatment were
not reversed by the PPARc antagonist T0070907 (Thal et al.,
7.1. Minocycline
2011). Notably, PPARc receptor agonist are used clinically for the
treatment of type 2 diabetes, and PPARc modulates macrophage
Minocycline is a second generation tetracycline that is known to
M2 alternative activation states thereby improving resistance to
have neuroprotective properties that are independent of its anti-
insulin (Odegaard et al., 2007). Furthermore, rosiglitazone also in-
microbial activity (Yrjanheikki et al., 1998). Minocycline is a potent
duces IL-4 expression in the brain and reduces microglial activa-
anti-inflammatory drug that suppresses the production of several
tion in an IL-4 dependent manner (Loane et al., 2009). Therefore,
pro-inflammatory cytokines (Choi et al., 2005; Seabrook et al.,
PPARc agonists could be used to promote an M2 microglial activa-
2006), and inhibits microglial-mediated neurotoxicity (Tikka
tion phenotype through IL-4 dependent mechanisms, which could
et al., 2001). It has been shown to have significant neuroprotective
potentially modulate the local microenvironment surrounding a
effects in spinal cord injury (SCI) models by reducing both inflam-
lesion, thereby promoting tissue repair and reducing the damaging
matory and apoptotic pathways (Festoff et al., 2006; Teng et al.,
effects of neuroinflammation after TBI.
2004; Wells et al., 2003). Minocycline has been used in experimen-
tal TBI models and it was demonstrated to improve neurological
recovery after injury due to its ability to inhibit post-traumatic
8. Multipotential drug treatments for traumatic brain injury
microglial activation and neuronal apoptosis (Sanchez Mejia
et al., 2001). However, another report demonstrated that minocy-
8.1. Cell cycle inhibitors
cline treatment resulted in only a transient improvement in neuro-
logical recovery after TBI, and that there was no difference in
The cell cycle is upregulated in both mitotic (astrocytes and
outcomes by day 4 post-injury when compared to vehicle-treated
microglia) and post-mitotic (neurons, oligodendrocytes) cells of
controls (Bye et al., 2007). Apoptotic mechanisms were not af-
the brain after CNS injury, and post-traumatic cell cycle activation
fected by minocycline treatment in this study, but IL-1b and IL-6
is associated with caspase-mediated neuronal cell death and glial
levels were decreased in the acute phase after injury and microglial
cell proliferation (Di Giovanni et al., 2005). Cell cycle inhibitors
activation was significantly reduced, which may account for the
have been extensively studied for their role in cancer treatment,
transient neuroprotective effects of minocycline. In contrast, recent
and inhibitors such as flavopiridol, roscovitine and olomoucine,
studies suggest that blockade of acute microglial activation by
have been shown to exert powerful neuroprotective effects in
minocycline is neuroprotective and promotes functional recovery
in vitro model systems for neuronal apoptosis (Cernak et al.,
up to 3 months post-injury (Homsi et al., 2010; Ng et al., 2012;
2005; Padmanabhan et al., 1999; Verdaguer et al., 2004), and they
Siopi et al., 2011, 2012). However, the short treatment protocol
also produce potent inhibitory effects on the proliferation and acti-
used in the latter studies requires that further investigation into
vation of astrocytes and microglia (Cernak et al., 2005; Di Giovanni
the therapeutic window for minocycline treatment needs to be
et al., 2005; Hilton et al., 2008). Given the multipotential properties
performed before it can be considered for clinical translation for
of cell cycle inhibitors they are considered to be promising candi-
human head injury.
date drugs for the treatment of TBI.
In experimental studies treatment with flavopiridol, an inhibi-
7.2. Peroxisome proliferator-activated receptor (PPAR) agonists tor of all major cyclic-dependent kinases (CDKs), after lateral fluid
percussion TBI in rats significantly reduced lesion volume, and im-
Another class of drugs that display strong anti-inflammatory proved long-term cognitive and sensorimotor recovery (Di Giovan-
properties are the synthetic agonists for PPARs. PPARs are li- ni et al., 2005). In addition, flavopiridol treatment blocked caspase-
gand-activated transcription factors of the nuclear hormone recep- mediated neuronal cell death and significantly reduced glial cell
tor family (Berger and Moller, 2002). Upon activation by specific activation, and these changes were associated with the suppres-
agonists, PPARs bind as heterodimers with a retinoid X receptor sion of the cell cycle in neurons, astrocytes, and microglia within
and translocate to the nucleus, where they act as agonist-depen- the injured cortex. Follow up studies demonstrated that delayed
dent transcription factors that regulate gene expression by binding treatment with flavopiridol as late as 24 h post-injury resulted in
to specific promoter regions of target genes. The PPARs play a crit- significantly reduced TBI lesion volumes (Cernak et al., 2005). Ros-
ical physiological role as lipid sensors and regulators of lipid covitine is a more selective cell cycle inhibitor, which acts specifi-
metabolism, and they are also involved in reproductive, develop- cally on CDKs 1, 2, and 5 (Meijer et al., 1997), and it too had
mental and immune responses. Activation of the PPAR a and c iso- significant neuroprotective actions in rat and mouse TBI models.
forms are known to have potent anti-inflammatory effects, In addition to improving functional recovery and reducing the
including attenuation of pro-inflammatory mediators, such as TBI lesion, central and systemic administration of roscovitine
iNOS and COX2 (Bernardo and Minghetti, 2006; Chawla et al., markedly reduced microglial-mediated neuroinflammation and
2001). PPAR agonists are neuroprotective in acute brain injury associated neurodegeneration up to 28 days post-injury (Hilton
models such as transient focal ischemia (Bernardo and Minghetti, et al., 2008; Kabadi et al., 2012a). Further, genetic deletion of cyclin
2006; Sundararajan et al., 2005), and similar anti-inflammatory d1, a key modulator of the cell cycle, resulted in significantly re-
and neuroprotective properties have been demonstrated in exper- duced microglial activation and improved histological and func-
imental TBI studies. For example, the PPARa receptor agonist, tional outcomes after TBI (Kabadi et al., 2012b), thereby
fenofibrate, reduced post-traumatic neuroinflammation, oxidative underpinning the key role that cell cycle activation plays in the
A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201 1197

neuroinflammatory response to TBI. Several cell cycle inhibitors nitive performance in the Morris water maze than males after
have been studied in randomized clinical trials for cancer (Fischer experimental TBI (Roof et al., 1993), and that progesterone-treated
and Gianella-Borradori, 2005). Although they are toxic when male rats were less impaired in the task than vehicle-treated ani-
administered chronically, cell cycle inhibitors have required only mals (Roof et al., 1994). Later studies demonstrated that progester-
single dose administration to exert powerful therapeutic effects one inhibited multiple secondary injury pathways in TBI. For
in experimental TBI models. Therefore, given their clinically rele- example, progesterone attenuates glutamate excitotoxicity (Smith,
vant therapeutic window and their multipotential neuroprotective 1991), modulates apoptotic pathways (Djebaili et al., 2005; Yao
effects, cell cycle inhibitors such as flavopiridol or roscovitine are et al., 2005), and decreases diffuse axonal injury (O’Connor et al.,
attractive candidates for future clinical translation. 2007). It also reduces lipid peroxidation (Roof et al., 1997), possibly
by upregulating superoxide dismutase (Moorthy et al., 2005), and
8.2. Statins reduces cerebral edema formation (O’Connor et al., 2007; Roof
et al., 1996). Notably, progesterone treatment reduces neuroin-
The 3-hydroxy-3-methyglutaryl coenzyme A (HMGCoA) reduc- flammation after TBI, by attenuating NFjB signaling, IL-1b, IL-6
tase inhibitors (also known as statins) are inhibitors of cholesterol and TNFa production, as well as the inflammatory C3 complement
biosynthesis, and they have additional pleiotropic properties that protein (Grossman et al., 2004; Pettus et al., 2005).
