Vous êtes sur la page 1sur 18

REVIEW ARTICLE

published: 04 March 2013


doi: 10.3389/fneur.2013.00018

The role of markers of inflammation in traumatic


brain injury
Thomas Woodcock 1 and Maria Cristina Morganti-Kossmann 2 *
1
Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
2
Department of Epidemiology and Preventive Medicine, Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, VIC, Australia

Edited by: Within minutes of a traumatic impact, a robust inflammatory response is elicited in the
Stefania Mondello, University of
injured brain. The complexity of this post-traumatic squeal involves a cellular compo-
Messina, USA
nent, comprising the activation of resident glial cells, microglia, and astrocytes, and the
Reviewed by:
V. Wee Yong, University of Calgary, infiltration of blood leukocytes. The second component regards the secretion immune
Canada mediators, which can be divided into the following sub-groups: the archetypal pro-
Amade Bregy, University of Miami inflammatory cytokines (Interleukin-1, Tumor Necrosis Factor, Interleukin-6), the anti-
Miller School of Medicine, USA
inflammatory cytokines (IL-4, Interleukin-10, and TGF-beta), and the chemotactic cytokines
*Correspondence:
or chemokines, which specifically drive the accumulation of parenchymal and peripheral
Maria Cristina Morganti-Kossmann,
Department of Epidemiology and immune cells in the injured brain region. Such mechanisms have been demonstrated in
Preventive Medicine, School of Public animal models, mostly in rodents, as well as in human brain. Whilst the humoral immune
Health and Preventive Medicine, response is particularly pronounced in the acute phase followingTraumatic brain injury (TBI),
Australian and New Zealand Intensive
the activation of glial cells seems to be a rather prolonged effect lasting for several months.
Care Research Centre, Monash
University, The Alfred Centre, 99 The complex interaction of cytokines and cell types installs a network of events, which
Commercial Road, Melbourne, VIC subsequently intersect with adjacent pathological cascades including oxidative stress, exci-
3004, Australia. totoxicity, or reparative events including angiogenesis, scarring, and neurogenesis. It is
e-mail: cristina.morganti-kossmann@
monash.edu
well accepted that neuroinflammation is responsible of beneficial and detrimental effects,
contributing to secondary brain damage but also facilitating neurorepair. Although such
mediators are clear markers of immune activation, to what extent cytokines can be defined
as diagnostic factors reflecting brain injury or as predictors of long term outcome needs to
be further substantiated. In clinical studies some groups reported a proportional cytokine
production in either the cerebrospinal fluid or intraparenchymal tissue with initial brain dam-
age, mortality, or poor outcome scores. However, the validity of cytokines as biomarkers
is not broadly accepted. This review article will discuss the evidence from both clinical and
laboratory studies exploring the validity of immune markers as a correlate to classification
and outcome following TBI.
Keywords: traumatic brain injury, biomarkers, inflammation, cytokines, chemokines

INTRODUCTION treatment has been found to confer neuroprotection by targeting


Traumatic brain injury (TBI) has long been called a silent epi- secondary injury mechanisms.
demic (Goldstein, 1990; Coburn, 1992). In the United States, it Currently, the main focus for TBI patient management is
accounted for at least 1.5 million emergency room visits and hos- monitoring and maintenance of normal intracranial pressure
pitalizations annually between 1995 and 2001, a number that has (ICP), and cerebral perfusion pressure (CPP). One of the effects
certainly increased as a result of the wars in Iraq and Afghanistan of elevated ICP is the reduction in CPP, which consequently
(Langlois et al., 2006). Many survivors of TBI are left with long leads to secondary ischemia. It is no surprise therefore that
term disabilities, and even a mild TBI can leave people with high ICP has been shown to be associated with mortality and
cognitive impairments, difficulty in concentrating, fatigue, and poor outcome in TBI patients (Narayan et al., 1982; Treggiari
headaches. It disproportionately affects young men, but is also et al., 2007). Although treatment of high ICP is certainly ben-
increasingly common in the elderly population (Hukkelhoven eficial, being able to predict elevations in ICP and apply pre-
et al., 2003). The financial burden to the United States has been ventive or early interventions would be more ideal. At present,
estimated to exceed $56 billion annually (Finkelstein et al., 2006; there is no method with which to predict changes in ICP,
Langlois et al., 2006; Rutland-Brown et al., 2006) while in 2008 since neurological examinations and routinely administered com-
alone it has been calculated to approximately AU$ 8.6 billion in puted tomography (CT) scans do not provide this information.
Australia. Despite advances in prevention measures, surgical, and To this end, the development of biomarkers that have predic-
diagnostic techniques, there have been relatively few changes in tive accuracy with regard to ICP would greatly improve patient
the way TBI patients are managed, and so far no pharmacological management.

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 1


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

In addition to lowering elevated ICP, treatment and manage- THE DEVELOPMENT OF BIOMARKERS IN TBI
ment of a TBI patient could depend on a wide range of fac- The most commonly used biomarkers include S100B, neuron-
tors. Human TBI is a very heterogeneous condition due to the specific enolase (NSE), and myelin basic protein (MBP; Palfrey-
intrinsic combination of focal and diffuse injuries and the indi- man et al., 1978; Thomas et al., 1978) reflecting the extent of
vidual response via secondary mechanisms of neurodegeneration. tissue damage as well as having prognostic value for long term out-
Understanding these processes would aid the development of an come (Baker et al., 2009; Svetlov et al., 2009; Kovesdi et al., 2010).
individual patients tailored treatment plan. At present patients There are several observational clinical TBI studies where S100B
are categorized based on admission characteristics including age, has been successfully correlated with initial brain injury severity
pupil reaction, Glasgow coma scale scores (GCS), body tempera- (GCS), size of brain damage (on CT/MRI scans), and neurological
ture, blood glucose, non-cranial injuries (Hukkelhoven et al., 2005; outcome (Glasgow Outcome Scale/Extended; GOSE). However,
Mushkudiani et al., 2008), and also observations from a CT scan it is only recently that biomarkers are being employed in the
(e.g., Marshall CT classification, primarily prognostically oriented context of clinical trials with prospective collection of physiolog-
Rotterdam score; Marshall et al., 1992; Maas et al., 2005). Mag- ical data, outcomes, and the clinical assessment of efficacy of an
netic resonance imaging (MRI) is routinely used in the clinic and intervention. The ultimate aim is to demonstrate the correlations
is becoming very popular for the quantification of brain damage between a drugs neuroprotection and the reduced concentration
particularly in diffuse TBI. The more recently developed diffu- of biomarkers in TBI patients. Biomarkers are defined as sensitive
sion tension (DT)-MRI can detect with in great detail alterations early measure of outcome than the currently available neurologi-
in the microstructure of the white matter and shows consid- cal scores assessed only 6 months after TBI. In addition due to the
erable promise in the assessment of axonal damage (Salmond common problem of TBI clinical trials being inadequate in patient
et al., 2006). However, these neuroimaging techniques reveal lit- numbers, the use of biomarkers could provide a more powerful
tle or no information regarding secondary injury processes such tool to detect outcome differences in TBI patient populations.
as excitotoxicity, neuroinflammation, blood-brain barrier (BBB) At present, the most studied TBI biomarker is S100B, a low-
breakdown, ischemic damage, and cell death. Biomarkers promise molecular weight calcium binding protein secreted by astrocytes.
to arm clinicians with all this additional, patient-specific infor- The validity of S100B as a potential TBI biomarker relies on its con-
mation. In the context of this review a biomarker is a molecule stitutively low expression in serum and cerebrospinal fluid (CSF),
that can be measured in a patient, which reflects the pathology of which is rapidly released into the CSF and serum following brain
TBI, how the pathology is likely to develop, and become predic- injury. A very strong correlation of S100B levels and severity of
tive factors for long term neurological outcome. It would be of injury has been reported (Savola et al., 2004), as well as a link
great benefit to the patient if an endogenous molecule could be between high S100B expression and unfavorable outcome (Her-
identified to have an expression profile, which can be linked to the rmann et al., 2001; Townend et al., 2002; Vos et al., 2004; Rainey
type or extent of secondary injury. With this information clini- et al., 2009). However, S100B is not ideal as a TBI biomarker
cians would be able to make more informed decisions regarding because it does not readily cross the BBB, its serum levels increase
treatments most likely to lead to an optimal outcome. Further- after peripheral trauma in the absence of brain injury (Ander-
more, detection of early biomarker levels would be invaluable in son et al., 2001; Savola et al., 2004; Torabian and Kashani-Sabet,
identifying patients that are most likely to benefit from a specific 2005), and it has not always been found to reliably predict outcome
experimental treatment. Indeed, the diverse nature of human TBI (Berger et al., 2007; Piazza et al., 2007). In the search for a reliable
has been cited as a potential explanation for lack of a successful biomarker of TBI, other molecules have been assessed, includ-
clinical trial (Statler et al., 2001). While it is now clear that there ing glial fibrillary acidic protein (GFAP), NSE, MBP, -II-spectrin
is no silver bullet with which all TBI patients can be treated, breakdown products (BDPs), ubiquitin C-terminal hydrolase-L1,
categorization of TBI patients using biomarkers in combination and various cytokines (Dash et al., 2010; Schiff et al., 2012).
with traditional methods might allow for specific treatments to be
given to those most likely to benefit. THE POTENTIAL OF INFLAMMATORY CYTOKINES AS
Identifying secondary injury mechanisms may also be useful BIOMARKERS OF TBI
in determining the type of injury that a person has received and One of the integral features of TBI is the inflammatory reaction
the degree of its progression in the long term. For example, the initiated and regulated by an array of pro- and anti-inflammatory
War on Terror has led to an increase in the incidence and aware- cytokines. Cytokines are small, short-lived proteins produced by
ness of blast injuries, which involve a rapid change in air pressure blood leukocytes and glial cells. They are quickly released in
around the body. Blast injury is becoming increasingly common, response to TBI and are rapidly sequestered. There are a large num-
yet symptoms often do not manifest until weeks or months after ber of different cytokines, many with overlapping functions that
the incident (Zeitzer and Brooks, 2008). Even a mild TBI can form a complex network of inflammatory mediators. Cytokines
leave survivors with long term cognitive deficits and behavioral that initiate or propagate an inflammatory response are said to
problems, which impact on their daily lives (Corrigan et al., 2004; be pro-inflammatory, while cytokines that inhibit the inflamma-
Pagulayan et al., 2006; Strandberg, 2009). Being able to specifi- tory response are called anti-inflammatory. The expression profile
cally predict which patients are likely to develop these neurological of each cytokine following brain injury has the potential to pro-
symptoms would enable doctors to make referrals to specific reha- vide information about the extent of tissue damage, and can be
bilitation programs, council patients and family members, and easily and rapidly measured via immunological assays. However,
encourage vigilance in reporting such changes. cytokine concentrations vary depending on the tissue or fluid they

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 2


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

are measured in (brain tissue, CSF blood, serum, plasma, etc.), and occurs between 12 and 24 h after injury (Fan et al., 1995; Ciallella
the temporal profile of the cerebral immune response in rodent et al., 2002; Ahn et al., 2004; Lu et al., 2005a,b, 2007; Kamm et al.,
versus human data can present differences as well as commonality. 2006; Maegele et al., 2007; Semple et al., 2010a,b; Shojo et al., 2010);
Some of the most studied cytokines with respect to brain injury and levels of IL-1 mRNA within 24 h of injury do appear to be
and their potential as biomarkers are discussed in this review. A associated with injury severity (Kinoshita et al., 2002). Despite the
summary of relevant literature is shown in Table 1. lack of direct correlations with brain damage or outcome, we have
shown that while IL-1 peaks at 2 h in the rat cortex following a dif-
INTERLEUKIN-1 fuse axonal injury, when combined with post-traumatic hypoxia
The Interleukin-1 (IL-1) family of cytokines are key mediators of the expression of IL-1 is significantly enhanced and prolonged
the inflammatory response peripherally and centrally. The IL-1 (Foda and Marmarou, 1994; Yan et al., 2011). The exacerbated
related molecules are perhaps the best known in relation to acute production of IL-1 is therefore important to consider, especially
TBI, having been widely studied in models of both focal and dif- when secondary injuries occur.
fuse injury (Fan et al., 1995; Benveniste, 1998; Yatsiv et al., 2002;
Brough et al., 2011). The IL-1 receptor type I (IL-1R) is thought to CLINICAL EVIDENCE
mediate many of the effects of the IL-1 cytokines, and is expressed IL-1 is barely detectable in the serum or CSF of healthy indi-
on multiple cell types in the brain (Holmin et al., 1997; Csuka et al., viduals, and has proved difficult to measure following human
2000; Pinteaux et al., 2002; Lu et al., 2005a). However, there is also TBI (Kossmann et al., 1996, 1997; Hergenroeder et al., 2010).
evidence to suggest that some of the effects of IL-1 cytokines are One recent study reported peak IL-1 in CSF of 1.425 pg/mL,
independent of the IL-1R (Touzani et al., 2002; Boutin et al., 2003; and serum concentrations of 0.87.6 pg/mL (Singhal et al., 2002).
Loscher et al., 2003). Intranuclear actions of IL-1 to regulate gene More recently, measurements of IL-1 concentrations in post-
transcription and RNA splicing may account for some of these mortem tissue from TBI patients have confirmed that a global
actions (Luheshi et al., 2009). upregulation occurs within a few minutes to hours of injury
The IL-1 family includes the closely related agonists IL-1 and (Frugier et al., 2010). Similar small increases in IL-1 concentra-
IL-1, the antagonist IL-1ra, and the other family member, the tions have been reported previously in stroke patients (Tarkowski
agonist IL-18 (Dinarello, 1994, 1998, 2009; Barksby et al., 2007). et al., 1995). Although changes in IL-1 expression in CSF and
Of these, the IL-1 isoform is by far the most often reported in TBI. serum following injury appear to be small, attempts have been
IL-1 is a pro-inflammatory cytokine and has been implicated in made to correlate IL-1 levels with outcome. Serum levels of IL-
the release of phospholipase-2 (PLA2), prostaglandins, and the 1 taken within 6 h of TBI have been found correlate with GCS
activation of cyclooxygenase-2 (COX-2; Chung and Benveniste, in a cohort of 48 patients (Tasci et al., 2003). In other studies
1990; Aloisi et al., 1992; Molina-Holgado et al., 2000; Rothwell, in severe brain injury patients high CSF concentrations of IL-1
2003). Furthermore, the primary mechanism of action for IL- were associated with poor outcome and increased ICP (Hayakata
1 is believed to be the regulation of release of other cytokines. et al., 2004; Shiozaki et al., 2005). In pediatric TBI, the CSF lev-
IL-1 has also been shown to play a role in apoptosis (Holmin els of IL-1 have been correlated with outcome assessed by the
and Mathiesen, 2000), adhesion of leukocytes to endothelial cells Glasgow outcome score (GOS; Chiaretti et al., 2005). Finally, in
(Bevilacqua et al., 1985), BBB disruption (Quagliarello et al., 1991), one study IL-1 and IL-1ra were measured in brain microdia-
and edema formation (Holmin and Mathiesen, 2000). The fun- lyzates of 15 TBI patients, and better outcomes were reported
damental pro-inflammatory and neurotoxic function of IL-1 is in patients with a high IL-1ra/IL-1 ratio (Hutchinson et al.,
demonstrated by studies that have aimed to attenuate IL-1 effects. 2007). Despite these results, other groups have failed to corre-
For example, the antagonist IL-1ra has been found to reduce neu- late IL-1 to ICP or outcome (Winter et al., 2004; Stein et al.,
ronal damage in rodent brain injury models (Relton and Rothwell, 2011).
1992; Yang et al., 1998). Improved cellular and behavioral out-
comes from brain injury have been reported in rats treated with TUMOR NECROSIS FACTOR
recombinant human IL-1ra (Toulmond and Rothwell, 1995), mice Tumor Necrosis Factor (TNF; formerly TNF) is a multifunctional
lacking the IL-1R (Basu et al., 2002), mice over-expressing IL-1ra cytokine most often referred to as a potent pro-inflammatory
(Tehranian et al., 2002), or by means of intraventricular admin- cytokine, produced by microglia and astrocytes. Early studies
istration of IL-1 or IL-1 antibodies to rats (Lu et al., 2005a,b). mostly in rat models of TBI, administration or inhibition of TNF
The intensive research into IL-1 with regard to TBI has led to it suggested that increased expression of TNF is detrimental (Ramilo
being considered as a biomarker of early neuroinflammation and et al., 1990; Kim et al., 1992; Shohami et al., 1996; Knoblach
consequent tissue damage. et al., 1999; Trembovler et al., 1999). However, more recent work
employing TNF and TNF receptor knockout mice have shown that
LABORATORY EVIDENCE mortality rates are increased and long term recovery impaired in
Studies in animal models of focal and diffuse TBI have consis- these models of focal TBI (Scherbel et al., 1999; Sullivan et al.,
tently shown that basal levels of IL-1 are very low (OConnor 1999; Stahel et al., 2000). These apparently conflicting data mirror
and Coogan, 1999; Krueger, 2008), and that an increase in IL-1 findings from other inflammatory mediators and demonstrate the
expression is detectable as early as 1 h after trauma (Fan et al., dual role of TNF as both a pro- and anti-inflammatory cytokine
1995; Kinoshita et al., 2002; Lu et al., 2005a,b; Kamm et al., 2006). (Shohami et al., 1999; Lenzlinger et al., 2001; Morganti-Kossmann
In rodent brain homogenates peak mRNA and protein expression et al., 2002; Schmidt et al., 2005).