make them attractive multipotential neuroprotective drugs (Wible Although progesterone has been shown to confer neuroprotec-
and Laskowitz, 2010). Statins increase endothelium-derived nitric tion in models of experimental stroke, SCI and TBI, a recent system-
oxide production (Eto et al., 2002) and reduce vascular inflamma- atic review questions its effectiveness in trauma models (Gibson
tion (Maeda et al., 2003), thereby improving the microvasculature et al., 2008). Despite the perceived limitations of the preclinical re-
after traumatic insult. In in vitro models statins protect cortical search, two randomized, double-blind, placebo-controlled phase II
neurons from NMDA-induced excitotoxic death (Zacco et al., clinical trials for progesterone have been conducted (Wright et al.,
2003), and statin treatment significantly improves neuronal sur- 2007; Xiao et al., 2008). Although these trials used different doses
vival following TBI (Lu et al., 2004, 2007; Qu et al., 2005; Wang and treatment regimens, both indicated trends toward improved
et al., 2007). Statins may also promote the growth and differentia- outcome in progesterone treated patients. The results are promis-
tion of new neurons after brain injury (Lu et al., 2007; Wu et al., ing since both clinical studies not only showed reduced mortality
2008b), and their ability to increase neurogenesis may be in part but also reduced morbidity as indicated by improved functional
due to upregulation of neurotrophic factors (e.g. BDNF) (Chen outcomes. In 2010, two major Phase III clinical trials for progester-
et al., 2005; Wu et al., 2008b). Notably, statins exert powerful one (ProTECT III and SyNAPSe trials) were set up to provide a more
anti-inflammatory effects, in part by decreasing the formation of robust answer on the effectiveness of progesterone treatment in
isoprenoids (Bi et al., 2004; Cordle and Landreth, 2005). In TBI TBI (Stein and Wright, 2010), and the results of these studies are
models, statins have been shown to significantly reduce pro- eagerly anticipated.
inflammatory cytokine production and attenuate microglial activa-
tion and cerebral edema formation, while increasing BBB integrity
(Chen et al., 2007a, 2009; Wang et al., 2007). Moreover, inhibition 9. Conclusions
of the TLR4 and NFjB signaling pathways are potential mecha-
nisms through which statins modulate the post-traumatic neuro- Neuroinflammation and microglial activation are key secondary
inflammatory response (Chen et al., 2009). injury mechanisms that contribute to chronic neurodegeneration
Experimental studies have shown that statins target multiple and loss of neurological function after TBI. However, the neuroin-
secondary injury pathways and significantly improve functional flammatory response to TBI possesses both beneficial and detri-
recovery after TBI (Chen et al., 2007a, 2009; Lu et al., 2004, 2007; mental effects, and these likely differ in the acute and delayed
Qu et al., 2005; Wang et al., 2007; Wu et al., 2008a). Furthermore, phases after injury. The key to developing future neuroprotective
the therapeutic window for this class of drugs is relatively large, treatments that target post-traumatic neuroinflammation and
with treatment 24 h after TBI resulting in long-term functional microglial activation is to minimize the detrimental and neuro-
improvements and reduced neuronal cell loss (Lu et al., 2004, toxic effects of neuroinflammation while promoting the beneficial
2007). As such, statins possess key preclinical neuroprotective and neurotrophic effects, thereby creating optimal conditions for
characteristics that make them suitable candidates for clinical regeneration and repair after injury. The complex and multifaceted
translation. Importantly, statins have a long clinical track record responses of microglia to injury are essential to these processes,
in critically ill patients; they are easy to administer, are well toler- and research must focus on identifying the signals and mecha-
ated and have well-defined side effects (Tseng et al., 2005). A small nisms by which microglia can be guided to promote optimal repair.
prospective, randomized, double-blind clinical trial in TBI has been Further detailed understanding of the role of classical/M1 and
performed using the statin drug rosuvastatin, and treatment alternatively/M2 activated microglial phenotypes, particularly in
showed modest improvements in TBI associated amnesia and dis- relation to the chronic neuroinflammatory responses to injury is
orientation time outcomes (Tapia-Perez et al., 2008). Other phase II required, so that therapeutic approaches that promote the alterna-
clinical trials to evaluate rosuvastatin and atorvastatin for the tive/M2 phenotype can be developed to support regeneration and
treatment of head injury are planned. repair after injury.
So complex and intertwined are the secondary injury responses
8.3. Progesterone to TBI that the concept of a single magic bullet strategy for neuro-
protection is no longer accepted. Focus has now turned to multipo-
Another neuroprotective drug undergoing clinical trials for head tential therapeutic strategies that modulate independent
injury is progesterone; a neurosteroid whose receptors are ex- secondary injury mechanisms simultaneously, and several treat-
pressed in the CNS of both males and females (Camacho-Arroyo ments have been identified experimentally and are currently in
et al., 1994). Progesterone is also thought to have pleiotropic neu- or are awaiting human clinical trials for head injury. Notably, many
roprotective properties, and has been reported to improve out- of these multipotential drug treatments target post-traumatic
comes in experimental models of SCI (Gonzalez Deniselle et al., microglial activation and neuroinflammatory mechanisms, thereby
2002), stroke (Jiang et al., 1996) and TBI (Roof and Hall, 2000). demonstrating the major role that these pathways play in the
Intriguingly, it was first discovered that female rats had better cog- pathogenesis of this disorder.
1198 A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201

Acknowledgment protects dopaminergic neurons in the substantia nigra in vivo. J. Neurochem.


95, 1755–1765.
Colton, C.A., 2009. Heterogeneity of microglial activation in the innate immune
This work is supported by a pilot award from the National response in the brain. J. Neuroimmune Pharmacol. 4, 399–418.
Capital Area Rehabilitation Research Network (R24HD050845) Colton, C.A., Mott, R.T., Sharpe, H., Xu, Q., Van Nostrand, W.E., Vitek, M.P., 2006.
Expression profiles for macrophage alternative activation genes in AD and in
(D.J.L.).
mouse models of AD. J. Neuroinflamm. 3, 27.
Combs, C.K., Johnson, D.E., Karlo, J.C., Cannady, S.B., Landreth, G.E., 2000.
Inflammatory mechanisms in Alzheimer’s disease: inhibition of beta-amyloid-
stimulated proinflammatory responses and neurotoxicity by PPARgamma
References agonists. J. Neurosci. 20, 558–567.
Conde, J.R., Streit, W.J., 2006a. Effect of aging on the microglial response to
peripheral nerve injury. Neurobiol. Aging 27, 1451–1461.
Allan, S.M., Tyrrell, P.J., Rothwell, N.J., 2005. Interleukin-1 and neuronal injury. Nat.
Conde, J.R., Streit, W.J., 2006b. Microglia in the aging brain. J. Neuropathol. Exp.
Rev. Immunol. 5, 629–640.
Neurol. 65, 199–203.
Basu, A., Krady, J.K., O’Malley, M., Styren, S.D., DeKosky, S.T., Levison, S.W., 2002. The
Cordle, A., Landreth, G., 2005. 3-Hydroxy-3-methylglutaryl-coenzyme A reductase
type 1 interleukin-1 receptor is essential for the efficient activation of microglia
inhibitors attenuate beta-amyloid-induced microglial inflammatory responses.
and the induction of multiple proinflammatory mediators in response to brain
J. Neurosci. 25, 299–307.
injury. J. Neurosci. 22, 6071–6082.
Csuka, E., Morganti-Kossmann, M.C., Lenzlinger, P.M., Joller, H., Trentz, O.,
Bendlin, B.B., Ries, M.L., Lazar, M., Alexander, A.L., Dempsey, R.J., Rowley, H.A.,
Kossmann, T., 1999. IL-10 levels in cerebrospinal fluid and serum of patients
Sherman, J.E., Johnson, S.C., 2008. Longitudinal changes in patients with
with severe traumatic brain injury: relationship to IL-6, TNF-alpha, TGF-beta1
traumatic brain injury assessed with diffusion-tensor and volumetric imaging.
and blood–brain barrier function. J. Neuroimmunol. 101, 211–221.
Neuroimage 42, 503–514.
Davalos, D., Grutzendler, J., Yang, G., Kim, J.V., Zuo, Y., Jung, S., Littman, D.R., Dustin,
Berger, J., Moller, D.E., 2002. The mechanisms of action of PPARs. Annu. Rev. Med.
M.L., Gan, W.B., 2005. ATP mediates rapid microglial response to local brain
53, 409–435.
injury in vivo. Nat. Neurosci. 8, 752–758.
Bernardo, A., Minghetti, L., 2006. PPAR-gamma agonists as regulators of microglial
Di Giovanni, S., Movsesyan, V., Ahmed, F., Cernak, I., Schinelli, S., Stoica, B., Faden,
activation and brain inflammation. Curr. Pharm. Des. 12, 93–109.
A.I., 2005. Cell cycle inhibition provides neuroprotection and reduces glial
Besson, V.C., Chen, X.R., Plotkine, M., Marchand-Verrecchia, C., 2005. Fenofibrate, a
proliferation and scar formation after traumatic brain injury. Proc. Natl. Acad.
peroxisome proliferator-activated receptor alpha agonist, exerts
Sci. USA 102, 8333–8338.
neuroprotective effects in traumatic brain injury. Neurosci. Lett. 388, 7–12.
Djebaili, M., Guo, Q., Pettus, E.H., Hoffman, S.W., Stein, D.G., 2005. The neurosteroids
Bi, X., Baudry, M., Liu, J., Yao, Y., Fu, L., Brucher, F., Lynch, G., 2004. Inhibition of
progesterone and allopregnanolone reduce cell death, gliosis, and functional
geranylgeranylation mediates the effects of 3-hydroxy-3-methylglutaryl
deficits after traumatic brain injury in rats. J. Neurotrauma 22, 106–118.
(HMG)-CoA reductase inhibitors on microglia. J. Biol. Chem. 279, 48238–48245.
Eto, M., Kozai, T., Cosentino, F., Joch, H., Luscher, T.F., 2002. Statin prevents tissue
Block, M.L., Hong, J.S., 2005. Microglia and inflammation-mediated
factor expression in human endothelial cells: role of Rho/Rho-kinase and Akt
neurodegeneration: multiple triggers with a common mechanism. Prog.
pathways. Circulation 105, 1756–1759.
Neurobiol. 76, 77–98.
Faden, A.I., Demediuk, P., Panter, S.S., Vink, R., 1989. The role of excitatory amino
Block, M.L., Zecca, L., Hong, J.S., 2007. Microglia-mediated neurotoxicity: uncovering
acids and NMDA receptors in traumatic brain injury. Science 244, 798–800.
the molecular mechanisms. Nat. Rev. 8, 57–69.
Fan, L., Young, P.R., Barone, F.C., Feuerstein, G.Z., Smith, D.H., McIntosh, T.K., 1995.
Bramlett, H.M., Dietrich, W.D., 2002. Quantitative structural changes in white and
Experimental brain injury induces expression of interleukin-1 beta mRNA in the
gray matter 1 year following traumatic brain injury in rats. Acta Neuropathol.
rat brain. Brain Res. Mol. Brain Res. 30, 125–130.
103, 607–614.
Fan, L., Young, P.R., Barone, F.C., Feuerstein, G.Z., Smith, D.H., McIntosh, T.K., 1996.
Butovsky, O., Talpalar, A.E., Ben-Yaakov, K., Schwartz, M., 2005. Activation of
Experimental brain injury induces differential expression of tumor necrosis
microglia by aggregated beta-amyloid or lipopolysaccharide impairs MHC-II
factor-alpha mRNA in the CNS. Brain Res. Mol. Brain Res. 36, 287–291.
expression and renders them cytotoxic whereas IFN-gamma and IL-4 render
Faul, M., Xu, L., Wald, M.M., Coronado, V.G., 2010. Traumatic brain injury in the
them protective. Mol. Cell Neurosci. 29, 381–393.
United States: Emergency Department Visits, Hospitalizations and Deaths
Bye, N., Habgood, M.D., Callaway, J.K., Malakooti, N., Potter, A., Kossmann, T.,
2002–2006. Centers for Disease Control and Prevention, National Center for
Morganti-Kossmann, M.C., 2007. Transient neuroprotection by minocycline
Injury Prevention and Control, Atlanta (GA).
following traumatic brain injury is associated with attenuated microglial
Festoff, B.W., Ameenuddin, S., Arnold, P.M., Wong, A., Santacruz, K.S., Citron, B.A.,
activation but no changes in cell apoptosis or neutrophil infiltration. Exp.
2006. Minocycline neuroprotects, reduces microgliosis, and inhibits caspase
Neurol. 204, 220–233.
protease expression early after spinal cord injury. J. Neurochem. 97, 1314–1326.
Byrnes, K.R., Loane, D.J., Stoica, B.A., Zhang, J., Faden, A.I., 2012. Delayed mGluR5
Fischer, P.M., Gianella-Borradori, A., 2005. Recent progress in the discovery and
activation limits neuroinflammation and neurodegeneration after traumatic
development of cyclin-dependent kinase inhibitors. Exp. Opin. Investig. Drugs
brain injury. J. Neuroinflamm. 9, 43.
14, 457–477.
Cafferty, W.B., Yang, S.H., Duffy, P.J., Li, S., Strittmatter, S.M., 2007. Functional axonal
Fisher, M., Feuerstein, G., Howells, D.W., Hurn, P.D., Kent, T.A., Savitz, S.I., Lo, E.H.,
regeneration through astrocytic scar genetically modified to digest chondroitin
2009. Update of the stroke therapy academic industry roundtable preclinical
sulfate proteoglycans. J. Neurosci. 27, 2176–2185.
recommendations. Stroke 40, 2244–2250.
Cagnin, A., Brooks, D.J., Kennedy, A.M., Gunn, R.N., Myers, R., Turkheimer, F.E., Jones,
Floyd, C.L., Lyeth, B.G., 2007. Astroglia: important mediators of traumatic brain
T., Banati, R.B., 2001. In-vivo measurement of activated microglia in dementia.
injury. Prog. Brain Res. 161, 61–79.
Lancet 358, 461–467.
Galbraith, S., 1987. Head injuries in the elderly. Br. Med. J. (Clin. Res. Ed.) 294, 325.