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 3


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

Table 1 | Studies relevant to the development of cytokines as biomarkers of TBI.

Cytokine Species Injury/model Tissue/fluid Findings Reference

IL-1 Rat LFP, Brain homogenates Increase in mRNA expression occurs within 1 h Fan et al. (1995), Kinoshita et al.
weight-drop and peak mRNA and protein expression is (2002), Lu et al. (2005a), Lu et al.
between 12 and 24 h after injury (2005b), Kamm et al. (2006)
Rat Weight-drop Plasma No change in IL-1 expression following TBI Kamm et al. (2006)
Rat LFP Brain homogenates mRNA expression of IL-1 is higher in severe Kinoshita et al. (2002)
versus moderate injury severity
Rat DAI-hypoxia Brain homogenates Increased and prolonged IL-1 expression when Yan et al. (2011)
TBI is combined with post-traumatic hypoxia
Human TBI Post-mortem tissue Increased mRNA expression within minutes of Frugier et al. (2010)
injury
Human TBI CSF and serum Peak expression of IL-1 in CSF and serum Kossmann et al. (1996), Kossmann
following TBI is very low et al. (1997), Singhal et al. (2002),
Hergenroeder et al. (2010)
Human TBI Serum IL-1 levels within 6 h of injury correlate with Tasci et al. (2003)
GCS
Human Severe TBI and CSF Elevated IL-1 expression associated with poor Hayakata et al. (2004), Chiaretti et al.
pediatric TBI outcome and increased ICP (2005), Shiozaki et al. (2005)
Human TBI Brain parenchyma, No correlation of IL-1 expression with ICP or Winter et al. (2004), Stein et al. (2011)
CSF, serum outcome

IL-1/IL-1ra Human Severe TBI Brain parenchyma High IL-1ra/IL-1 ratio is associated with better (Hutchinson et al. (2007)
outcome

TNF Rat TBI Brain homogenates, Increased mRNA and protein expression Taupin et al. (1993), Shohami et al.
brain slices detectable at 1 h, and peak expression between (1994), Fan et al. (1996), Knoblach et al.
4 and 8 h post-TBI (1999), Dalgard et al. (2012)
Rat LFP Brain homogenates TNF expression increases after severe TBI, but Knoblach et al. (1999)
not mild TBI
Rat DAI-hypoxia Brain homogenates DAI and post-traumatic hypoxia leads to Yan et al. (2011)
increased expression of TNF versus DAI alone
Rat CCI CSF Peak levels of TNF in CSF are not reached until Stover et al. (2000)
24 h after TBI
Human TBI CSF, serum, plasma TNF is increased in CSF, serum, and plasma Goodman et al. (1990), Ross et al.
following TBI (1994), Morganti-Kossmann et al.
(1997), Csuka et al. (1999)
Human TBI Post-mortem tissue TNF mRNA and protein can be detected in the Frugier et al. (2010)
brain within minutes of injury
Human Severe TBI CSF TNF protein concentrations peak in the CSF Hayakata et al. (2004)
within 24 h
Human Severe TBI CSF, serum Six hours after TBI, TNF expression is higher in Shiozaki et al. (2005)
CSF than in serum. TNF expression does not
correlate with outcome
Human Severe TBI CSF, serum Increased serum TNF levels correlate with Stein et al. (2011)
increased ICP and decreased CPP, but not
outcome. TNF concentrations in CSF not linked
to ICP, CPP, or outcome

IL-10 Rat LFP Brain homogenates IL-10 expression increases rapidly, and remains Knoblach and Faden (1998)
elevated from 4 to at least 20 h after TBI
Rat CCI Brain homogenates IL-10 expression is reduced in the brains of TBI Lee et al. (2012)
versus sham animals 1 day after surgery

(Continued)

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 4


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

Table 1 | Continued

Cytokine Species Injury/model Tissue/fluid Findings Reference

Human Severe TBI CSF, serum, plasma IL-10 expression in both CSF and serum Csuka et al. (1999), Maier et al.
increases rapidly following TBI, and is higher in (2001), Woiciechowsky et al. (2002)
CSF than in serum or plasma. There is no
correlation of IL-10 with BBB integrity
Human Severe TBI CSF, serum IL-10 expression is higher in serum than in CSF Hayakata et al. (2004)
following TBI
Human Pediatric TBI CSF Increased IL-10 levels in CSF are linked to Bell et al. (1997)
mortality
Human Severe TBI CSF, serum Increased IL-10 is linked to BBB dysfunction and Kirchhoff et al. (2008)
mortality
Human Severe TBI CSF IL-10 expression is higher in patients that had an Shiozaki et al. (2005)
unfavorable outcome
Human Severe TBI CSF, serum No link between IL-10 and outcome Maier et al. (2001), Woiciechowsky
et al. (2002), Lo et al. (2009, 2010),
Stein et al. (2011)

IL-6 Rat, mouse LFP, PBBI, Brain homogenates, IL-6 expression is undetectable in normal brain, Woodroofe et al. (1991), Taupin et al.
weight-drop brain parenchyma but increases rapidly, peaking at 28 h following (1993), Shohami et al. (1994),
TBI Maegele et al. (2007), Williams
et al. (2007), Ziebell et al. (2011),
Weckbach et al. (2012)
Rat CCI, DAI CSF, serum IL-6 expression is higher in CSF than in serum. Woodroofe et al. (1991), Hans et al.
IL-6 expression increases from 1 h and peaks at (1999), Stover et al. (2000)
25 h after injury
Rat CHI, Serum IL-6 expression in serum cannot discriminate Maegele et al. (2007), Weckbach
poly-trauma between peripheral and CNS injuries et al. (2012)
Human TBI CSF, serum Following TBI IL-6 expression increases to a Kossmann et al. (1996), Winter et al.
greater extent in CSF than serum (2004), Hillman et al. (2007),
Chiaretti et al. (2008)
Human TBI Plasma IL-6 concentrations greater than 100 pg/mL are Woiciechowsky et al. (2002)
associated with severe TBI
Human TBI Plasma Increased IL-6 concentrations correlate with Arand et al. (2001), Woiciechowsky
poor outcomes et al. (2002)
Human Pediatric TBI Serum No correlation between IL-6 levels and outcome Kalabalikis et al. (1999)
Human TBI Serum IL-6 levels within 17 h of injury can be used to Hergenroeder et al. (2010)
predict elevated ICP
Human TBI Brain parenchyma Higher parenchymal levels of IL-6 correlate with Winter et al. (2004)
better outcomes
Human TBI Brain parenchyma No relationship between IL-6 and ICP, brain Perez-Barcena et al. (2011)
oxygenation, or edema

IL-8 Rat, mouse CCI, Brain homogenates IL-8 functional homologs CXCL-1 and CXCL-2 Otto et al. (2002), Dalgard et al.
weight-drop exhibit peak expression at 412 h after TBI (2012)
Human TBI CSF, serum Following TBI, increased IL-8 expression can be Kossmann et al. (1997),
measured in CSF and to a lesser extent in serum Morganti-Kossman et al. (1997),
Whalen et al. (2000), Kushi et al.
(2003a)
Human Pediatric TBI CSF IL-8 levels following TBI correlate with mortality Whalen et al. (2000)
Human Severe TBI CSF, plasma Lower IL-8 levels in plasma are associated with Gopcevic et al. (2007)
survival. CSF IL-8 levels do not vary between
survivors and non-survivors
Human Severe TBI, Serum Increases in IL-8 after TBI correlate with Mussack et al. (2002), Kushi et al.
pediatric TBI unfavorable outcome and are associated with (2003b); Lo et al. (2010)
mortality

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 5


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

LABORATORY EVIDENCE ICP, or neurological outcome. More recently, Stein et al. (2011)
In injured rat brain, increased TNF mRNA can be detected prior analyzed CSF and serum samples from 24 patients at 12 h inter-
to the cytokine protein itself, and upregulation of TNF was shown vals for 7 days after having sustained a severe TBI. In the same
to precede leukocyte infiltration to the site of injury (Riva-Depaty patients ICP and CPP were continually monitored so that associ-
et al., 1994; Shohami et al., 1997). This suggests that TNF is pro- ations between cytokine levels and subsequent changes in ICP or
duced early by resident brain cells in response to neuronal injury. CPP could be investigated. They reported that increased serum,
Increases in TNF protein have been measured at 1 h, and peak lev- and not CSF, concentrations of TNF moderately correlate with
els were found between 4 and 8 h after injury (Taupin et al., 1993; subsequent increases in ICP or decreases in CPP. However, they
Shohami et al., 1994; Fan et al., 1996; Knoblach et al., 1999; Dal- did not find any relationship of any cytokine concentration [IL-1,
gard et al., 2012). Early increases in TNF expression could prove Interleukin-6 (IL-6), Interleukin-8 (IL-8), Interleukin-10 (IL-10),
to be useful in the clinical setting as a diagnostic/prognostic fac- or TNF] with outcome (GOS < 5).
tor. In fact TNF was shown to reflect injury severity, since one
study using the lateral fluid percussion (LFP) injury reported that INTERLEUKIN-10
while increases could be measured for severe injury, no change Interleukin-10 is regarded to be primarily an anti-inflammatory
in TNF was recorded for a mild injury (Knoblach et al., 1999). cytokine, having a potent inhibitory effect on production of
Furthermore, in a recent study from our laboratory, we found several pro-inflammatory mediators including IL-1 and TNF,
that TNF is only increased in rats subjected to a combined diffuse but also IL-1, granulocyte-macrophage colony stimulating fac-
brain injury and hypoxia, but not in diffuse brain injury alone tor (GM-CSF), IL-6, IL-8, IL-12, and IL-18 (de Waal Malefyt
(Yan et al., 2011). In contrast to previous published work, when et al., 1991, 1993; Fiorentino et al., 1991; DAndrea et al., 1993;
using the closed brain injury model of focal TBI, we did not find Gruber et al., 1994). The inhibition of IL-1 and TNF is its
increased expression of TNF in brain homogenates over 24 h (Bye most important function, since these cytokines are known to
et al., 2007; Semple et al., 2010a). Another interesting finding from play central roles in initiation and propagation of the inflam-
animal studies is a difference in the timing of TNF peaks in the matory response. Indeed, rats subjected to LFP injury and
parenchyma versus CSF of rats. In brains of rats subjected to LFP treated with IL-10 have improved outcomes and reduced lev-
injury TNF expression peaks at 46 h and is resolved by 24 h (Fan els of IL-1 and TNF in brain tissues (Knoblach and Faden,
et al., 1996; Knoblach et al., 1999), yet in the controlled corti- 1998).
cal impact (CCI) model TNF expression in the CSF did not peak
until 24 h post-injury (Stover et al., 2000). Whether this represents LABORATORY EVIDENCE
a differential response to injury type, or a delay in movement of Although the anti-inflammatory properties of IL-10 following TBI
cytokine from parenchyma to CSF remains to be determined. are well established, there is relatively little information regarding
the expression profile of IL-10 following TBI in animals. Using
CLINICAL EVIDENCE the FPI model, one study found that in the brain IL-10 increased
Following TBI increases in TNF levels have been reported in the rapidly during the first 4 h following injury and remained elevated
CSF and serum of patients (Goodman et al., 1990; Ross et al., for at least 20 h thereafter (Knoblach and Faden, 1998). However,
1994). In work from our group TNF concentrations were mea- a more recent study reported a reduction in IL-10 expression in
sured in the CSF and serum of TBI patients at 24 h intervals, brains of rats 1 day after CCI (Lee et al., 2012). No changes instead
and TNF was significantly elevated from controls (Morganti- were shown in diffuse brain injured rats over 4 days post-injury,
Kossmann et al., 1997; Csuka et al., 1999). Given that TNF expres- whether with or without the addition of hypoxia (Yan et al., 2011).
sion in animal models usually peaks and resolves within the first This suggests that such anti-inflammatory cytokine may play a
24 h this is perhaps not surprising. The delayed and sustained role in a delayed phase after TBI. The discrepancy between the two
increase in TNF measurements in human CSF (3 days to 3 weeks) studies indicates that there may be a differential expression profile
as compared to rapid fluctuations observed in brain tissue may of IL-10 based on the type of injury, since the weight-drop model
reflect different mechanisms of cytokine metabolism and degrada- produces a diffuse axonal injury in the absence of focal damage,
tion in these environments. Indeed, more recently we have shown whereas CCI is primarily a focal injury.
that TNF mRNA and protein can be detected in post-mortem
brain tissue from TBI patients that died within 17 min of injury CLINICAL EVIDENCE
(Frugier et al., 2010). In another study, Hayakata et al. (2004) We have shown that IL-10 is elevated in the CSF and serum of
examined CSF from 23 severe TBI patients and a peak in TNF of patients with isolated, severe TBI (Csuka et al., 1999). In the CSF
2030 pg/mL was recorded within 24 h. They then attempted to IL-10 was elevated in 26 out of 28 patients (range: 1.341.7 pg/mL)
analyze the associations of TNF levels with raised ICP and poor versus controls, but in serum only 7 patients displayed elevated
outcome (GOS < 4 at 6 months), but no correlation was found. In IL-10 (range: 5.423 pg/mL). The temporal profile was similar in
a subsequent study the same group measured TNF in both the CSF both fluids, exhibiting a rapid early rise and peak followed by a
and serum of 35 TBI patients with or without additional injury slow decline. In addition to cytokine measurements, BBB function
at exactly 6 h after injury (Shiozaki et al., 2005). They reported has been assessed in TBI patients using the CSF/serum albumin
that TNF levels were higher in CSF (median 18 pg/mL) versus ratio, and no correlation between the two variables has been found
serum (median 5 pg/mL) regardless of the presence of additional (Csuka et al., 1999; Maier et al., 2001). The lack of association of
injury. Again, there was no correlation between TNF and GCS, IL-10 levels with BBB dysfunction, combined with the fact that