Camacho-Arroyo, I., Perez-Palacios, G., Pasapera, A.M., Cerbon, M.A., 1994.
Gentile, N.T., McIntosh, T.K., 1993. Antagonists of excitatory amino acids and
Intracellular progesterone receptors are differentially regulated by sex steroid
endogenous opioid peptides in the treatment of experimental central nervous
hormones in the hypothalamus and the cerebral cortex of the rabbit. J. Steroid
system injury. Ann. Emerg. Med. 22, 1028–1034.
Biochem. Mol. Biol. 50, 299–303.
Gentleman, S.M., Leclercq, P.D., Moyes, L., Graham, D.I., Smith, C., Griffin, W.S.,
Cernak, I., Stoica, B., Byrnes, K.R., Di Giovanni, S., Faden, A.I., 2005. Role of the cell
Nicoll, J.A., 2004. Long-term intracerebral inflammatory response after
cycle in the pathobiology of central nervous system trauma. Cell cycle
traumatic brain injury. Forensic Sci. Int. 146, 97–104.
(Georgetown Tex) 4, 1286–1293.
Gibson, C.L., Gray, L.J., Bath, P.M., Murphy, S.P., 2008. Progesterone for the treatment
Chao, C.C., Hu, S., Ehrlich, L., Peterson, P.K., 1995. Interleukin-1 and tumor necrosis
of experimental brain injury; a systematic review. Brain 131, 318–328.
factor-alpha synergistically mediate neurotoxicity: involvement of nitric oxide
Godbout, J.P., Chen, J., Abraham, J., Richwine, A.F., Berg, B.M., Kelley, K.W., Johnson,
and of N-methyl-D-aspartate receptors. Brain Behav. Immun. 9, 355–365.
R.W., 2005. Exaggerated neuroinflammation and sickness behavior in aged mice
Chawla, A., Barak, Y., Nagy, L., Liao, D., Tontonoz, P., Evans, R.M., 2001. PPAR-gamma
following activation of the peripheral innate immune system. FASEB J. 19,
dependent and independent effects on macrophage-gene expression in lipid
1329–1331.
metabolism and inflammation. Nat. Med. 7, 48–52.
Gonzalez Deniselle, M.C., Lopez Costa, J.J., Gonzalez, S.L., Labombarda, F., Garay, L.,
Chen, J., Zhang, C., Jiang, H., Li, Y., Zhang, L., Robin, A., Katakowski, M., Lu, M., Chopp,
Guennoun, R., Schumacher, M., De Nicola, A.F., 2002. Basis of progesterone
M., 2005. Atorvastatin induction of VEGF and BDNF promotes brain plasticity
protection in spinal cord neurodegeneration. J. Steroid Biochem. Mol. Biol. 83,
after stroke in mice. J. Cereb. Blood Flow Metab. 25, 281–290.
199–209.
Chen, S.F., Hung, T.H., Chen, C.C., Lin, K.H., Huang, Y.N., Tsai, H.C., Wang, J.Y., 2007a.
Goodman, J.C., Robertson, C.S., Grossman, R.G., Narayan, R.K., 1990. Elevation of
Lovastatin improves histological and functional outcomes and reduces
tumor necrosis factor in head injury. J. Neuroimmunol. 30, 213–217.
inflammation after experimental traumatic brain injury. Life Sci. 81, 288–298.
Gordon, S., 2003. Alternative activation of macrophages. Nat. Rev. Immunol. 3, 23–35.
Chen, X.R., Besson, V.C., Palmier, B., Garcia, Y., Plotkine, M., Marchand-Leroux, C.,
Graves, A.B., White, E., Koepsell, T.D., Reifler, B.V., van Belle, G., Larson, E.B., Raskind,
2007b. Neurological recovery-promoting, anti-inflammatory, and anti-
M., 1990. The association between head trauma and Alzheimer’s disease. Am. J.
oxidative effects afforded by fenofibrate, a PPAR alpha agonist, in traumatic
Epidemiol. 131, 491–501.
brain injury. J. Neurotrauma 24, 1119–1131.
Grossman, K.J., Goss, C.W., Stein, D.G., 2004. Effects of progesterone on the
Chen, G., Zhang, S., Shi, J., Ai, J., Qi, M., Hang, C., 2009. Simvastatin reduces secondary
inflammatory response to brain injury in the rat. Brain Res. 1008, 29–39.
brain injury caused by cortical contusion in rats: possible involvement of TLR4/
Hall, E.D., Detloff, M.R., Johnson, K., Kupina, N.C., 2004. Peroxynitrite-mediated
NF-kappaB pathway. Exp. Neurol. 216, 398–406.
protein nitration and lipid peroxidation in a mouse model of traumatic brain
Choi, S.H., Lee, D.Y., Chung, E.S., Hong, Y.B., Kim, S.U., Jin, B.K., 2005. Inhibition of
injury. J. Neurotrauma 21, 9–20.
thrombin-induced microglial activation and NADPH oxidase by minocycline
A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201 1199

Hanisch, U.K., Kettenmann, H., 2007. Microglia: active sensor and versatile effector Lucin, K.M., Wyss-Coray, T., 2009. Immune activation in brain aging and
cells in the normal and pathologic brain. Nat. Neurosci. 10, 1387–1394. neurodegeneration: too much or too little? Neuron 64, 110–122.
Harrison, J.K., Jiang, Y., Chen, S., Xia, Y., Maciejewski, D., McNamara, R.K., Streit, W.J., Lynch, M.A., 2009. The multifaceted profile of activated microglia. Mol. Neurobiol.
Salafranca, M.N., Adhikari, S., Thompson, D.A., Botti, P., Bacon, K.B., Feng, L., 40, 139–156.
1998. Role for neuronally derived fractalkine in mediating interactions between Maas, A.I., Stocchetti, N., Bullock, R., 2008. Moderate and severe traumatic brain
neurons and CX3CR1-expressing microglia. Proc. Natl. Acad. Sci. USA 95, injury in adults. Lancet Neurol. 7, 728–741.
10896–10901. Maas, A.I., Roozenbeek, B., Manley, G.T., 2010. Clinical trials in traumatic brain
Haynes, S.E., Hollopeter, G., Yang, G., Kurpius, D., Dailey, M.E., Gan, W.B., Julius, D., injury: past experience and current developments. Neurotherapeutics 7, 115–
2006. The P2Y12 receptor regulates microglial activation by extracellular 126.
nucleotides. Nat. Neurosci. 9, 1512–1519. Maeda, T., Kawane, T., Horiuchi, N., 2003. Statins augment vascular endothelial
Herrmann, J.E., Imura, T., Song, B., Qi, J., Ao, Y., Nguyen, T.K., Korsak, R.A., Takeda, K., growth factor expression in osteoblastic cells via inhibition of protein
Akira, S., Sofroniew, M.V., 2008. STAT3 is a critical regulator of astrogliosis and prenylation. Endocrinology 144, 681–692.
scar formation after spinal cord injury. J. Neurosci. 28, 7231–7243. Mantovani, A., Sica, A., Sozzani, S., Allavena, P., Vecchi, A., Locati, M., 2004. The
Hilton, G.D., Stoica, B.A., Byrnes, K.R., Faden, A.I., 2008. Roscovitine reduces neuronal chemokine system in diverse forms of macrophage activation and polarization.
loss, glial activation, and neurologic deficits after brain trauma. J. Cereb. Blood Trends Immunol. 25, 677–686.