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 6


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

IL-10 CSF levels exceeded serum levels in most patients suggest Shohami et al., 1994; Williams et al., 2007; Ziebell et al., 2011).
an intrathecal origin for this cytokine. However, not all studies In rodent models, experimental TBI induces an increase in IL-6
have corroborated this hypothesis, since serum levels have been mRNA expression in brain tissue after 1 h (Williams et al., 2007),
reported to be significantly higher than CSF levels in some studies and peaks in protein expression have been reported between 2 and
(Hayakata et al., 2004). The presence of additional injuries could 8 h after injury (Taupin et al., 1993; Shohami et al., 1994; Hang
easily account for this difference (Hensler et al., 2000; Dziurdzik et al., 2004; Ziebell et al., 2011). In CSF, increases in IL-6 protein
et al., 2004; Shiozaki et al., 2005). Other studies have confirmed can be detected within 1 h, with peak expression between 2 and 5 h
that in severe TBI IL-10 expression increases early, reaching a peak after an experimental brain injury (Woodroofe et al., 1991; Hans
within 28 h of injury (Woiciechowsky et al., 2002). Higher lev- et al., 1999; Stover et al., 2000). The rapid increase in IL-6 expres-
els of IL-10 have been linked to better outcome in some studies, sion following injury, and its maximal levels detected within a few
but not in others. For instance, an early study reported a link hours makes this cytokine a promising candidate biomarker. How-
between increased IL-10 levels in CSF and mortality in pediatric ever, it may have limited utility in stratification of patients since a
TBI (Bell et al., 1997), and more recent work in adult severe TBI similar temporal profile of IL-6 production has been demonstrated
made a similar link between increased IL-10 and mortality (Kirch- in most studies irrespective of the injury model used (weight-drop,
hoff et al., 2008). The concentration of IL-10 in the CSF has also FPI, CCI, or stab wound). The concentration of IL-6 in serum is
been shown to be higher in patients that have an unfavorable out- rarely reported, but lower magnitude increases can be detected fol-
come (GOS < 4) assessed 6 months after injury (Shiozaki et al., lowing injury (Maegele et al., 2007; Weckbach et al., 2012). Since
2005). However, other studies have failed to find any connection the primary source of IL-6 following TBI originates in the brain,
between IL-10 and outcome (Maier et al., 2001; Woiciechowsky a limited ability of IL-6 to cross the BBB could explain in part
et al., 2002). In pediatric TBI, Lo et al. measured serum IL-10 lev- this discrepancy. Indeed, studies in rodents and ovines have indi-
els on day 1, and found they could not differentiate severe and cated that IL-6 has a limited ability to cross the BBB following a
non-severe injury or predict favorable outcome (Lo et al., 2009), peripheral injection (Banks et al., 1994), and that a specific and
even when paired with GCS (Lo et al., 2010). In a more recent saturable transport mechanism is involved in movement of IL-
study there was no correlation of IL-10 in serum or CSF with out- 6 across the BBB (Threlkeld et al., 2010). These findings suggest
come assessed at 6 months using the GOSE scores (Stein et al., that measurement of IL-6 in serum is unlikely to be truly indica-
2011). The IL-10 response to peripheral injuries as reported in tive of brain concentrations, but rather the integrity of the BBB.
multi-trauma patients could be part to blame for the difficulty in Furthermore, since peripheral injuries can lead to changes in cir-
making associations between TBI variables and IL-10 levels (Shi- culating IL-6 levels, models of poly-trauma are being developed
monkevitz et al., 1999; Hensler et al., 2000; Dziurdzik et al., 2004; (Maegele et al., 2005; Weckbach et al., 2012). Using these models,
Shiozaki et al., 2005). the specificity of serum IL-6 as a biomarker for brain injury has
been found to be poor. While serum IL-6 concentrations were sig-
INTERLEUKIN-6 nificantly higher in poly-trauma versus LFP or tibia fracture alone
Interleukin-6 has been extensively studied, and has been found to at 6 and 24 h after injury, there was no difference between the
be involved in a large number of physiological and pathophysio- latter two groups (Maegele et al., 2007). In another model involv-
logical processes. IL-6 is known to regulate inflammation, immu- ing combined blunt bilateral chest trauma, lower limb fracture,
nity, bone metabolism, hematopoiesis, and neural development and closed head injury (CHI), serum IL-6 was measured at 2 and
(Romano et al., 1997). In addition, a role for IL-6 has been impli- 4 h after injury and found to be significantly higher in three-hit
cated in aging, osteoporosis, autoimmune disease, Alzheimers dis- poly-trauma (CHI, chest trauma, and limb fracture) versus other
ease, and brain injury. Although IL-6 is not exclusively expressed groups at 4 h (Weckbach et al., 2012). Again there were no differ-
in the CNS, it does exhibit a significant upregulation following ences between single-hit groups or even combined CHI and chest
brain injury (Morganti-Kossmann et al., 1992; Kossmann et al., trauma versus chest trauma and limb fracture. Therefore, current
1995). data on IL-6 in animal models is controversial. The rapid increase
in IL-6 expression observed after brain injury and its peak within
LABORATORY STUDIES hours make it suited as a biomarker. Unfortunately, its limited abil-
Laboratory studies have shown that in the brain IL-6 is expressed ity to cross the BBB and apparent lack of ability to discriminate
by astrocytes (Benveniste et al., 1990; Van Wagoner and Benveniste, injury types may limit its usefulness.
1999), microglia (Woodroofe et al., 1991; Sebire et al., 1993), and
neurons (Schobitz et al., 1993; Gadient and Otten, 1994; Ringheim CLINICAL EVIDENCE
et al., 1995; Sallmann et al., 2000). It inhibits the synthesis of Under physiological conditions in humans, IL-6 expression in
TNF (Aderka et al., 1989), induces synthesis of nerve growth fac- plasma is accepted as being 042 pg/mL, whereas in CSF there
tor (NGF; Kossmann et al., 1996), inhibits N -methyl-d-aspartate are only a few studies which have detected IL-6 under physio-
(NMDA) mediated toxicity (Wang et al., 2009), and promotes neu- logically normal conditions (Froon et al., 1996; Kossmann et al.,
ronal differentiation and survival (Islam et al., 2009). Evidence 1996; Maier et al., 2005). The largest of these studies measured
suggests that expression of IL-6 is beneficial following neuronal IL-6 in the CSF of 113 patients, and reported IL-6 concen-
injury (Penkowa et al., 2000, 2003). While IL-6 is often unde- trations of 123 pg/mL (Maier et al., 2005). Following injury,
tectable in normal brain, its acute release in response to injury IL-6 concentrations in CSF can reach concentrations as high
is well documented (Woodroofe et al., 1991; Taupin et al., 1993; as 35,500 pg/mL, but there appears to be a lot of variability

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 7


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

in this response (Kossmann et al., 1996; Hillman et al., 2007). In inconclusive. Its inability to discriminate between brain damage
addition, increases in serum IL-6 have been also been reported and peripheral injuries may limit its usefulness in poly-trauma
after TBI, with lower peak concentrations of 93269 pg/mL (Win- patients, but further development of parenchymal microdialy-
ter et al., 2004; Chiaretti et al., 2005). Plasma levels of IL-6 greater sis and its use in combination with other biomarkers may prove
than 100 pg/mL in the first 24 h following injury have been found fruitful.
to be associated with severe brain injury (Woiciechowsky et al.,
2002). Data on the predictive ability of IL-6 in serum is limited and INTERLEUKIN-8/CXCL8 AND MONOCYTE
conflicting; in pediatric TBI serum IL-6 was reported to have no CHEMOATTRACTANT PROTEIN/CCL2
associations with neurological outcome (Kalabalikis et al., 1999), Interleukin-8 is a member of the CXC chemokine family (CXCL8),
while others demonstrated that high IL-6 correlated with poor and is secreted by glial cells, macrophages, and endothelial cells
outcome (Arand et al., 2001; Woiciechowsky et al., 2002). More (Aloisi et al., 1992; Nitta et al., 1992; Zhang et al., 2011). It is an
recently, measurements of serum IL-6 within 17 h of injury have important mediator for the activation and chemotaxis of neu-
been shown to identify patients at risk of developing elevated ICP trophils (Bickel, 1993). Early studies showed that IL-8 is released
(Hergenroeder et al., 2010). However, the authors also noted that from astrocytes in response to other cytokines including IL-1 and
lack of prognostic value of IL-6 for elevated ICP when patients TNF (Kasahara et al., 1991), both of which are expressed soon after
also presented with extracranial injuries. Indeed, the presence of brain injury (McClain et al., 1987; Woodroofe et al., 1991; Taupin
multiple injuries is common in TBI patients (Gennarelli et al., et al., 1993). Increased IL-8 expression has been reported in many
1994; Meixensberger and Roosen, 1998) and should be considered cancers (Xie, 2001), bacterial infections (Hirao et al., 2000), and is
when searching for or with the purpose of developing a biomarker. linked to cardiovascular disease (Apostolakis et al., 2009).
This is substantiated by evidence showing increased serum con- The monocyte chemoattractant protein-1 (MCP-1) or CCL2 is
centrations of IL-6 following orthopedic injury (Hergenroeder produced by astrocytes within hours after injury and its levels cor-
et al., 2010), burns (Agay et al., 2008), and exercise (Nybo et al., relate with the amounts of recruited macrophages (Semple et al.,
2002; Febbraio and Pedersen, 2005). The poor predictive ability 2010c). Since MCP-1 is regulated in an autocrine fashion, subse-
of IL-6 in the presence of multiple injuries is not surprising, and quent release of MCP-1 by infiltrated macrophages and microglia
is corroborated by studies on experimental poly-trauma models perpetuates cell migration in the injured brain. MCP-1 overexpres-
described above. However, since the primary source of IL-6 in sion increased macrophage infiltration and neurological deficit in
TBI are the cells of the CNS, including microglia, astrocytes, and ischemia whereas its deletion attenuated infiltrates in brain injury,
neurons (Marz et al., 1998; Van Wagoner et al., 1999; Lau and Yu, stroke, and multiple sclerosis models (Lu et al., 1998; Huang et al.,
2001), more specific information on injury to the brain might be 2001; Hughes et al., 2002; Chen et al., 2003).
obtained from measurement of parenchymal cytokine production.
This can be achieved using the technique of cerebral microdialysis, LABORATORY EVIDENCE
which can be adapted to recover cytokines and allow for continual Rodents lack a direct homolog for IL-8, but chemokine (CXC
sampling of brain parenchyma (Winter et al., 2002; Helmy et al., motif) ligand-1 (CXCL-1), CXCL-2, CXCL-5, and CXCL-6 appear
2007). A wide range of cytokine concentrations can be measured to be functional homologs, residing in the same gene cluster as IL-
in brain injured patients using this technique (Helmy et al., 2011). 8 (human chromosome 4q13.3) and contributing to neutrophil
In a study involving 14 severe TBI patients, higher peak parenchy- recruitment in a number of animal models. In rat CCI, the expres-
mal levels of IL-6 were found to correlate with better GOS (Winter sion of the chemokine CXCL-1 peaks at 4 h, and is reduced by
et al., 2004). Concentrations of NGF were also measured in this 12 h, but remains elevated versus control for up to 7 days after
study, and while overall levels of NGF or IL-6 alone could not injury (Dalgard et al., 2012). In our model of CHI, the synthesis
predict outcome, the ratio of NGF:IL-6 was significantly lower of MIP-2 as well as other chemokines (MCP-1, MIP-1, RANTES,
in survivors, and was correlated with GCS and GOS. In a more and KC) in the cortex increased at 412 h preceding the infil-
recent study including 16 patients with diffuse TBI, there was no tration of neutrophils and macrophages which peak at 2448 h
relationship between parenchymal levels of IL-6 and ICP, brain tis- and 47 days, respectively. MIP-1 and MIP-2 concentration was
sue oxygenation, or the presence of brain swelling (Perez-Barcena reduced in the brain of TNF-KO mice after TBI, implying a role for
et al., 2011). However, it must be noted that this study used aver- TNF in regulating their expression (Otto et al., 2002). Amplified
ages of samples collected over 8-h periods, and could have missed expression of chemokine receptors CXCR2 and CCR2 was local-
some of he large-scale changes in cytokine concentrations known ized on infiltrating neutrophils and macrophages, respectively at 1
to occur in TBI. Furthermore, both of these studies suffered from and 4 days post-TBI (Otto et al., 2001; Semple et al., 2010b). While
a lack of statistical power, having relatively few patients. While neutrophils depart by 1 week from the injured brain, the accumu-
further development of microdialysis techniques will undoubt- lation of macrophages persists over 4 weeks (Semple et al., 2010a).
edly provide us with some very useful information regarding the The prolonged presence of activated leukocytes within the peri-
brains response to injury, it is limited by its invasive nature, the contusional tissue is likely detrimental due to their ability to secrete
expense of the probes, and the highly region-specific information neurotoxins leading to delayed neuronal death. The role of MCP-1
obtained. and IL-8/MIP-2 in secondary brain degeneration and neurological
In summary, IL-6 is highly sensitive to brain injury and can function was recently explored using MCP-1-KO and CXCR2-KO
be easily detected in serum, although current data on its ability mice. The most striking data in MCP-1-KO mice showed a signif-
to predict outcome and its correlation with ICP are limited and icant reduction in lesion volume, neuronal loss and macrophage

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 8


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

accumulation up to 46% over 4 weeks after TBI as opposed to wild- Upregulation of both IL-8 and MCP-1 at mRNA and pro-
type mice (Semple et al., 2010a). Improved brain damage resulted tein level in post-mortem human brain, underlining the rel-
in faster neurological recovery from 1 to 4 weeks. In CXCR2-KO evance of the chemokine network in human TBI (Frugier
mice, an 80% decline of neutrophil infiltration occurred at 12 h et al., 2010). Specifically, a 140-fold increase in IL-8 mRNA
after TBI and coincided with reduced lesion and neuronal loss over was detected in the injured brain compared to control, being
wild-type controls (Semple et al., 2010b). the mediator with the strongest increase, while MCP-1 mRNA
was increased by almost 20-fold (unpublished data). Overex-
CLINICAL EVIDENCE pression of chemokines in these human samples was associ-
In humans, IL-8 is detected at very low levels in the CSF and ated with tissue infiltration of CD68-positive macrophages and
plasma of healthy individuals. Physiological plasma concentra- GFAP-positive reactive astrocytes (Frugier et al., 2010). Com-
tions are 518 pg/mL, and in CSF are 572 pg/mL (Maier et al., bined, these experimental studies provide compelling evidence
2005). Following TBI there is an increase in IL-8 concentration that signaling through chemokine networks contributes to sec-
in serum and CSF (Kossmann et al., 1997; Morganti-Kossman ondary tissue and neurological damage and could be consid-
et al., 1997). IL-8 appears to peak early following a TBI, with mean ered in future studies assessing their meaning as biomarkers
levels up to 29,000 pg/mL reported in CSF (Whalen et al., 2000; of TBI.
Kushi et al., 2003a). Increases in plasma levels of IL-8 following
brain trauma have been reported, but are of lower magnitude and OTHER CYTOKINES
more variable. Peak concentrations of approximately 100 pg/mL The cytokines described above are the best characterized in terms
are commonly demonstrated in severe TBI (Kossmann et al., 1997; of their role in neuronal injury and their potential as markers for
Mussack et al., 2002; Kushi et al., 2003a). Increased IL-8 in CSF TBI, however, there are a large number of less-well characterized
has been associated with mortality. In one study, CSF was obtained immune modulators that could be useful TBI biomarkers.
from 27 children who had sustained a severe TBI, 7 with menin- Another potential biomarker is GM-CSF, a pro-inflammatory
gitis, and 24 children with neither diagnosis. The increase in IL-8 cytokine that is expressed in the CNS by neurons, astrocytes,
levels in children with TBI was of similar magnitude to children and microglia (Franzen et al., 2004). GM-CSF is secreted by vas-
with meningitis. Heightened IL-8 expression persisted for several cular endothelial cells, it crosses the BBB and can be detected
days, and a significant correlation was found between IL-8 and in CSF (Coxon et al., 1999). We have found this cytokine to
mortality (Whalen et al., 2000). In another study, Gopcevic et be significantly upregulated in post-mortem tissue from TBI
al. collected blood and CSF samples at time of admittance from patients that died 6122 h after injury (Frugier et al., 2010).
20 adults who had sustained severe TBI, 10 of which died. They More recently we have found that GM-CSF is more highly
showed that plasma IL-8 was significantly lower in survivors versus expressed in the CSF of patients suffering from secondary hypoxia
non-survivors (71 and 111 pg/mL respectively), however, CSF IL- versus normoxic TBI patients, and is also increased in dif-
8 concentrations were not different. They also showed that IL-8 fuse TBI versus focal TBI (unpublished results). The differ-
had a prognostic value for GCS, patient age, and Acute Physio- ent levels of expression of this cytokine between injury types
logic and Chronic Health Evaluation (APACHE II; Gopcevic et al., merits further investigation of this molecule as a TBI bio-
2007). Although plasma levels of IL-8 are considerably lower than marker.
CSF, several studies have found correlations between peripheral Also of potential interest is endothelial monocyte-activating
IL-8 and outcomes. Mussack et al. (2002) measured serum IL-8 polypeptide II precursor (p43/pro-EMAP II), a pro-inflammatory
at intake and 12 h later in 20 TBI patients and found a significant cytokine linked with apoptosis (Knies et al., 1998; van Horssen
correlation of increased IL-8 levels 12 h after admission with out- et al., 2006). In a high-throughput immunoblotting screen of 998
comes assessed by GOS. In another study, serum concentrations proteins in rats 24 h after ischemic injury versus penetrating TBI
of IL-8 72 h after TBI were significantly higher in non-survivors differential expression was found in only the cytokine p43/pro-
versus survivors (Kushi et al., 2003a). Most recently, Lo et al. took EMAP II (Yao et al., 2008). In a subsequent study, tissue, blood,
blood samples from 28 pediatric TBI patients at precisely 24 h and CSF concentrations of this cytokine were shown in vivo to dis-
following injury and correlated serum biomarker levels with unfa- criminate between ischemic brain injury and TBI modalities (Yao
vorable outcomes 6 months later (GOS < 4). Increased serum IL-8 et al., 2009). Specifically, Yao et al. found that p43/pro-EMAP II is
was found to correlate with unfavorable outcome. Furthermore, constitutively expressed in the brain of nave rats, but significantly
when combined with GCS and increased specificity, an sensitivity increases in CSF and plasma 24-h after penetrating ballistic-like
was observed (Lo et al., 2010). The weakness of this study was brain injury, whereas a significant decrease was found in CSF and
that only 4 patients had an unfavorable outcome, nonetheless, it plasma following middle cerebral artery occlusion. Western blot-
demonstrates the potential for using paired markers to predict ting of brain tissue at 6, 24, 48, and 72 h showed similar increases
outcomes with greater accuracy. in p43/pro-EMAP II expression at all time-points post-TBI, and
Our group has demonstrated that in CSF samples from significant decreases in expression following ischemic brain injury.
21 severe TBI patients, MCP-1 concentrations increased to Immunohistochemistry revealed that changes in p43/pro-EMAP
19,000 pg/mL on day 1, falling to approximately 3000 pg/mL from II levels were due to changes in neuronal expression and decreases
day 3 onward (Semple et al., 2010a). The rapid peak in CCL2 levels did not represent neuronal loss. Taken together, these data show
in CSF and its elevated expression in the CSF for several days merit that the little-studied p43/pro-EMAP II cytokine has potential to
investigation of this cytokine as a potential biomarker for TBI. be a useful brain-specific biomarker.

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 9


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

OTHER MARKERS OF INFLAMMATION method for predicting outcomes. Neurological assessment has
In addition to cytokines, several other molecules could potentially been shown to be predictive for outcome, but cannot be admin-
be useful as biomarkers of brain injury. For example, activation istered within the first critical hours of injury (Lammertse et al.,
of the JAK/STAT pathway by IL-6 regulates GFAP expression. It is 2007). Biomarkers for SCI could allow clinicians to make earlier
well established that GFAP expression increases in serum follow- prognoses and decide on the best course of treatment. Research
ing TBI (Missler et al., 1999; van Geel et al., 2002). In severe TBI into biomarkers of SCI is very limited and has focused almost
patients, serum GFAP levels have been shown to be able to pre- exclusively on S100B and NSE. Laboratory research has shown
dict mortality and outcome (Pelinka et al., 2004a,b; Nylen et al., that in the CSF, expression of S100B, NSE, and neurofilament pro-
2006; Vos et al., 2010). More recently, GFAP-BDPs in serum of tein (NFL) are increased (Skouen et al., 1999; Nagy et al., 2002;
mild and moderate TBI patients within 4-h of injury has been Cao et al., 2008). A similar increase can be measured in the serum
found to correlate with injury severity (GCS), and maybe be asso- of animals subjected to an experimental SCI (Ma et al., 2001; Loy
ciated with CT lesions (Papa et al., 2012). In a recent study it was et al., 2005; Cao et al., 2008). There is some evidence from these
found that combining measurements of GFAP in CSF and serum studies that NSE and S100B can distinguish between injury sever-
with the IMPACT Outcome Calculator a significant improvement ities. Both NSE and S100B expression was shown to correlate with
in outcome prediction could be achieved (Czeiter et al., 2012). In injury severity in a rat weight-drop model of SCI (Cao et al., 2008).
addition, the expression of II-spectrin breakdown product 145 However, a similar study failed to find a difference in serum or CSF
(SBDP145) was measured in CSF and correlated significantly with NSE expression between graded injury levels (Loy et al., 2005).
6-month mortality (Czeiter et al., 2012). Another study measured Human studies into SCI biomarkers have focused on detecting
SBDPs in the CSF of severe TBI patients at the time of admis- ischemic injury in patients undergoing thoracoabdominal aortic
sion and every 6 h thereafter for up to 7 days (Mondello et al., aneurism surgery (van Dongen et al., 1998, 1999; Kunihara et al.,
2010). It was shown that in addition to an increased expression of 2001; Winnerkvist et al., 2007), and other studies have looked at
SBDPs in all TBI patients versus controls, there was a significant S100B or NSE after surgery for lumbar disk herniation (Brisby
correlation of SBDP145 with injury severity (assessed by GCS). et al., 1999), spinal epidural empyema (Marquardt et al., 2004b),
Furthermore, levels of SBDP145 and SBDP120 were significantly or paresis due to spinal metastasis (Marquardt et al., 2004a). Data
higher in patients that died, suggesting that these markers may be from these studies is inconclusive with regards to either S100B or
able to predict mortality (Mondello et al., 2010). Another marker NSE being useful as markers of ischemic injury. While several stud-
of note is the microtubule-associated proteins (MAP-2), which are ies have reported increases in expression of these biomarkers, little
neuronal specific proteins found in dendrites (Drewes et al., 1998). is known with regard to how this relates to outcome. In the case of
Laboratory studies in models of ischemic and traumatic injury spinal epidural empyema or paresis due to spinal metastasis, longer
have established that MAP-2 expression is lost from injured brain periods of elevated serum S100B were related to unfavorable out-
regions, and increases in MAP-2 expression can be detected in come (Marquardt et al., 2004a,b). Lastly, two studies have assessed
serum shortly after injury (Kitagawa et al., 1989; Posmantur et al., biomarkers for human traumatic SCI. The first measured GFAP
1996; Park et al., 2012). More recently, it has been shown that serum and NFL expression in CSF of patients with traumatic SCI, and
MAP-2 expression measured 6 months after severe TBI is still ele- although both markers were increased, no statistical analysis was
vated. Furthermore, increased serum MAP-2 expression correlates done and only six patients were included (Guez et al., 2003). The
with better outcome (GOSE), and was found to be higher in non- second study took CSF from 27 patients with complete or incom-
vegetative state patients versus vegetative state patients (Mondello plete SCI and measured protein expression of IL-6, IL-8, MCP-1,
et al., 2012). This suggests that MAP-2 has potential as a marker S100B, and GFAP (Kwon et al., 2010). All of the markers were
for outcome prediction, and increased serum MAP-2 expression increased in SCI patients, and there were significant differences in
may signal the emergence of higher cognitive function in severe expression of each marker between injury severities. Furthermore,
TBI patients. Lastly, the inflammasome is responsible for the pro- a model was developed that accurately predicted injury severity
duction of mature IL-1 and IL-18, and may therefore provide and outcome at 6-months post-injury. The results of this study
us with useful brain injury biomarkers. In a recent study, CSF suggest that biomarkers may be useful in SCI patients.
was collected from 23 patients who had suffered a moderate or
severe TBI, and levels of inflammasome proteins were measured DISCUSSION
(Adamczak et al., 2012). Apoptosis-associated speck-like protein Brain injury biomarkers promise to improve patient diagnosis,
containing a caspase recruitment domain (ASC), caspase-1, and management and outcomes, and aid in the development of novel
Nacht leucine-rich-repeat protein-1 (NALP-1) were all elevated in therapeutics. The now well accepted inflammatory response that
the CSF of TBI patients versus controls. Furthermore, all 3 pro- occurs in the injured brain has the potential to offer clinicians a
teins correlated significantly with outcome (GOS at 5-months; number of markers that could provide specific information on
Adamczak et al., 2012). the injury. Experiments in animal models of TBI have revealed a
plethora of inflammatory mediators that are expressed in the brain
COMPARABLE STUDIES IN SPINAL CORD INJURY following injury. Many of these exhibit rapid changes in expres-
Development of biomarkers for CNS injury has focused almost sion, reaching peaks of over 1000 orders of magnitude greater
exclusively on brain injury, however, markers for spinal cord injury than physiological levels within hours of injury. The magnitude,
(SCI) are also much needed. MRI after SCI can be used to detect timing, and duration of expression of these mediators might be
hemorrhage, transaction, and lesions, but is not always the best able to provide not only information about the injury but also of

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 10


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

the complexity deriving from the combination of multiple insults. TBI and established associations or correlations with other injury
For example, in a rat Marmarou model of diffuse brain injury, parameters and outcomes. Among the cytokines, perhaps the most
TNF production is of a higher magnitude, and IL-1 expression promising to date is IL-6, since 100-fold increases can be readily
is heightened and prolonged, when the injury is followed by a measured in serum following TBI (Winter et al., 2004; Chiaretti
30-min period of hypoxia (Yan et al., 2011). This suggests that et al., 2005). IL-6 levels have been correlated with ICP (Hergen-
cytokines may indeed reveal various degrees of brain damage with roeder et al., 2010), outcome (Arand et al., 2001; Woiciechowsky
overlapping insults. et al., 2002), but not in multi-trauma patients. To overcome this
Translating information from animal studies into clinically rel- problem sampling for cytokine biomarkers could be done in CSF
evant concepts can prove challenging, since human TBI is a very or in the brain parenchyma itself by microdialysis to get a CNS
varied condition that is often accompanied by additional periph- specific measurement of the immune response. Since ICP moni-
eral injuries, especially if we consider that animal models mostly toring and management is common in TBI patients, CSF samples
reproduce a single form of TBI. Additionally, the human brain is could be obtained from severely injured patients. Changes in
gyroencephalic, and with a larger mass than the rodent brain it CSF cytokine concentrations can be orders of magnitude greater
also presents a different ratio of white to gray matter. The devel- than serum concentrations, and do not appear to be affected by
opment of inflammatory mediators as reliable markers of brain peripheral injuries. However, data is still mixed with regard to the
injury is jeopardized by the fact that peripheral injuries induce ability of cytokines to distinguish injury severity and type, pre-
an immune response that may mask or be indistinguishable from dict or correlate with ICP, or to predict outcome characteristics.
the inflammatory response occurring in the brain. Indeed, several Further study of the cellular origin and biochemical meaning of
studies have noted that in the presence of multiple injuries, mark- raised or lowered level of a specific cytokine may help to interpret
ers of inflammation cannot discriminate for the presence of brain these data.
injury (Hensler et al., 2000; Dziurdzik et al., 2004; Shiozaki et al., Given the problems discussed above, improved sensitivity may
2005; Hergenroeder et al., 2010). To this end, models of so called be achieved by combining a multitude of biomarkers with conven-
poly-trauma are being developed, which combine experimen- tional neuroimaging techniques, and neurological scoring (e.g.,
tal TBI with peripheral injuries (Maegele et al., 2005; Weckbach GCS, GOS/E). Novel cytokine biomarkers could be measured
et al., 2012). These can then be used to search for specific brain in parallel with classical TBI biomarkers such as S100B and
injury markers. Another consideration is that in most animal stud- NSE to improve sensitivity. We have shown that CSF concentra-
ies cytokine concentrations are measured directly in homogenized tions of sICAM-1 correlate well with tissue and BBB damage,
brain tissue rather than in CSF or blood samples. While this gives giving an indication of the degree of immunologic activation
very useful description of the specific response of the brain to in the injured CNS (Pleines et al., 1998), and in a subsequent
injury, such measurements are not possible in the clinic and if done publication measured sICAM-1 together with well known TBI
they would have little relevance when considering the definition of biomarkers (Pleines et al., 2001). In the latter publication we
a biomarker for early diagnostic and prognostic significance. An showed that mean CSF protein concentrations of S100B corre-
additional consideration should be differences in immune acti- late with IL-6, contusion size assessed by CT, and GOS, while
vation occurring in the species utilized in modeling TBI. Higher serum S100B correlates with contusion size and GOS. In this
brain TNF levels have been reported in rats as compared to mice study we also showed that NSE serum levels correlate with IL-6,
in equivalent severity of cerebral ischemia (Schroeter et al., 2003), and that NSE levels in CSF correlate with sICAM-1 and con-
so different responses between rodents and humans must be also tusion size (but not GOS). Taken together, the correlation of
expected. Even within the human population there is evidence serum S100B with contusions size and outcome shows that it
that polymorphisms in cytokine genes could affect not only out- reflects the extent of injury well, but NSE and cytokine bio-
come, but also the subject level of cytokine synthesis in response markers give a better indication of the degree of inflammation
to injury (Hadjigeorgiou et al., 2005; Uzan et al., 2005). Finally, the in the brain. Other groups have gone one to show that by com-
redundancy inherent in the inflammatory response is important bining pairs of biomarkers including IL-6, IL-8, S100B, and NSE,
to consider, since it makes it quite likely that in a genetically diverse a higher degree of outcome predictability can be achieved versus
population of humans a degree of variability will be observed in any single biomarker (Winter et al., 2004; Berger et al., 2009; Lo
cytokine responses. et al., 2009). Similarly, combining GCS score with a single bio-
In studies that have concomitantly measured cytokine levels marker such as IL-8 also improves outcome predictability (Lo
in CSF and serum differences in concentration and even tim- et al., 2010). Recently developed methods for predicting outcomes
ing of peaks has been observed (Kossmann et al., 1995; Csuka based on age, motor score, pupillary reactivity, and CT charac-
et al., 1999; Shiozaki et al., 2005). This undoubtedly reflects the teristics (IMPACT, Steyerberg et al., 2008) have been shown to
compartmentalization of the CNS from the periphery and the benefit from the inclusion of brain injury biomarkers (Czeiter
limited diffusion of cytokines out of the brain parenchyma and et al., 2012). Although inflammatory markers were not included
vice versa. Changes in BBB compliance following TBI are also in that study, several cytokines have been shown to have power to
known to occur, and may affect the CSF:serum ratio of some predict outcome, including IL-1, IL-6, IL-8, and IL-10 (Bell et al.,
cytokines (Kossmann et al., 1995; Csuka et al., 1999), as well as 1997; Whalen et al., 2000; Arand et al., 2001; Mussack et al., 2002;
sequestration of cytokines by the liver (Wu and Pardridge, 1999). Woiciechowsky et al., 2002; Kushi et al., 2003b; Chiaretti et al.,
Nonetheless, many groups have been able to successfully detect sig- 2005; Shiozaki et al., 2005; Gopcevic et al., 2007; Kirchhoff et al.,
nificant increases in inflammatory markers in the blood following 2008).