Flow Metab. 28, 1845–1859. Margulies, S., Hicks, R., 2009. Combination therapies for traumatic brain injury:
Hoek, R.M., Ruuls, S.R., Murphy, C.A., Wright, G.J., Goddard, R., Zurawski, S.M., Blom, prospective considerations. J. Neurotrauma 26, 925–939.
B., Homola, M.E., Streit, W.J., Brown, M.H., Barclay, A.N., Sedgwick, J.D., 2000. Maxwell, W.L., MacKinnon, M.A., Smith, D.H., McIntosh, T.K., Graham, D.I., 2006.
Down-regulation of the macrophage lineage through interaction with OX2 Thalamic nuclei after human blunt head injury. J. Neuropathol. Exp. Neurol. 65,
(CD200). Science 290, 1768–1771. 478–488.
Holmin, S., Mathiesen, T., 1999. Long-term intracerebral inflammatory response McGeer, P.L., Itagaki, S., Tago, H., McGeer, E.G., 1987. Reactive microglia in patients
after experimental focal brain injury in rat. NeuroReport 10, 1889–1891. with senile dementia of the Alzheimer type are positive for the
Homsi, S., Piaggio, T., Croci, N., Noble, F., Plotkine, M., Marchand-Leroux, C., Jafarian- histocompatibility glycoprotein HLA-DR. Neurosci. Lett. 79, 195–200.
Tehrani, M., 2010. Blockade of acute microglial activation by minocycline McIntosh, T.K., Smith, D.H., Meaney, D.F., Kotapka, M.J., Gennarelli, T.A., Graham,
promotes neuroprotection and reduces locomotor hyperactivity after closed D.I., 1996. Neuropathological sequelae of traumatic brain injury: relationship to
head injury in mice: a twelve-week follow-up study. J. Neurotrauma 27, 911– neurochemical and biomechanical mechanisms. Lab. Invest. 74, 315–342.
921. Meijer, L., Borgne, A., Mulner, O., Chong, J.P., Blow, J.J., Inagaki, N., Inagaki, M.,
Ii, M., Sunamoto, M., Ohnishi, K., Ichimori, Y., 1996. Beta-Amyloid protein- Delcros, J.G., Moulinoux, J.P., 1997. Biochemical and cellular effects of
dependent nitric oxide production from microglial cells and neurotoxicity. roscovitine, a potent and selective inhibitor of the cyclin-dependent kinases
Brain Res. 720, 93–100. cdc2, cdk2 and cdk5. Eur. J. Biochem. 243, 527–536.
Ikonomovic, M.D., Uryu, K., Abrahamson, E.E., Ciallella, J.R., Trojanowski, J.Q., Lee, Menet, V., Prieto, M., Privat, A., Gimenez y Ribotta, M., 2003. Axonal plasticity and
V.M., Clark, R.S., Marion, D.W., Wisniewski, S.R., DeKosky, S.T., 2004. Alzheimer’s functional recovery after spinal cord injury in mice deficient in both glial
pathology in human temporal cortex surgically excised after severe brain fibrillary acidic protein and vimentin genes. Proc. Natl. Acad. Sci. USA 100,
injury. Exp. Neurol. 190, 192–203. 8999–9004.
Israelsson, C., Bengtsson, H., Kylberg, A., Kullander, K., Lewen, A., Hillered, L., Moorthy, K., Sharma, D., Basir, S.F., Baquer, N.Z., 2005. Administration of estradiol
Ebendal, T., 2008. Distinct cellular patterns of upregulated chemokine and progesterone modulate the activities of antioxidant enzyme and
expression supporting a prominent inflammatory role in traumatic brain aminotransferases in naturally menopausal rats. Exp. Gerontol. 40, 295–302.
injury. J. Neurotrauma 25, 959–974. Morganti-Kossmann, M.C., Hans, V.H., Lenzlinger, P.M., Dubs, R., Ludwig, E., Trentz,
Jiang, N., Chopp, M., Stein, D., Feit, H., 1996. Progesterone is neuroprotective after O., Kossmann, T., 1999. TGF-beta is elevated in the CSF of patients with severe
transient middle cerebral artery occlusion in male rats. Brain Res. 735, 101–107. traumatic brain injuries and parallels blood-brain barrier function. J.
Johnson, V.E., Stewart, W., Smith, D.H., 2010. Traumatic brain injury and amyloid- Neurotrauma 16, 617–628.
beta pathology: a link to Alzheimer’s disease? Nat. Rev. Neurosci.. Morganti-Kossmann, M.C., Rancan, M., Otto, V.I., Stahel, P.F., Kossmann, T., 2001.
Kabadi, S.V., Stoica, B.A., Byrnes, K.R., Hanscom, M., Loane, D.J., Faden, A.I., 2012a. Role of cerebral inflammation after traumatic brain injury: a revisited concept.
Selective CDK inhibitor limits neuroinflammation and progressive Shock 16, 165–177.
neurodegeneration after brain trauma. J. Cereb. Blood Flow Metab. 32, 137–149. Morganti-Kossmann, M.C., Satgunaseelan, L., Bye, N., Kossmann, T., 2007.
Kabadi, S.V., Stoica, B.A., Loane, D.J., Byrnes, K.R., Hanscom, M., Cabatbat, R.M., Tan, Modulation of immune response by head injury. Injury 38, 1392–1400.
M.T., Faden, A.I., 2012b. Cyclin d1 gene ablation confers neuroprotection in Mortimer, J.A., French, L.R., Hutton, J.T., Schuman, L.M., 1985. Head injury as a risk
traumatic brain injury. J. Neurotrauma 29, 813–827. factor for Alzheimer’s disease. Neurology 35, 264–267.
Kigerl, K.A., Gensel, J.C., Ankeny, D.P., Alexander, J.K., Donnelly, D.J., Popovich, P.G., Myer, D.J., Gurkoff, G.G., Lee, S.M., Hovda, D.A., Sofroniew, M.V., 2006. Essential
2009. Identification of two distinct macrophage subsets with divergent effects protective roles of reactive astrocytes in traumatic brain injury. Brain 129,
causing either neurotoxicity or regeneration in the injured mouse spinal cord. J. 2761–2772.
Neurosci. 29, 13435–13444. Nadler, Y., Alexandrovich, A., Grigoriadis, N., Hartmann, T., Rao, K.S., Shohami, E.,
Knoblach, S.M., Faden, A.I., 1998. Interleukin-10 improves outcome and alters Stein, R., 2008. Increased expression of the gamma-secretase components
proinflammatory cytokine expression after experimental traumatic brain presenilin-1 and nicastrin in activated astrocytes and microglia following
injury. Exp. Neurol. 153, 143–151. traumatic brain injury. Glia 56, 552–567.
Kobori, N., Clifton, G.L., Dash, P., 2002. Altered expression of novel genes in the Natale, J.E., Ahmed, F., Cernak, I., Stoica, B., Faden, A.I., 2003. Gene expression profile
cerebral cortex following experimental brain injury. Brain Res. Mol. Brain Res. changes are commonly modulated across models and species after traumatic
104, 148–158. brain injury. J. Neurotrauma 20, 907–927.
Kremlev, S.G., Palmer, C., 2005. Interleukin-10 inhibits endotoxin-induced pro- Ng, S.Y., Semple, B.D., Morganti-Kossmann, M.C., Bye, N., 2012. Attenuation of
inflammatory cytokines in microglial cell cultures. J. Neuroimmunol. 162, 71–80. microglial activation with minocycline is not associated with changes in
Liao, Y.F., Wang, B.J., Cheng, H.T., Kuo, L.H., Wolfe, M.S., 2004. Tumor necrosis factor- neurogenesis after focal traumatic brain injury in adult mice. J. Neurotrauma
alpha, interleukin-1beta, and interferon-gamma stimulate gamma-secretase- 29, 1410–1425.
mediated cleavage of amyloid precursor protein through a JNK-dependent Nimmerjahn, A., Kirchhoff, F., Helmchen, F., 2005. Resting microglial cells are
MAPK pathway. J. Biol. Chem. 279, 49523–49532. highly dynamic surveillants of brain parenchyma in vivo. Science 308, 1314–
Loane, D.J., Byrnes, K.R., 2010. Role of microglia in neurotrauma. Neurotherapeutics 1318.
7, 366–377. O’Connor, C.A., Cernak, I., Johnson, F., Vink, R., 2007. Effects of progesterone on
Loane, D.J., Faden, A.I., 2010. Neuroprotection for traumatic brain injury: neurologic and morphologic outcome following diffuse traumatic brain injury
translational challenges and emerging therapeutic strategies. Trends in rats. Exp. Neurol. 205, 145–153.
Pharmacol. Sci. 31, 596–604. Odegaard, J.I., Ricardo-Gonzalez, R.R., Goforth, M.H., Morel, C.R., Subramanian, V.,
Loane, D.J., Deighan, B.F., Clarke, R.M., Griffin, R.J., Lynch, A.M., Lynch, M.A., 2009. Mukundan, L., Red Eagle, A., Vats, D., Brombacher, F., Ferrante, A.W., Chawla, A.,
Interleukin-4 mediates the neuroprotective effects of rosiglitazone in the aged 2007. Macrophage-specific PPARgamma controls alternative activation and
brain. Neurobiol. Aging 30, 920–931. improves insulin resistance. Nature 447, 1116–1120.
Lu, D., Goussev, A., Chen, J., Pannu, P., Li, Y., Mahmood, A., Chopp, M., 2004. Padmanabhan, J., Park, D.S., Greene, L.A., Shelanski, M.L., 1999. Role of cell cycle
Atorvastatin reduces neurological deficit and increases synaptogenesis, regulatory proteins in cerebellar granule neuron apoptosis. J. Neurosci. 19,
angiogenesis, and neuronal survival in rats subjected to traumatic brain 8747–8756.
injury. J. Neurotrauma 21, 21–32. Pennings, J.L., Bachulis, B.L., Simons, C.T., Slazinski, T., 1993. Survival after severe
Lu, K.T., Wang, Y.W., Wo, Y.Y., Yang, Y.L., 2005a. Extracellular signal-regulated brain injury in the aged. Arch. Surg. 128, 787–793, discussion 793–784.
kinase-mediated IL-1-induced cortical neuron damage during traumatic brain Pettus, E.H., Wright, D.W., Stein, D.G., Hoffman, S.W., 2005. Progesterone treatment
injury. Neurosci. Lett. 386, 40–45. inhibits the inflammatory agents that accompany traumatic brain injury. Brain
Lu, K.T., Wang, Y.W., Yang, J.T., Yang, Y.L., Chen, H.I., 2005b. Effect of interleukin-1 Res. 1049, 112–119.
on traumatic brain injury-induced damage to hippocampal neurons. J. Pinteaux, E., Parker, L.C., Rothwell, N.J., Luheshi, G.N., 2002. Expression of
Neurotrauma 22, 885–895. interleukin-1 receptors and their role in interleukin-1 actions in murine
Lu, D., Qu, C., Goussev, A., Jiang, H., Lu, C., Schallert, T., Mahmood, A., Chen, J., Li, Y., microglial cells. J. Neurochem. 83, 754–763.
Chopp, M., 2007. Statins increase neurogenesis in the dentate gyrus, reduce Ponomarev, E.D., Maresz, K., Tan, Y., Dittel, B.N., 2007. CNS-derived interleukin-4 is
delayed neuronal death in the hippocampal CA3 region, and improve spatial essential for the regulation of autoimmune inflammation and induces a state of
learning in rat after traumatic brain injury. J. Neurotrauma 24, 1132–1146. alternative activation in microglial cells. J. Neurosci. 27, 10714–10721.
1200 A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201

Popa-Wagner, A., Carmichael, S.T., Kokaia, Z., Kessler, C., Walker, L.C., 2007. The Smith, D.H., Chen, X.H., Pierce, J.E., Wolf, J.A., Trojanowski, J.Q., Graham, D.I.,
response of the aged brain to stroke: too much, too soon? Curr. Neurovasc. Res. McIntosh, T.K., 1997. Progressive atrophy and neuron death for one year
4, 216–227. following brain trauma in the rat. J. Neurotrauma 14, 715–727.
Qin, L., Liu, Y., Cooper, C., Liu, B., Wilson, B., Hong, J.S., 2002. Microglia enhance beta- STAIR, 1999. Recommendations for standards regarding preclinical neuroprotective
amyloid peptide-induced toxicity in cortical and mesencephalic neurons by and restorative drug development. Stroke 30, 2752–2758.
producing reactive oxygen species. J. Neurochem. 83, 973–983. Stein, D.G., Wright, D.W., 2010. Progesterone in the clinical treatment of acute
Qu, C., Lu, D., Goussev, A., Schallert, T., Mahmood, A., Chopp, M., 2005. Effect of traumatic brain injury. Exp. Opin. Investig. Drugs 19, 847–857.
atorvastatin on spatial memory, neuronal survival, and vascular density in Stover, J.F., Schoning, B., Beyer, T.F., Woiciechowsky, C., Unterberg, A.W., 2000.
female rats after traumatic brain injury. J. Neurosurg. 103, 695–701. Temporal profile of cerebrospinal fluid glutamate, interleukin-6, and tumor
Raghavendra Rao, V.L., Dhodda, V.K., Song, G., Bowen, K.K., Dempsey, R.J., 2003. necrosis factor-alpha in relation to brain edema and contusion following
Traumatic brain injury-induced acute gene expression changes in rat cerebral controlled cortical impact injury in rats. Neurosci. Lett. 288, 25–28.
cortex identified by GeneChip analysis. J. Neurosci. Res. 71, 208–219. Sullivan, P.G., Bruce-Keller, A.J., Rabchevsky, A.G., Christakos, S., Clair, D.K., Mattson,
Ramlackhansingh, A.F., Brooks, D.J., Greenwood, R.J., Bose, S.K., Turkheimer, F.E., M.P., Scheff, S.W., 1999. Exacerbation of damage and altered NF-kappaB
Kinnunen, K.M., Gentleman, S., Heckemann, R.A., Gunanayagam, K., Gelosa, G., activation in mice lacking tumor necrosis factor receptors after traumatic
Sharp, D.J., 2011. Inflammation after trauma: microglial activation and brain injury. J. Neurosci. 19, 6248–6256.
traumatic brain injury. Ann. Neurol. 70, 374–383. Sundararajan, S., Gamboa, J.L., Victor, N.A., Wanderi, E.W., Lust, W.D., Landreth, G.E.,
Roberts, G.W., Gentleman, S.M., Lynch, A., Graham, D.I., 1991. Beta A4 amyloid 2005. Peroxisome proliferator-activated receptor-gamma ligands reduce
protein deposition in brain after head trauma. Lancet 338, 1422–1423. inflammation and infarction size in transient focal ischemia. Neuroscience
Roberts, G.W., Gentleman, S.M., Lynch, A., Murray, L., Landon, M., Graham, D.I., 130, 685–696.