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 11


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

In addition to being useful in prediction of changes in ICP, mouse brain subjected to CHI when minocycline is administered
mortality, or 6 month outcomes, biomarkers could be used to (Bye et al., 2007). Although interestingly, there was no significant
categorize patients based on specific pathophysiological processes reduction in other cytokines, including TNF, IL-6, G-CSF, MCP-1,
occurring in the injured brain. This information could help to pro- and MIP-2.
vide individualized treatment based on the specific type or severity The future for inflammatory mediators as biomarkers for use
of injury. Using a rat model of diffuse injury we have shown that in TBI is still uncertain, in large part due to their lack of specificity.
an additional hypoxic insult enhances cortical production of the However, development of animal models of multi-trauma, the use
cytokines IL-1, IL-6, and TNF (Yan et al., 2011). In human studies, of two or more markers, and new sampling techniques may over-
correlations have been found between injury severity and concen- come this problem. There is also a need to gain a more complete
trations of a specific cytokine, such as IL-1 (Aly et al., 2006), IL-6 understanding of the temporal expression profile of each cytokine
(Kossmann et al., 1996; Kalabalikis et al., 1999; Woiciechowsky in specific types and severities of injury, since data is currently
et al., 2002; Chiaretti et al., 2005), and IL-10 (Neidhardt et al., limited. The use of multiplex assays now allows for simultaneous
1997). An improved understanding of the roles of these cytokines measurement of several cytokines in brain samples and is provid-
in the secondary injury process may pave the way for targeted treat- ing useful information in this regard (Yan et al., 2011; Dalgard et al.,
ment strategies tailored specifically to the patient. Measurement 2012). In addition, the use of microdialysis technology in patients,
of these cytokines could also be used to track the effects of a poten- while invasive and expensive, has potential to provide us with
tial pharmacological treatment. For example, methylprednisolone continual cytokine concentrations within the brain parenchyma
(MP) has anti-oxidant and anti-inflammatory effects and is used itself (Winter et al., 2002; Helmy et al., 2007). Intraparenchy-
in the treatment of SCI. Administration of MP to rats subjected mal measurement of this kind negates the need to consider BBB
to experimental SCI or subjected to an inflammatory stimulus has disturbances and may be more biologically relevant.
been found to reduce TNF expression (Buttini et al., 1997; Xu et al., In conclusion, the monitoring of the inflammatory process has
1998), and expression of other cytokines (Fu and Saporta, 2005). the potential to provide specific information on the injury and
Another potential treatment is the tetracycline antibiotic, minocy- make predictions about probable outcomes. Several cytokines have
cline, which has been found to have anti-inflammatory effects. shown potential in this area, but a more complete understanding
We have shown that IL-1 expression is significantly reduced in of their specific roles and expression profiles is needed.

REFERENCES IL-1 beta and tumor necrosis factor- blood-brain barrier. Neurosci. Lett. D. (2007). Serum biomarker con-
Adamczak, S., Dale, G., De Rivero Vac- alpha. J. Immunol. 149, 23582366. 179, 5356. centrations and outcome after pedi-
cari, J. P., Bullock, M. R., Dietrich, W. Aly, H., Khashaba, M. T., El-Ayouty, Barksby, H. E., Lea, S. R., Pre- atric traumatic brain injury. J. Neu-
D., and Keane, R. W. (2012). Inflam- M., El-Sayed, O., and Hasanein, B. shaw, P. M., and Taylor, J. J. rotrauma 24, 17931801.
masome proteins in cerebrospinal M. (2006). IL-1beta, IL-6 and TNF- (2007). The expanding family of Berger, R. P., Taasan, S., Rand, A., Lok-
fluid of brain-injured patients as alpha and outcomes of neonatal interleukin-1 cytokines and their shin, A., and Kochanek, P. (2009).
biomarkers of functional outcome: hypoxic ischemic encephalopathy. role in destructive inflammatory dis- Multiplex assessment of serum bio-
clinical article. J. Neurosurg. 117, Brain Dev. 28, 178182. orders. Clin. Exp. Immunol. 149, marker concentrations in well-
11191125. Anderson, R. E., Hansson, L. O., 217225. appearing children with inflicted
Aderka, D., Le, J. M., and Vil- Nilsson, O., Dijlai-Merzoug, R., Basu, A., Krady, J. K., OMalley, M., traumatic brain injury. Pediatr. Res.
cek, J. (1989). IL-6 inhibits and Settergren, G. (2001). High Styren, S. D., Dekosky, S. T., and 65, 97102.
lipopolysaccharide-induced tumor serum S100B levels for trauma Levison, S. W. (2002). The type Bevilacqua, M. P., Pober, J. S., Wheeler,
necrosis factor production in cul- patients without head injuries. Neu- 1 interleukin-1 receptor is essen- M. E., Cotran, R. S., and Gim-
tured human monocytes, U937 rosurgery 48, 12551258; discussion tial for the efficient activation of brone, M. A. Jr. (1985). Interleukin
cells, and in mice. J. Immunol. 143, 12581260. microglia and the induction of mul- 1 acts on cultured human vascular
35173523. Apostolakis, S., Vogiatzi, K., Amanati- tiple proinflammatory mediators in endothelium to increase the adhe-
Agay, D., Andriollo-Sanchez, M., dou, V., and Spandidos, D. A. (2009). response to brain injury. J. Neurosci. sion of polymorphonuclear leuko-
Claeyssen, R., Touvard, L., Denis, Interleukin 8 and cardiovascular dis- 22, 60716082. cytes, monocytes, and related leuko-
J., Roussel, A. M., et al. (2008). ease. Cardiovasc. Res. 84, 353360. Bell, M. J., Kochanek, P. M., Doughty, cyte cell lines. J. Clin. Invest. 76,
Interleukin-6, TNF-alpha and Arand, M., Melzner, H., Kinzl, L., L. A., Carcillo, J. A., Adelson, P. 20032011.
interleukin-1 beta levels in blood Bruckner, U. B., and Gebhard, F. D., Clark, R. S., et al. (1997). Bickel, M. (1993). The role of
and tissue in severely burned (2001). Early inflammatory media- Interleukin-6 and interleukin-10 in interleukin-8 in inflammation
rats. Eur. Cytokine Netw. 19, tor response following isolated trau- cerebrospinal fluid after severe trau- and mechanisms of regulation. J.
17. matic brain injury and other major matic brain injury in children. J. Periodontol. 64, 456460.
Ahn, M. J., Sherwood, E. R., Prough, D. trauma in humans. Langenbecks Neurotrauma 14, 451457. Boutin, H., Kimber, I., Rothwell, N.
S., Lin, C. Y., and Dewitt, D. S. (2004). Arch. Surg. 386, 241248. Benveniste, E. N. (1998). Cytokine J., and Pinteaux, E. (2003). The
The effects of traumatic brain injury Baker, A. J., Rhind, S. G., Morrison, L. J., actions in the central nervous sys- expanding interleukin-1 family and
on cerebral blood flow and brain tis- Black, S., Crnko, N. T., Shek, P. N., et tem. Cytokine Growth Factor Rev. 9, its receptors: do alternative IL-
sue nitric oxide levels and cytokine al. (2009). Resuscitation with hyper- 259275. 1 receptor/signaling pathways exist
expression. J. Neurotrauma 21, tonic saline-dextran reduces serum Benveniste, E. N., Sparacio, S. M., Nor- in the brain? Mol. Neurobiol. 27,
14311442. biomarker levels and correlates with ris, J. G., Grenett, H. E., and Fuller, G. 239248.
Aloisi, F., Care, A., Borsellino, G., outcome in severe traumatic brain M. (1990). Induction and regulation Brisby, H., Olmarker, K., Rosengren,
Gallo, P., Rosa, S., Bassani, A., et injury patients. J. Neurotrauma 26, of interleukin-6 gene expression in L., Cederlund, C. G., and Rydevik,
al. (1992). Production of hemolym- 12271240. rat astrocytes. J. Neuroimmunol. 30, B. (1999). Markers of nerve tissue
phopoietic cytokines (IL-6, IL-8, Banks, W. A., Kastin, A. J., and Gutier- 201212. injury in the cerebrospinal fluid in
colony-stimulating factors) by nor- rez, E. G. (1994). Penetration of Berger, R. P., Beers, S. R., Richichi, patients with lumbar disc herniation
mal human astrocytes in response to interleukin-6 across the murine R., Wiesman, D., and Adelson, P. and sciatica. Spine 24, 742746.