1994. Beta amyloid protein deposition in the brain after severe head injury: Tapia-Perez, J.H., Sanchez-Aguilar, M., Torres-Corzo, J.G., Gordillo-Moscoso, A.,
implications for the pathogenesis of Alzheimer’s disease. J. Neurol. Neurosurg. Martinez-Perez, P., Madeville, P., de la Cruz-Mendoza, E., Chalita-Williams, J.,
Psychiatry 57, 419–425. 2008. Effect of rosuvastatin on amnesia and disorientation after traumatic brain
Roof, R.L., Hall, E.D., 2000. Gender differences in acute CNS trauma and stroke: injury (NCT003229758). J. Neurotrauma 25, 1011–1017.
neuroprotective effects of estrogen and progesterone. J. Neurotrauma 17, 367– Tehranian, R., Andell-Jonsson, S., Beni, S.M., Yatsiv, I., Shohami, E., Bartfai, T.,
388. Lundkvist, J., Iverfeldt, K., 2002. Improved recovery and delayed cytokine
Roof, R.L., Zhang, Q., Glasier, M.M., Stein, D.G., 1993. Gender-specific impairment on induction after closed head injury in mice with central overexpression of the
Morris water maze task after entorhinal cortex lesion. Behav. Brain Res. 57, 47– secreted isoform of the interleukin-1 receptor antagonist. J. Neurotrauma 19,
51. 939–951.
Roof, R.L., Duvdevani, R., Braswell, L., Stein, D.G., 1994. Progesterone facilitates Teng, Y.D., Choi, H., Onario, R.C., Zhu, S., Desilets, F.C., Lan, S., Woodard, E.J., Snyder,
cognitive recovery and reduces secondary neuronal loss caused by cortical E.Y., Eichler, M.E., Friedlander, R.M., 2004. Minocycline inhibits contusion-
contusion injury in male rats. Exp. Neurol. 129, 64–69. triggered mitochondrial cytochrome c release and mitigates functional deficits
Roof, R.L., Duvdevani, R., Heyburn, J.W., Stein, D.G., 1996. Progesterone rapidly after spinal cord injury. Proc. Natl. Acad. Sci. USA 101, 3071–3076.
decreases brain edema: treatment delayed up to 24 hours is still effective. Exp. Thal, S.C., Heinemann, M., Luh, C., Pieter, D., Werner, C., Engelhard, K., 2011.
Neurol. 138, 246–251. Pioglitazone reduces secondary brain damage after experimental brain trauma
Roof, R.L., Hoffman, S.W., Stein, D.G., 1997. Progesterone protects against lipid by PPAR-gamma-independent mechanisms. J. Neurotrauma 28, 983–993.
peroxidation following traumatic brain injury in rats. Mol. Chem. Neuropathol. Tikka, T., Fiebich, B.L., Goldsteins, G., Keinanen, R., Koistinaho, J., 2001. Minocycline,
31, 1–11. a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting
Ross, S.A., Halliday, M.I., Campbell, G.C., Byrnes, D.P., Rowlands, B.J., 1994. The activation and proliferation of microglia. J. Neurosci. 21, 2580–2588.
presence of tumour necrosis factor in CSF and plasma after severe head injury. Toulmond, S., Rothwell, N.J., 1995. Interleukin-1 receptor antagonist inhibits
Br. J. Neurosurg. 8, 419–425. neuronal damage caused by fluid percussion injury in the rat. Brain Res. 671,
Rothwell, N., 2003. Interleukin-1 and neuronal injury: mechanisms, modification, 261–266.
and therapeutic potential. Brain Behav. Immun. 17, 152–157. Tremblay, M.E., Stevens, B., Sierra, A., Wake, H., Bessis, A., Nimmerjahn, A., 2011. The
Saatman, K.E., Duhaime, A.C., Bullock, R., Maas, A.I., Valadka, A., Manley, G.T., 2008. role of microglia in the healthy brain. J. Neurosci. 31, 16064–16069.
Classification of traumatic brain injury for targeted therapies. J. Neurotrauma Tseng, M.Y., Czosnyka, M., Richards, H., Pickard, J.D., Kirkpatrick, P.J., 2005. Effects of
25, 719–738. acute treatment with pravastatin on cerebral vasospasm, autoregulation, and
Sanchez Mejia, R.O., Ona, V.O., Li, M., Friedlander, R.M., 2001. Minocycline reduces delayed ischemic deficits after aneurysmal subarachnoid hemorrhage: a phase
traumatic brain injury-mediated caspase-1 activation, tissue damage, and II randomized placebo-controlled trial. Stroke 36, 1627–1632.
neurological dysfunction. Neurosurgery 48, 1393–1399, discussion 1399–1401. Tyor, W.R., Avgeropoulos, N., Ohlandt, G., Hogan, E.L., 2002. Treatment of spinal cord
Sandhir, R., Onyszchuk, G., Berman, N.E., 2008. Exacerbated glial response in the impact injury in the rat with transforming growth factor-beta. J. Neurol. Sci.
aged mouse hippocampus following controlled cortical impact injury. Exp. 200, 33–41.
Neurol. 213, 372–380. van Duijn, C.M., Tanja, T.A., Haaxma, R., Schulte, W., Saan, R.J., Lameris, A.J.,
Sauerbeck, A., Gao, J., Readnower, R., Liu, M., Pauly, J.R., Bing, G., Sullivan, P.G., 2011. Antonides-Hendriks, G., Hofman, A., 1992. Head trauma and the risk of
Pioglitazone attenuates mitochondrial dysfunction, cognitive impairment, Alzheimer’s disease. Am. J. Epidemiol. 135, 775–782.
cortical tissue loss, and inflammation following traumatic brain injury. Exp. Verdaguer, E., Jimenez, A., Canudas, A.M., Jorda, E.G., Sureda, F.X., Pallas, M., Camins,
Neurol. 227, 128–135. A., 2004. Inhibition of cell cycle pathway by flavopiridol promotes survival of
Scherbel, U., Raghupathi, R., Nakamura, M., Saatman, K.E., Trojanowski, J.Q., cerebellar granule cells after an excitotoxic treatment. J. Pharmacol. Exp. Ther.
Neugebauer, E., Marino, M.W., McIntosh, T.K., 1999. Differential acute and 308, 609–616.
chronic responses of tumor necrosis factor-deficient mice to experimental brain Vink, R., Nimmo, A.J., 2009. Multifunctional drugs for head injury.
injury. Proc. Natl. Acad. Sci. USA 96, 8721–8726. Neurotherapeutics 6, 28–42.
Schousboe, A., Waagepetersen, H.S., 2005. Role of astrocytes in glutamate Wang, H., Lynch, J.R., Song, P., Yang, H.J., Yates, R.B., Mace, B., Warner, D.S., Guyton,
homeostasis: implications for excitotoxicity. Neurotox. Res. 8, 221–225. J.R., Laskowitz, D.T., 2007. Simvastatin and atorvastatin improve behavioral
Seabrook, T.J., Jiang, L., Maier, M., Lemere, C.A., 2006. Minocycline affects microglia outcome, reduce hippocampal degeneration, and improve cerebral blood flow
activation, Abeta deposition, and behavior in APP-tg mice. Glia 53, 776–782. after experimental traumatic brain injury. Exp. Neurol. 206, 59–69.
Shohami, E., Novikov, M., Bass, R., Yamin, A., Gallily, R., 1994. Closed head injury Wells, J.E., Hurlbert, R.J., Fehlings, M.G., Yong, V.W., 2003. Neuroprotection by
triggers early production of TNF alpha and IL-6 by brain tissue. J. Cereb. Blood minocycline facilitates significant recovery from spinal cord injury in mice.
Flow Metab. 14, 615–619. Brain 126, 1628–1637.
Shohami, E., Gallily, R., Mechoulam, R., Bass, R., Ben-Hur, T., 1997. Cytokine Wible, E.F., Laskowitz, D.T., 2010. Statins in traumatic brain injury.
production in the brain following closed head injury: dexanabinol (HU-211) is a Neurotherapeutics 7, 62–73.
novel TNF-alpha inhibitor and an effective neuroprotectant. J. Neuroimmunol. Wilhelmsson, U., Li, L., Pekna, M., Berthold, C.H., Blom, S., Eliasson, C., Renner, O.,
72, 169–177. Bushong, E., Ellisman, M., Morgan, T.E., Pekny, M., 2004. Absence of glial
Shohami, E., Cohen-Yeshurun, A., Magid, L., Algali, M., Mechoulam, R., 2011. fibrillary acidic protein and vimentin prevents hypertrophy of astrocytic
Endocannabinoids and traumatic brain injury. Br. J. Pharmacol. 163, 1402–1410. processes and improves post-traumatic regeneration. J. Neurosci. 24, 5016–
Simard, J.M., Kahle, K.T., Gerzanich, V., 2010. Molecular mechanisms of 5021.
microvascular failure in central nervous system injury – synergistic roles of Winter, C.D., Iannotti, F., Pringle, A.K., Trikkas, C., Clough, G.F., Church, M.K., 2002. A
NKCC1 and SUR1/TRPM4. J. Neurosurg. 113, 622–629. microdialysis method for the recovery of IL-1beta, IL-6 and nerve growth factor
Siopi, E., Cho, A.H., Homsi, S., Croci, N., Plotkine, M., Marchand-Leroux, C., Jafarian- from human brain in vivo. J. Neurosci. Methods 119, 45–50.
Tehrani, M., 2011. Minocycline restores sAPPalpha levels and reduces the late Woodroofe, M.N., Sarna, G.S., Wadhwa, M., Hayes, G.M., Loughlin, A.J., Tinker, A.,
histopathological consequences of traumatic brain injury in mice. J. Cuzner, M.L., 1991. Detection of interleukin-1 and interleukin-6 in adult rat
Neurotrauma 28, 2135–2143. brain, following mechanical injury, by in vivo microdialysis: evidence of a role
Siopi, E., Llufriu-Daben, G., Fanucchi, F., Plotkine, M., Marchand-Leroux, C., Jafarian- for microglia in cytokine production. J. Neuroimmunol. 33, 227–236.
Tehrani, M., 2012. Evaluation of late cognitive impairment and anxiety states Wright, D.W., Kellermann, A.L., Hertzberg, V.S., Clark, P.L., Frankel, M., Goldstein,
following traumatic brain injury in mice. The effect of minocycline. Neurosci. F.C., Salomone, J.P., Dent, L.L., Harris, O.A., Ander, D.S., Lowery, D.W., Patel, M.M.,
Lett. 511, 110–115. Denson, D.D., Gordon, A.B., Wald, M.M., Gupta, S., Hoffman, S.W., Stein, D.G.,
Smith, S.S., 1991. Progesterone administration attenuates excitatory amino acid 2007. ProTECT: a randomized clinical trial of progesterone for acute traumatic
responses of cerebellar Purkinje cells. Neuroscience 42, 309–320. brain injury. Ann. Emerg. Med. 49, 391–402, 402 e391–392.
A. Kumar, D.J. Loane / Brain, Behavior, and Immunity 26 (2012) 1191–1201 1201

Wu, H., Lu, D., Jiang, H., Xiong, Y., Qu, C., Li, B., Mahmood, A., Zhou, D., Chopp, M., Yao, X.L., Liu, J., Lee, E., Ling, G.S., McCabe, J.T., 2005. Progesterone differentially
2008a. Increase in phosphorylation of Akt and its downstream signaling targets regulates pro- and anti-apoptotic gene expression in cerebral cortex following
and suppression of apoptosis by simvastatin after traumatic brain injury. J. traumatic brain injury in rats. J. Neurotrauma 22, 656–668.
Neurosurg. 109, 691–698. Yi, J.H., Park, S.W., Brooks, N., Lang, B.T., Vemuganti, R., 2008. PPARgamma agonist
Wu, H., Lu, D., Jiang, H., Xiong, Y., Qu, C., Li, B., Mahmood, A., Zhou, D., Chopp, M., rosiglitazone is neuroprotective after traumatic brain injury via anti-
2008b. Simvastatin-mediated upregulation of VEGF and BDNF, activation of the inflammatory and anti-oxidative mechanisms. Brain Res. 1244, 164–172.
PI3K/Akt pathway, and increase of neurogenesis are associated with therapeutic Yrjanheikki, J., Keinanen, R., Pellikka, M., Hokfelt, T., Koistinaho, J., 1998.
improvement after traumatic brain injury. J. Neurotrauma 25, 130–139. Tetracyclines inhibit microglial activation and are neuroprotective in global
Xiao, G., Wei, J., Yan, W., Wang, W., Lu, Z., 2008. Improved outcomes from the brain ischemia. Proc. Natl. Acad. Sci. USA 95, 15769–15774.
administration of progesterone for patients with acute severe traumatic brain Zacco, A., Togo, J., Spence, K., Ellis, A., Lloyd, D., Furlong, S., Piser, T., 2003. 3-
injury: a randomized controlled trial. Crit. Care 12, R61. Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors protect cortical
Xiong, Y., Gu, Q., Peterson, P.L., Muizelaar, J.P., Lee, C.P., 1997. Mitochondrial neurons from excitotoxicity. J. Neurosci. 23, 11104–11111.
dysfunction and calcium perturbation induced by traumatic brain injury. J. Zhang, D., Hu, X., Qian, L., O’Callaghan, J.P., Hong, J.S., 2010. Astrogliosis in CNS
Neurotrauma 14, 23–34. pathologies: is there a role for microglia? Mol. Neurobiol. 41, 232–241.
Yakovlev, A.G., Knoblach, S.M., Fan, L., Fox, G.B., Goodnight, R., Faden, A.I., 1997. Zhao, Z., Alam, S., Oppenheim, R.W., Prevette, D.M., Evenson, A., Parsadanian, A.,
Activation of CPP32-like caspases contributes to neuronal apoptosis and 2004. Overexpression of glial cell line-derived neurotrophic factor in the CNS
neurological dysfunction after traumatic brain injury. J. Neurosci. 17, 7415– rescues motoneurons from programmed cell death and promotes their long-
7424. term survival following axotomy. Exp. Neurol. 190, 356–372.
Yankner, B.A., Dawes, L.R., Fisher, S., Villa-Komaroff, L., Oster-Granite, M.L., Neve, Ziebell, J.M., Morganti-Kossmann, M.C., 2010. Involvement of pro- and anti-
R.L., 1989. Neurotoxicity of a fragment of the amyloid precursor associated with inflammatory cytokines and chemokines in the pathophysiology of traumatic
Alzheimer’s disease. Science 245, 417–420. brain injury. Neurotherapeutics 7, 22–30.

You might also like