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 12


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

Brough, D., Tyrrell, P. J., and Allan, S. M. following traumatic brain injury. J. Comparison with IL-4 and modu- patients after repair of abdominal
(2011). Regulation of interleukin-1 Head Trauma Rehabil. 19, 205216. lation by IFN-gamma or IL-10. J. aortic aneurysm. Eur. J. Surg. 162,
in acute brain injury. Trends Phar- Coxon, A., Tang, T., and Mayadas, Immunol. 151, 63706381. 287296.
macol. Sci. 32, 617622. T. N. (1999). Cytokine-activated Dinarello, C. A. (1994). The interleukin- Frugier, T., Morganti-Kossmann, M. C.,
Buttini, M., Mir, A., Appel, K., endothelial cells delay neutrophil 1 family: 10 years of discovery. OReilly, D., and McLean, C. A.
Wiederhold, K. H., Limonta, S., apoptosis in vitro and in vivo. FASEB J. 8, 13141325. (2010). In situ detection of inflam-
Gebicke-Haerter, P. J., et al. (1997). A role for granulocyte/macrophage Dinarello, C. A. (1998). Interleukin- matory mediators in post mortem
Lipopolysaccharide induces expres- colony-stimulating factor. J. Exp. 1, interleukin-1 receptors and human brain tissue after traumatic
sion of tumour necrosis factor alpha Med. 190, 923934. interleukin-1 receptor antagonist. injury. J. Neurotrauma 27, 497507.
in rat brain: inhibition by methyl- Csuka, E., Hans, V. H., Ammann, Int. Rev. Immunol. 16, 457499. Fu, E. S., and Saporta, S. (2005). Methyl-
prednisolone and by rolipram. Br. J. E., Trentz, O., Kossmann, T., and Dinarello, C. A. (2009). Immunologi- prednisolone inhibits production of
Pharmacol. 122, 14831489. Morganti-Kossmann, M. C. (2000). cal and inflammatory functions of interleukin-1beta and interleukin-6
Bye, N., Habgood, M. D., Callaway, J. K., Cell activation and inflammatory the interleukin-1 family. Annu. Rev. in the spinal cord following com-
Malakooti, N., Potter, A., Kossmann, response following traumatic axonal Immunol. 27, 519550. pression injury in rats. J. Neurosurg.
T., et al. (2007). Transient neuro- injury in the rat. Neuroreport 11, Drewes, G., Ebneth, A., and Man- Anesthesiol. 17, 8285.
protection by minocycline following 25872590. delkow, E. M. (1998). MAPs, MARKs Gadient, R. A., and Otten, U. (1994).
traumatic brain injury is associated Csuka, E., Morganti-Kossmann, M. C., and microtubule dynamics. Trends Identification of interleukin-6 (IL-
with attenuated microglial activa- Lenzlinger, P. M., Joller, H., Trentz, Biochem. Sci. 23, 307311. 6)-expressing neurons in the cere-
tion but no changes in cell apop- O., and Kossmann, T. (1999). IL- Dziurdzik, P., Krawczyk, L., Jalowiecki, bellum and hippocampus of nor-
tosis or neutrophil infiltration. Exp. 10 levels in cerebrospinal fluid and P., Kondera-Anasz, Z., and Menon, L. mal adult rats. Neurosci. Lett. 182,
Neurol. 204, 220233. serum of patients with severe trau- (2004). Serum interleukin-10 in ICU 243246.
Cao, F., Yang, X. F., Liu, W. G., Hu, W. matic brain injury: relationship to patients with severe acute central Gennarelli, T. A., Champion, H. R.,
W., Li, G., Zheng, X. J., et al. (2008). IL-6, TNF-alpha, TGF-beta1 and nervous system injuries. Inflamm. Copes, W. S., and Sacco, W. J. (1994).
Elevation of neuron-specific enolase blood-brain barrier function. J. Neu- Res. 53, 338343. Comparison of mortality, morbidity,
and S-100beta protein level in exper- roimmunol. 101, 211221. Fan, L., Young, P. R., Barone, F. C., and severity of 59,713 head injured
imental acute spinal cord injury. J. Czeiter, E., Mondello, S., Kovacs, N., Feuerstein, G. Z., Smith, D. H., and patients with 114,447 patients with
Clin. Neurosci. 15, 541544. Sandor, J., Gabrielli, A., Schmid, McIntosh, T. K. (1995). Experimen- extracranial injuries. J. Trauma 37,
Chen, Y., Hallenbeck, J. M., Ruetzler, C., K., et al. (2012). Brain injury bio- tal brain injury induces expression 962968.
Bol, D., Thomas, K., Berman, N. E., et markers may improve the predic- of interleukin-1 beta mRNA in the Goldstein, M. (1990). Traumatic brain
al. (2003). Overexpression of mono- tive power of the IMPACT out- rat brain. Brain Res. Mol. Brain Res. injury: a silent epidemic. Ann. Neu-
cyte chemoattractant protein 1 in come calculator. J. Neurotrauma 29, 30, 125130. rol. 27, 327.
the brain exacerbates ischemic brain 17701778. Fan, L., Young, P. R., Barone, F. C., Goodman, J. C., Robertson, C. S., Gross-
injury and is associated with recruit- Dalgard, C. L., Cole, J. T., Kean, Feuerstein, G. Z., Smith, D. H., and man, R. G., and Narayan, R. K.
ment of inflammatory cells. J. Cereb. W. S., Lucky, J. J., Sukumar, G., McIntosh, T. K. (1996). Experimen- (1990). Elevation of tumor necrosis
Blood Flow Metab. 23, 748755. McMullen, D. C., et al. (2012). tal brain injury induces differential factor in head injury. J. Neuroim-
Chiaretti, A., Antonelli, A., Mastrangelo, The cytokine temporal profile in expression of tumor necrosis factor- munol. 30, 213217.
A., Pezzotti, P., Tortorolo, L., Tosi, F., rat cortex after controlled cortical alpha mRNA in the CNS. Brain Res. Gopcevic, A., Mazul-Sunko, B., Marout,
et al. (2008). Interleukin-6 and nerve impact. Front. Mol. Neurosci. 5:6. Mol. Brain Res. 36, 287291. J., Sekulic,A.,Antoljak, N., Siranovic,
growth factor upregulation corre- doi:10.3389/fnmol.2012.00006 Febbraio, M. A., and Pedersen, B. M., et al. (2007). Plasma interleukin-
lates with improved outcome in chil- DAndrea, A., Aste-Amezaga, M., K. (2005). Contraction-induced 8 as a potential predictor of mortal-
dren with severe traumatic brain Valiante, N. M., Ma, X., Kubin, M., myokine production and release: is ity in adult patients with severe trau-
injury. J. Neurotrauma 25, 225234. and Trinchieri, G. (1993). Inter- skeletal muscle an endocrine organ? matic brain injury. Tohoku J. Exp.
Chiaretti, A., Genovese, O., Aloe, L., leukin 10 (IL-10) inhibits human Exerc. Sport Sci. Rev. 33, 114119. Med. 211, 387393.
Antonelli, A., Piastra, M., Polidori, lymphocyte interferon gamma- Finkelstein, E., Corso, P. S., and Miller, Gruber, M. F., Williams, C. C.,
G., et al. (2005). Interleukin 1beta production by suppressing natural T. R. (2006). The Incidence And Eco- and Gerrard, T. L. (1994).
and interleukin 6 relationship with killer cell stimulatory factor/IL-12 nomic Burden of Injuries in the United Macrophage-colony-stimulating
paediatric head trauma severity and synthesis in accessory cells. J. Exp. States. New York, NY: Oxford Uni- factor expression by anti-CD45
outcome. Childs Nerv. Syst. 21, Med. 178, 10411048. versity Press. stimulated human monocytes is
185193; discussion 194. Dash, P. K., Zhao, J., Hergenroeder, G., Fiorentino, D. F., Zlotnik, A., Mosmann, transcriptionally up-regulated by
Chung, I. Y., and Benveniste, E. N. and Moore, A. N. (2010). Biomark- T. R., Howard, M., and OGarra, A. IL-1 beta and inhibited by IL-4 and
(1990). Tumor necrosis factor-alpha ers for the diagnosis, prognosis, and (1991). IL-10 inhibits cytokine pro- IL-10. J. Immunol. 152, 13541361.
production by astrocytes. Induction evaluation of treatment efficacy for duction by activated macrophages. J. Guez, M., Hildingsson, C., Rosen-
by lipopolysaccharide, IFN-gamma, traumatic brain injury. Neurothera- Immunol. 147, 38153822. gren, L., Karlsson, K., and Toola-
and IL-1 beta. J. Immunol. 144, peutics 7, 100114. Foda, M. A., and Marmarou, A. (1994). nen, G. (2003). Nervous tissue dam-
29993007. de Waal Malefyt, R., Abrams, J., Ben- A new model of diffuse brain age markers in cerebrospinal fluid
Ciallella, J. R., Ikonomovic, M. D., nett, B., Figdor, C. G., and De injury in rats. Part II: morphologi- after cervical spine injuries and
Paljug, W. R., Wilbur, Y. I., Dixon, Vries, J. E. (1991). Interleukin 10(IL- cal characterization. J. Neurosurg. 80, whiplash trauma. J. Neurotrauma
C. E., Kochanek, P. M., et al. (2002). 10) inhibits cytokine synthesis by 301313. 20, 853858.
Changes in expression of amyloid human monocytes: an autoreg- Franzen, R., Bouhy, D., and Schoe- Hadjigeorgiou, G. M., Paterakis, K.,
precursor protein and interleukin- ulatory role of IL-10 produced nen, J. (2004). Nervous system Dardiotis, E., Dardioti, M., Agge-
1beta after experimental traumatic by monocytes. J. Exp. Med. 174, injury: focus on the inflammatory lakis, K., Tasiou, A., et al. (2005).
brain injury in rats. J. Neurotrauma 12091220. cytokine granulocyte-macrophage IL-1RN and IL-1B gene polymor-
19, 15551567. de Waal Malefyt, R., Figdor, C. G., colony stimulating factor. Neurosci. phisms and cerebral hemorrhagic
Coburn, K. (1992). Traumatic brain Huijbens, R., Mohan-Peterson, S., Lett. 361, 7678. events after traumatic brain injury.
injury: the silent epidemic. AACN Bennett, B., Culpepper, J., et al. Froon, A. H., Greve, J. W., Van Der Neurology 65, 10771082.
Clin. Issues Crit. Care Nurs. 3, 918. (1993). Effects of IL-13 on pheno- Linden, C. J., and Buurman, W. A. Hang, C. H., Shi, J. X., Tian, J., Li, J. S.,
Corrigan, J. D., Whiteneck, G., and type, cytokine production, and cyto- (1996). Increased concentrations of Wu, W., and Yin, H. X. (2004). Effect
Mellick, D. (2004). Perceived needs toxic function of human monocytes. cytokines and adhesion molecules in of systemic LPS injection on cortical

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 13


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

NF-kappaB activity and inflamma- Holmin, S., and Mathiesen, T. (2000). interleukin-10 in the rat. J. Trauma Kossmann, T., Hans, V. H., Imhof, H.
tory response following traumatic Intracerebral administration of 60, 152157. G., Stocker, R., Grob, P., Trentz, O.,
brain injury in rats. Brain Res. 1026, interleukin-1beta and induction Kasahara, T., Mukaida, N., Yamashita, et al. (1995). Intrathecal and serum
2332. of inflammation, apoptosis, and K., Yagisawa, H., Akahoshi, T., and interleukin-6 and the acute-phase
Hans, V. H., Kossmann, T., Len- vasogenic edema. J. Neurosurg. 92, Matsushima, K. (1991). IL-1 and response in patients with severe
zlinger, P. M., Probstmeier, R., Imhof, 108120. TNF-alpha induction of IL-8 and traumatic brain injuries. Shock 4,
H. G., Trentz, O., et al. (1999). Holmin, S., Schalling, M., Hojeberg, B., monocyte chemotactic and activat- 311317.
Experimental axonal injury triggers Nordqvist, A. C., Skeftruna, A. K., ing factor (MCAF) mRNA expres- Kossmann, T., Stahel, P. F., Lenzlinger,
interleukin-6 mRNA, protein syn- and Mathiesen, T. (1997). Delayed sion in a human astrocytoma cell P. M., Redl, H., Dubs, R. W.,
thesis and release into cerebrospinal cytokine expression in rat brain fol- line. Immunology 74, 6067. Trentz, O., et al. (1997). Interleukin-
fluid. J. Cereb. Blood Flow Metab. 19, lowing experimental contusion. J. Kim, K. S., Wass, C. A., Cross, A. S., 8 released into the cerebrospinal
184194. Neurosurg. 86, 493504. and Opal, S. M. (1992). Modula- fluid after brain injury is associated
Hayakata, T., Shiozaki, T., Tasaki, O., Huang, D. R., Wang, J., Kivisakk, P., tion of blood-brain barrier perme- with blood-brain barrier dysfunc-
Ikegawa, H., Inoue, Y., Toshiyuki, Rollins, B. J., and Ransohoff, R. ability by tumor necrosis factor and tion and nerve growth factor pro-
F., et al. (2004). Changes in CSF M. (2001). Absence of monocyte antibody to tumor necrosis factor in duction. J. Cereb. Blood Flow Metab.
S100B and cytokine concentra- chemoattractant protein 1 in mice the rat. Lymphokine Cytokine Res. 11, 17, 280289.
tions in early-phase severe traumatic leads to decreased local macrophage 293298. Kovesdi, E., Luckl, J., Bukovics, P.,
brain injury. Shock 22, 102107. recruitment and antigen-specific Kinoshita, K., Chatzipanteli, K.,Vitarbo, Farkas, O., Pal, J., Czeiter, E., et
Helmy, A., Carpenter, K. L., and T helper cell type 1 immune E., Truettner, J. S., Alonso, O. F., and al. (2010). Update on protein bio-
Hutchinson, P. J. (2007). Microdialy- response in experimental autoim- Dietrich, W. D. (2002). Interleukin- markers in traumatic brain injury
sis in the human brain and its poten- mune encephalomyelitis. J. Exp. 1beta messenger ribonucleic acid with emphasis on clinical use in
tial role in the development and clin- Med. 193, 713726. and protein levels after fluid- adults and pediatrics. Acta Neu-
ical assessment of drugs. Curr. Med. Hughes, P. M.,Allegrini, P. R., Rudin, M., percussion brain injury in rats: rochir. (Wien) 152, 117.
Chem. 14, 15251537. Perry, V. H., Mir, A. K., and Wiess- importance of injury severity and Krueger, J. M. (2008). The role of
Helmy, A., Carpenter, K. L., Menon, D. ner, C. (2002). Monocyte chemoat- brain temperature. Neurosurgery 51, cytokines in sleep regulation. Curr.
K., Pickard, J. D., and Hutchinson, tractant protein-1 deficiency is pro- 195203; discussion 203. Pharm. Des. 14, 34083416.
P. J. (2011). The cytokine response tective in a murine stroke model. Kirchhoff, C., Buhmann, S., Bogner, V., Kunihara, T., Shiiya, N., and Yasuda, K.
to human traumatic brain injury: J. Cereb. Blood Flow Metab. 22, Stegmaier, J., Leidel, B. A., Braun- (2001). Changes in S100beta pro-
temporal profiles and evidence for 308317. stein, V., et al. (2008). Cerebrospinal tein levels in cerebrospinal fluid
cerebral parenchymal production. Hukkelhoven, C. W., Steyerberg, E. W., IL-10 concentration is elevated in after thoracoabdominal aortic oper-
J. Cereb. Blood Flow Metab. 31, Habbema, J. D., Farace, E., Mar- non-survivors as compared to sur- ations. J. Thorac. Cardiovasc. Surg.
658670. marou, A., Murray, G. D., et al. vivors after severe traumatic brain 122, 10191020.
Hensler, T., Sauerland, S., Riess, P., Hess, (2005). Predicting outcome after injury. Eur. J. Med. Res. 13, 464468. Kushi, H., Saito, T., Makino, K., and
S., Helling, H. J., Andermahr, J., traumatic brain injury: development Kitagawa, K., Matsumoto, M., Niinobe, Hayashi, N. (2003a). IL-8 is a
et al. (2000). The effect of addi- and validation of a prognostic score M., Mikoshiba, K., Hata, R., Ueda, key mediator of neuroinflammation
tional brain injury on systemic inter- based on admission characteristics. H., et al. (1989). Microtubule- after traumatic brain injury. Crit.
leukin (IL)-10 and IL-13 levels in J. Neurotrauma 22, 10251039. associated protein 2 as a sensitive Care Med. 31, A82A82.
trauma patients. Inflamm. Res. 49, Hukkelhoven, C. W., Steyerberg, E. W., marker for cerebral ischemic dam- Kushi, H., Saito, T., Makino, K., and
524528. Rampen, A. J., Farace, E., Habbema, age immunohistochemical investi- Hayashi, N. (2003b). L-8 is a key
Hergenroeder, G. W., Moore, A. N., J. D., Marshall, L. F., et al. (2003). gation of dendritic damage. Neuro- mediator of neuroinflammation in
McCoy, J. P. Jr., Samsel, L., Ward, N. Patient age and outcome following science 31, 401411. severe traumatic brain injuries. Acta
H. III, Clifton, G. L., et al. (2010). severe traumatic brain injury: an Knies, U. E., Behrensdorf, H. A., Neurochir. Suppl. 86, 347350.
Serum IL-6: a candidate biomarker analysis of 5600 patients. J. Neuro- Mitchell, C. A., Deutsch, U., Risau, Kwon, B. K., Stammers, A. M., Belanger,
for intracranial pressure elevation surg. 99, 666673. W., Drexler, H. C., et al. (1998). Reg- L. M., Bernardo, A., Chan, D.,
following isolated traumatic brain Hutchinson, P. J., OConnell, M. T., ulation of endothelial monocyte- Bishop, C. M., et al. (2010).
injury. J. Neuroinflammation 7, 19. Rothwell, N. J., Hopkins, S. J., activating polypeptide II release by Cerebrospinal fluid inflammatory
Herrmann, M., Curio, N., Jost, S., Gru- Nortje, J., Carpenter, K. L., et al. apoptosis. Proc. Natl. Acad. Sci. cytokines and biomarkers of injury
bich, C., Ebert, A. D., Fork, M. L., (2007). Inflammation in human U.S.A. 95, 1232212327. severity in acute human spinal cord
et al. (2001). Release of biochemi- brain injury: intracerebral concen- Knoblach, S. M., and Faden, A. injury. J. Neurotrauma 27, 669682.
cal markers of damage to neuronal trations of IL-1alpha, IL-1beta, and I. (1998). Interleukin-10 improves Lammertse, D., Dungan, D., Dreisbach,
and glial brain tissue is associated their endogenous inhibitor IL-1ra. J. outcome and alters proinflamma- J., Falci, S., Flanders, A., Marino,
with short and long term neuropsy- Neurotrauma 24, 15451557. tory cytokine expression after exper- R., et al. (2007). Neuroimaging in
chological outcome after traumatic Islam, O., Gong, X., Rose-John, S., and imental traumatic brain injury. Exp. traumatic spinal cord injury: an
brain injury. J. Neurol. Neurosurg. Heese, K. (2009). Interleukin-6 and Neurol. 153, 143151. evidence-based review for clinical
Psychiatr. 70, 95100. neural stem cells: more than gliogen- Knoblach, S. M., Fan, L., and Faden, practice and research. J. Spinal Cord
Hillman, J., Aneman, O., Persson, esis. Mol. Biol. Cell 20, 188199. A. I. (1999). Early neuronal expres- Med. 30, 205214.
M., Andersson, C., Dabrosin, C., Kalabalikis, P., Papazoglou, K., Gou- sion of tumor necrosis factor-alpha Langlois, J. A., Rutland-Brown, W., and
and Mellergard, P. (2007). Vari- riotis, D., Papadopoulos, N., Kar- after experimental brain injury con- Wald, M. M. (2006). The epidemiol-
ations in the response of inter- dara, M., Papageorgiou, F., et al. tributes to neurological impairment. ogy and impact of traumatic brain
leukins in neurosurgical inten- (1999). Correlation between serum J. Neuroimmunol. 95, 115125. injury: a brief overview. J. Head
sive care patients monitored using IL-6 and CRP levels and severity Kossmann, T., Hans, V., Imhof, H. G., Trauma Rehabil. 21, 375378.
intracerebral microdialysis. J. Neu- of head injury in children. Intensive Trentz, O., and Morganti-Kossmann, Lau, L. T., and Yu, A. C. (2001).
rosurg. 106, 820825. Care Med. 25, 288292. M. C. (1996). Interleukin-6 released Astrocytes produce and release
Hirao, Y., Kanda, T., Aso, Y., Mit- Kamm, K., Vanderkolk, W., Lawrence, in human cerebrospinal fluid fol- interleukin-1, interleukin-6, tumor
suhashi, M., and Kobayashi, I. C., Jonker, M., and Davis, A. T. lowing traumatic brain injury may necrosis factor alpha and interferon-
(2000). Interleukin-8 an early (2006). The effect of traumatic brain trigger nerve growth factor produc- gamma following traumatic and
marker for bacterial infection. Lab. injury upon the concentration and tion in astrocytes. Brain Res. 713, metabolic injury. J. Neurotrauma 18,
Med. 31, 3944. expression of interleukin-1beta and 143152. 351359.

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 14


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

Lee, H. F., Lee, T. S., and Kou, Y. R. Ma, J., Novikov, L. N., Karlsson, K., et al. (1998). Sympathetic neurons Mushkudiani, N. A., Hukkelhoven, C.
(2012). Anti-inflammatory and neu- Kellerth, J. O., and Wiberg, M. can produce and respond to inter- W., Hernandez, A. V., Murray, G.
roprotective effects of triptolide on (2001). Plexus avulsion and spinal leukin 6. Proc. Natl. Acad. Sci. U.S.A. D., Choi, S. C., Maas, A. I., et al.
traumatic brain injury in rats. Respir. cord injury increase the serum con- 95, 32513256. (2008). A systematic review finds
Physiol. Neurobiol. 182, 18. centration of S-100 protein: an McClain, C. J., Cohen, D., Ott, L., methodological improvements nec-
Lenzlinger, P. M., Morganti-Kossmann, experimental study in rats. Scand. J. Dinarello, C. A., and Young, essary for prognostic models in
M. C., Laurer, H. L., and McIn- Plast. Reconstr. Surg. Hand. Surg. 35, B. (1987). Ventricular fluid determining traumatic brain injury
tosh, T. K. (2001). The duality of the 355359. interleukin-1 activity in patients outcomes. J. Clin. Epidemiol. 61,
inflammatory response to traumatic Maas, A. I., Hukkelhoven, C. W., with head injury. J. Lab. Clin. Med. 331343.
brain injury. Mol. Neurobiol. 24, Marshall, L. F., and Steyerberg, 110, 4854. Mussack, T., Biberthaler, P., Kanz,
169181. E. W. (2005). Prediction of out- Meixensberger, J., and Roosen, K. K. G., Wiedemann, E., Gippner-
Lo, T. Y., Jones, P. A., and Minns, R. A. come in traumatic brain injury (1998). Clinical and pathophys- Steppert, C., Mutschler, W., et
(2009). Pediatric brain trauma out- with computed tomographic char- iological significance of severe al. (2002). Serum S-100B and
come prediction using paired serum acteristics: a comparison between neurotrauma in polytraumatized interleukin-8 as predictive mark-
levels of inflammatory mediators the computed tomographic classi- patients. Langenbecks Arch. Surg. ers for comparative neurologic out-
and brain-specific proteins. J. Neu- fication and combinations of com- 383, 214219. come analysis of patients after car-
rotrauma 26, 14791487. puted tomographic predictors. Neu- Missler, U., Wiesmann, M., Wittmann, diac arrest and severe traumatic
Lo, T. Y., Jones, P. A., and Minns, R. rosurgery 57, 11731182; discussion G., Magerkurth, O., and Hagen- brain injury. Crit. Care Med. 30,
A. (2010). Combining coma score 11731182. strom, H. (1999). Measurement of 26692674.
and serum biomarker levels to pre- Maegele, M., Riess, P., Sauerland, S., glial fibrillary acidic protein in Nagy, G., Dzsinich, C., Selmeci, L., Sepa,
dict unfavorable outcome following Bouillon, B., Hess, S., McIntosh, T. human blood: analytical method G., Dzsinich, M., Kekesi, V., et al.
childhood brain trauma. J. Neuro- K., et al. (2005). Characterization and preliminary clinical results. Clin. (2002). Biochemical alterations in
trauma 27, 21392145. of a new rat model of experimental Chem. 45, 138141. cerebrospinal fluid during thora-
Loscher, C. E., Mills, K. H., and Lynch, combined neurotrauma. Shock 23, Molina-Holgado, E., Ortiz, S., Molina- coabdominal aortic cross-clamping
M. A. (2003). Interleukin-1 recep- 476481. Holgado, F., and Guaza, C. (2000). in dogs. Ann. Vasc. Surg. 16,
tor antagonist exerts agonist activity Maegele, M., Sauerland, S., Bouil- Induction of COX-2 and PGE(2) 436441.
in the hippocampus independent of lon, B., Schafer, U., Trubel, H., biosynthesis by IL-1beta is mediated Narayan, R. K., Kishore, P. R., Becker, D.
the interleukin-1 type I receptor. J. Riess, P., et al. (2007). Differential by PKC and mitogen-activated pro- P., Ward, J. D., Enas, G. G., Green-
Neuroimmunol. 137, 117124. immunoresponses following experi- tein kinases in murine astrocytes. Br. berg, R. P., et al. (1982). Intracranial
Loy, D. N., Sroufe, A. E., Pelt, J. mental traumatic brain injury, bone J. Pharmacol. 131, 152159. pressure: to monitor or not to moni-
L., Burke, D. A., Cao, Q. L., Tal- fracture and two-hit-combined Mondello, S., Gabrielli, A., Catani, S., tor? A review of our experience with
bott, J. F., et al. (2005). Serum neurotrauma. Inflamm. Res. 56, DIppolito, M., Jeromin, A., Cia- severe head injury. J. Neurosurg. 56,
biomarkers for experimental acute 318323. ramella, A., et al. (2012). Increased 650659.
spinal cord injury: rapid eleva- Maier, B., Laurer, H. L., Rose, S., Buur- levels of serum MAP-2 at 6-months Neidhardt, R., Keel, M., Steckholzer,
tion of neuron-specific enolase man, W. A., and Marzi, I. (2005). correlate with improved outcome in U., Safret, A., Ungethuem, U.,
and S-100beta. Neurosurgery 56, Physiological levels of pro- and anti- survivors of severe traumatic brain Trentz, O., et al. (1997). Relation-
391397; discussion 391397. inflammatory mediators in cere- injury. Brain Inj. 26, 16291635. ship of interleukin-10 plasma levels
Lu, B., Rutledge, B. J., Gu, L., Fiorillo, brospinal fluid and plasma: a nor- Mondello, S., Robicsek, S. A., Gabrielli, to severity of injury and clinical out-
J., Lukacs, N. W., Kunkel, S. L., et al. mative study. J. Neurotrauma 22, A., Brophy, G. M., Papa, L., Tepas, come in injured patients. J. Trauma
(1998). Abnormalities in monocyte 822835. J., et al. (2010). AlphaII-spectrin 42, 863870; discussion 870861.
recruitment and cytokine expression Maier, B., Schwerdtfeger, K., Mautes, A., breakdown products (SBDPs): diag- Nitta, T., Allegretta, M., Okumura, K.,
in monocyte chemoattractant pro- Holanda, M., Muller, M., Steudel, W. nosis and outcome in severe trau- Sato, K., and Steinman, L. (1992).
tein 1-deficient mice. J. Exp. Med. I., et al. (2001). Differential release matic brain injury patients. J. Neu- Neoplastic and reactive human
187, 601608. of interleukines 6, 8, and 10 in rotrauma 27, 12031213. astrocytes express interleukin-8
Lu, K. T., Wang, Y. W., Wo, Y. Y., cerebrospinal fluid and plasma after Morganti-Kossman, M. C., Lenzlinger, gene. Neurosurg. Rev. 15, 203207.
and Yang, Y. L. (2005a). Extracellu- traumatic brain injury. Shock 15, P. M., Hans, V., Stahel, P., Csuka, Nybo, L., Nielsen, B., Pedersen, B.
lar signal-regulated kinase-mediated 421426. E., Ammann, E., et al. (1997). Pro- K., Moller, K., and Secher, N. H.
IL-1-induced cortical neuron dam- Marquardt, G., Setzer, M., and Seifert, duction of cytokines following brain (2002). Interleukin-6 release from
age during traumatic brain injury. V. (2004a). Protein S-100b as serum injury: beneficial and deleterious for the human brain during prolonged
Neurosci. Lett. 386, 4045. marker for prediction of functional the damaged tissue. Mol. Psychiatry exercise. J. Physiol. (Lond.) 542,
Lu, K. T., Wang, Y. W., Yang, J. T., outcome in metastatic spinal cord 2, 133136. 991995.
Yang, Y. L., and Chen, H. I. (2005b). compression. Acta Neurochir. (Wien) Morganti-Kossmann, M. C., Kossmann, Nylen, K., Ost, M., Csajbok, L. Z., Nils-
Effect of interleukin-1 on traumatic 146, 449452. T., and Wahl, S. M. (1992). Cytokines son, I., Blennow, K., Nellgard, B., et
brain injury-induced damage to hip- Marquardt, G., Setzer, M., and Seifert, and neuropathology. Trends Phar- al. (2006). Increased serum-GFAP
pocampal neurons. J. Neurotrauma V. (2004b). Protein S-100b for indi- macol. Sci. 13, 286291. in patients with severe traumatic
22, 885895. vidual prediction of functional out- Morganti-Kossmann, M. C., Lenzlinger, brain injury is related to outcome.
Lu, K. T., Wu, C. Y., Yen, H. H., Peng, come in spinal epidural empyema. P. M., Hans, V., Stahel, P., Csuka, J. Neurol. Sci. 240, 8591.
J. H., Wang, C. L., and Yang, Y. L. Spine 29, 5962. E., Ammann, E., et al. (1997). Pro- OConnor, J. J., and Coogan, A.
(2007). Bumetanide administration Marshall, L. F., Marshall, S. B., duction of cytokines following brain N. (1999). Actions of the pro-
attenuated traumatic brain injury Klauber, M. R., Van Berkum Clark, injury: beneficial and deleterious for inflammatory cytokine IL-1 beta on
through IL-1 overexpression. Neu- M., Eisenberg, H., Jane, J. A., the damaged tissue. Mol. Psychiatry central synaptic transmission. Exp.
rol. Res. 29, 404409. et al. (1992). The diagnosis of 2, 133136. Physiol. 84, 601614.
Luheshi, N. M., Rothwell, N. J., and head injury requires a classification Morganti-Kossmann, M. C., Rancan, Otto, V. I., Gloor, S. M., Frentzel, S.,
Brough, D. (2009). Dual functional- based on computed axial tomog- M., Stahel, P. F., and Kossmann, Gilli, U., Ammann, E., Hein, A. E.,
ity of interleukin-1 family cytokines: raphy. J. Neurotrauma 9(Suppl. 1), T. (2002). Inflammatory response et al. (2002). The production of
implications for anti-interleukin- S287S292. in acute traumatic brain injury: a macrophage inflammatory protein-
1 therapy. Br. J. Pharmacol. 157, Marz, P., Cheng, J. G., Gadient, R. A., double-edged sword. Curr. Opin. 2 induced by soluble intercellu-
13181329. Patterson, P. H., Stoyan, T., Otten, U., Crit. Care 8, 101105. lar adhesion molecule-1 in mouse

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 15


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

astrocytes is mediated by src tyro- Perez-Barcena, J., Ibanez, J., Brell, M., neuronal damage in the rat. Brain review of the feasibility of glial fibril-
sine kinases and p42/44 mitogen- Crespi, C., Frontera, G., Llompart- Res. Bull. 29, 243246. lary acidic protein as a biomarker for
activated protein kinase. J. Neu- Pou, J. A., et al. (2011). Lack Ringheim, G. E., Burgher, K. L., and stroke and traumatic brain injury.
rochem. 80, 824834. of correlation among intracerebral Heroux, J. A. (1995). Interleukin-6 Mol. Diagn. Ther. 16, 7992.
Otto, V. I., Stahel, P. F., Rancan, cytokines, intracranial pressure, and mRNA expression by cortical neu- Schmidt, O. I., Heyde, C. E., Ertel,
M., Kariya, K., Shohami, E., Yat- brain tissue oxygenation in patients rons in culture: evidence for neu- W., and Stahel, P. F. (2005). Closed
siv, I., et al. (2001). Regulation of with traumatic brain injury and dif- ronal sources of interleukin-6 pro- head injury an inflammatory dis-
chemokines and chemokine recep- fuse lesions. Crit. Care Med. 39, duction in the brain. J. Neuroim- ease? Brain Res. Brain Res. Rev. 48,
tors after experimental closed head 533540. munol. 63, 113123. 388399.
injury. Neuroreport 12, 20592064. Piazza, O., Storti, M. P., Cotena, S., Riva-Depaty, I., Fardeau, C., Mari- Schobitz, B., De Kloet, E. R., Sutanto,
Pagulayan, K. F., Temkin, N. R., Stoppa, F., Perrotta, D., Esposito, G., ani, J., Bouchaud, C., and Delhaye- W., and Holsboer, F. (1993). Cel-
Machamer, J., and Dikmen, S. S. et al. (2007). S100B is not a reliable Bouchaud, N. (1994). Contribu- lular localization of interleukin 6
(2006). A longitudinal study of prognostic index in paediatric TBI. tion of peripheral macrophages and mRNA and interleukin 6 receptor
health-related quality of life after Pediatr. Neurosurg. 43, 258264. microglia to the cellular reaction mRNA in rat brain. Eur. J. Neurosci.
traumatic brain injury. Arch. Phys. Pinteaux, E., Parker, L. C., Rothwell, after mechanical or neurotoxin- 5, 14261435.
Med. Rehabil. 87, 611618. N. J., and Luheshi, G. N. (2002). induced lesions of the rat brain. Exp. Schroeter, M., Kury, P., and Jander, S.
Palfreyman, J. W., Thomas, D. G., and Expression of interleukin-1 recep- Neurol. 128, 7787. (2003). Inflammatory gene expres-
Ratcliffe, J. G. (1978). Radioim- tors and their role in interleukin-1 Romano, M., Sironi, M., Toniatti, C., sion in focal cortical brain ischemia:
munoassay of human myelin basic actions in murine microglial cells. J. Polentarutti, N., Fruscella, P., Ghezzi, differences between rats and mice.
protein in tissue extract, cere- Neurochem. 83, 754763. P., et al. (1997). Role of IL-6 and Brain Res. Mol. Brain Res. 117, 17.
brospinal fluid and serum and its Pleines, U. E., Morganti-Kossmann, M. its soluble receptor in induction of Sebire, G., Emilie, D., Wallon, C., Hery,
clinical application to patients with C., Rancan, M., Joller, H., Trentz, chemokines and leukocyte recruit- C., Devergne, O., Delfraissy, J. F., et
head injury. Clin. Chim. Acta 82, O., and Kossmann, T. (2001). S- ment. Immunity 6, 315325. al. (1993). In vitro production of
259270. 100 beta reflects the extent of injury Ross, S. A., Halliday, M. I., Campbell, IL-6, IL-1 beta, and tumor necro-
Papa, L., Lewis, L. M., Falk, J. L., Zhang, and outcome, whereas neuronal spe- G. C., Byrnes, D. P., and Rowlands, sis factor-alpha by human embry-
Z., Silvestri, S., Giordano, P., et al. cific enolase is a better indicator B. J. (1994). The presence of tumour onic microglial and neural cells. J.
(2012). Elevated levels of serum glial of neuroinflammation in patients necrosis factor in CSF and plasma Immunol. 150, 15171523.
fibrillary acidic protein breakdown with severe traumatic brain injury. after severe head injury. Br. J. Neuro- Semple, B. D., Bye, N., Rancan,
products in mild and moderate trau- J. Neurotrauma 18, 491498. surg. 8, 419425. M., Ziebell, J. M., and Morganti-
matic brain injury are associated Pleines, U. E., Stover, J. F., Kossmann, T., Rothwell, N. (2003). Interleukin-1 and Kossmann, M. C. (2010a). Role of
with intracranial lesions and neu- Trentz, O., and Morganti-Kossmann, neuronal injury: mechanisms, mod- CCL2 (MCP-1) in traumatic brain
rosurgical intervention. Ann. Emerg. M. C. (1998). Soluble ICAM-1 in ification, and therapeutic poten- injury (TBI): evidence from severe
Med. 59, 471483. CSF coincides with the extent of tial. Brain Behav. Immun. 17, TBI patients and CCL2-/- mice.
Park, D., Joo, S. S., Lee, H. J., Choi, cerebral damage in patients with 152157. J. Cereb. Blood Flow Metab. 30,
K. C., Kim, S. U., and Kim, Y. B. severe traumatic brain injury. J. Neu- Rutland-Brown, W., Langlois, J. A., 769782.
(2012). Microtubule-associated pro- rotrauma 15, 399409. Thomas, K. E., and Xi, Y. L. (2006). Semple, B. D., Bye, N., Ziebell, J.
tein 2, an early blood marker of Posmantur, R. M., Kampfl, A., Taft, W. Incidence of traumatic brain injury M., and Morganti-Kossmann, M.
ischemic brain injury. J. Neurosci. C., Bhattacharjee, M., Dixon, C. E., in the United States, 2003. J. Head C. (2010b). Deficiency of the
Res. 90, 461467. Bao, J., et al. (1996). Diminished Trauma Rehabil. 21, 544548. chemokine receptor CXCR2 attenu-
Pelinka, L. E., Kroepfl, A., Leixner- microtubule-associated protein 2 Sallmann, S., Juttler, E., Prinz, S., ates neutrophil infiltration and cor-
ing, M., Buchinger, W., Raabe, A., (MAP2) immunoreactivity follow- Petersen, N., Knopf, U., Weiser, T., et tical damage following closed head
and Redl, H. (2004a). GFAP versus ing cortical impact brain injury. J. al. (2000). Induction of interleukin- injury. Neurobiol. Dis. 40, 394403.
S100B in serum after traumatic brain Neurotrauma 13, 125137. 6 by depolarization of neurons. J. Semple, B. D., Kossmann, T., and
injury: relationship to brain dam- Quagliarello, V. J., Wispelwey, B., Long, Neurosci. 20, 86378642. Morganti-Kossmann, M. C. (2010c).
age and outcome. J. Neurotrauma 21, W. J. Jr., and Scheld, W. M. (1991). Salmond, C. H., Menon, D. K., Chat- Role of chemokines in CNS health
15531561. Recombinant human interleukin-1 field, D. A., Williams, G. B., Pena, and pathology: a focus on the
Pelinka, L. E., Kroepfl, A., Schmid- induces meningitis and blood-brain A., Sahakian, B. J., et al. (2006). CCL2/CCR2 and CXCL8/CXCR2
hammer, R., Krenn, M., Buchinger, barrier injury in the rat. Characteri- Diffusion tensor imaging in chronic networks. J. Cereb. Blood Flow
W., Redl, H., et al. (2004b). Glial zation and comparison with tumor head injury survivors: correlations Metab. 30, 459473.
fibrillary acidic protein in serum necrosis factor. J. Clin. Invest. 87, with learning and memory indices. Shimonkevitz, R., Bar-Or, D., Harris,
after traumatic brain injury and 13601366. Neuroimage 29, 117124. L., Dole, K., McLaughlin, L., and
multiple trauma. J. Trauma 57, Rainey, T., Lesko, M., Sacho, R., Lecky, Savola, O., Pyhtinen, J., Leino, T. K., Yukl, R. (1999). Transient monocyte
10061012. F., and Childs, C. (2009). Predict- Siitonen, S., Niemela, O., and Hill- release of interleukin-10 in response
Penkowa, M., Camats, J., Hadberg, ing outcome after severe traumatic bom, M. (2004). Effects of head and to traumatic brain injury. Shock 12,
H., Quintana, A., Rojas, S., Giralt, brain injury using the serum S100B extracranial injuries on serum pro- 1016.
M., et al. (2003). Astrocyte-targeted biomarker: results using a single tein S100B levels in trauma patients. Shiozaki, T., Hayakata, T., Tasaki,
expression of interleukin-6 protects (24h) time-point. Resuscitation 80, J. Trauma 56, 12291234; discussion O., Hosotubo, H., Fuijita, K.,
the central nervous system during 341345. 1234. Mouri, T., et al. (2005). Cere-
neuroglial degeneration induced by Ramilo, O., Saez-Llorens, X., Mertsola, Scherbel, U., Raghupathi, R., Nakamura, brospinal fluid concentrations of
6-aminonicotinamide. J. Neurosci. J., Jafari, H., Olsen, K. D., Hansen, M., Saatman, K. E., Trojanowski, J. anti-inflammatory mediators in
Res. 73, 481496. E. J., et al. (1990). Tumor necro- Q., Neugebauer, E., et al. (1999). Dif- early-phase severe traumatic brain
Penkowa, M., Giralt, M., Carrasco, sis factor alpha/cachectin and inter- ferential acute and chronic responses injury. Shock 23, 406410.
J., Hadberg, H., and Hidalgo, leukin 1 beta initiate meningeal of tumor necrosis factor-deficient Shohami, E., Bass, R., Wallach, D.,
J. (2000). Impaired inflamma- inflammation. J. Exp. Med. 172, mice to experimental brain injury. Yamin, A., and Gallily, R. (1996).
tory response and increased oxida- 497507. Proc. Natl. Acad. Sci. U.S.A. 96, Inhibition of tumor necrosis fac-
tive stress and neurodegeneration Relton, J. K., and Rothwell, N. J. (1992). 87218726. tor alpha (TNFalpha) activity in rat
after brain injury in interleukin-6- Interleukin-1 receptor antagonist Schiff, L., Hadker, N., Weiser, S., and brain is associated with cerebro-
deficient mice. Glia 32, 271285. inhibits ischaemic and excitotoxic Rausch, C. (2012). A literature protection after closed head injury.

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 16


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

J. Cereb. Blood Flow Metab. 16, of serum and cerebrospinal fluid closed head injury in mice with cen- thoracoabdominal aortic aneurysm
378384. biomarkers with intracranial hyper- tral overexpression of the secreted surgery: is S-100 protein a biochemi-
Shohami, E., Gallily, R., Mechoulam, R., tension and cerebral hypoperfusion isoform of the interleukin-1 recep- cal marker of clinical value in detect-
Bass, R., and Ben-Hur, T. (1997). after severe traumatic brain injury. J. tor antagonist. J. Neurotrauma 19, ing spinal cord ischemia? J. Vasc.
Cytokine production in the brain Trauma 70, 10961103. 939951. Surg. 27, 344346.
following closed head injury: dex- Steyerberg, E. W., Mushkudiani, N., Thomas, D. G., Palfreyman, J. W., van Dongen, E. P., Ter Beek, H. T.,
anabinol (HU-211) is a novel TNF- Perel, P., Butcher, I., Lu, J., McHugh, and Ratcliffe, J. G. (1978). Serum- Schepens, M. A., Morshuis, W. J.,
alpha inhibitor and an effective neu- G. S., et al. (2008). Predicting out- myelin-basic-protein assay in diag- Haas, F. J., De Boer, A., et al. (1999).
roprotectant. J. Neuroimmunol. 72, come after traumatic brain injury: nosis and prognosis of patients with The relationship between evoked
169177. development and international head injury. Lancet 1, 113115. potentials and measurements of S-
Shohami, E., Ginis, I., and Hallen- validation of prognostic scores Threlkeld, S. W., Lynch, J. L., Lynch, 100 protein in cerebrospinal fluid
beck, J. M. (1999). Dual role of based on admission characteristics. K. M., Sadowska, G. B., Banks, W. during and after thoracoabdomi-
tumor necrosis factor alpha in brain PLoS Med. 5:e165; discussion e165. A., and Stonestreet, B. S. (2010). nal aortic aneurysm surgery. J. Vasc.
injury. Cytokine Growth Factor Rev. doi:10.1371/journal.pmed.0050165 Ovine proinflammatory cytokines Surg. 30, 293300.
10, 119130. Stover, J. F., Schoning, B., Beyer, T. cross the murine blood-brain barrier van Geel, W. J., De Reus, H. P., Nijz-
Shohami, E., Novikov, M., Bass, R., F., Woiciechowsky, C., and Unter- by a common saturable transport ing, H., Verbeek, M. M., Vos, P. E.,
Yamin, A., and Gallily, R. (1994). berg, A. W. (2000). Temporal pro- mechanism. Neuroimmunomodula- and Lamers, K. J. (2002). Measure-
Closed head injury triggers early file of cerebrospinal fluid glutamate, tion 17, 405410. ment of glial fibrillary acidic protein
production of TNF alpha and IL-6 interleukin-6, and tumor necrosis Torabian, S., and Kashani-Sabet, M. in blood: an analytical method. Clin.
by brain tissue. J. Cereb. Blood Flow factor-alpha in relation to brain (2005). Biomarkers for melanoma. Chim. Acta 326, 151154.
Metab. 14, 615619. edema and contusion following con- Curr. Opin. Oncol. 17, 167171. van Horssen, R., Eggermont, A. M.,
Shojo, H., Kaneko, Y., Mabuchi, T., trolled cortical impact injury in rats. Toulmond, S., and Rothwell, N. J. and Ten Hagen, T. L. (2006).
Kibayashi, K., Adachi, N., and Bor- Neurosci. Lett. 288, 2528. (1995). Interleukin-1 receptor Endothelial monocyte-activating
longan, C. V. (2010). Genetic and Strandberg, T. (2009). Adults with antagonist inhibits neuronal dam- polypeptide-II and its functions
histologic evidence implicates role acquired traumatic brain injury: age caused by fluid percussion in (patho)physiological processes.
of inflammation in traumatic brain experiences of a changeover process injury in the rat. Brain Res. 671, Cytokine Growth Factor Rev. 17,
injury-induced apoptosis in the rat and consequences in everyday life. 261266. 339348.
cerebral cortex following moderate Soc. Work Health Care 48, 276297. Touzani, O., Boutin, H., Lefeuvre, R., Van Wagoner, N. J., and Benveniste,
fluid percussion injury. Neuroscience Sullivan, P. G., Bruce-Keller, A. J., Parker, L., Miller, A., Luheshi, G., et E. N. (1999). Interleukin-6 expres-
171, 12731282. Rabchevsky, A. G., Christakos, S., al. (2002). Interleukin-1 influences sion and regulation in astrocytes. J.
Singhal, A., Baker, A. J., Hare, G. Clair, D. K., Mattson, M. P., et ischemic brain damage in the mouse Neuroimmunol. 100, 124139.
M., Reinders, F. X., Schlichter, L. al. (1999). Exacerbation of damage independently of the interleukin- Van Wagoner, N. J., Oh, J. W., Repovic,
C., and Moulton, R. J. (2002). and altered NF-kappaB activation 1 type I receptor. J. Neurosci. 22, P., and Benveniste, E. N. (1999).
Association between cerebrospinal in mice lacking tumor necrosis fac- 3843. Interleukin-6 (IL-6) production by
fluid interleukin-6 concentrations tor receptors after traumatic brain Townend, W. J., Guy, M. J., Pani, M. A., astrocytes: autocrine regulation by
and outcome after severe human injury. J. Neurosci. 19, 62486256. Martin, B., and Yates, D. W. (2002). IL-6 and the soluble IL-6 receptor.
traumatic brain injury. J. Neuro- Svetlov, S. I., Larner, S. F., Kirk, D. Head injury outcome prediction in J. Neurosci. 19, 52365244.
trauma 19, 929937. R., Atkinson, J., Hayes, R. L., and the emergency department: a role Vos, P. E., Jacobs, B., Andriessen, T.
Skouen, J. S., Brisby, H., Otani, K., Wang, K. K. (2009). Biomarkers of for protein S-100B? J. Neurol. Neu- M., Lamers, K. J., Borm, G. F.,
Olmarker, K., Rosengren, L., and blast-induced neurotrauma: profil- rosurg. Psychiatry 73, 542546. Beems, T., et al. (2010). GFAP
Rydevik, B. (1999). Protein markers ing molecular and cellular mecha- Treggiari, M. M., Schutz, N., Yanez, N. and S100B are biomarkers of trau-
in cerebrospinal fluid in experimen- nisms of blast brain injury. J. Neu- D., and Romand, J. A. (2007). Role matic brain injury: an observa-
tal nerve root injury. A study of slow- rotrauma 26, 913921. of intracranial pressure values and tional cohort study. Neurology 75,
onset chronic compression effects or Tarkowski, E., Rosengren, L., Blom- patterns in predicting outcome in 17861793.
the biochemical effects of nucleus strand, C., Wikkelso, C., Jensen, traumatic brain injury: a systematic Vos, P. E., Lamers, K. J., Hendriks, J.
pulposus on sacral nerve roots. Spine C., Ekholm, S., et al. (1995). review. Neurocrit. Care 6, 104112. C., Van Haaren, M., Beems, T., Zim-
24, 21952200. Early intrathecal production of Trembovler, V., Beit-Yannai, E., You- merman, C., et al. (2004). Glial and
Stahel, P. F., Shohami, E., Younis, F. interleukin-6 predicts the size of nis, F., Gallily, R., Horowitz, M., neuronal proteins in serum predict
M., Kariya, K., Otto, V. I., Len- brain lesion in stroke. Stroke 26, and Shohami, E. (1999). Antiox- outcome after severe traumatic brain
zlinger, P. M., et al. (2000). Exper- 13931398. idants attenuate acute toxicity of injury. Neurology 62, 13031310.
imental closed head injury: analy- Tasci, A., Okay, O., Gezici, A. R., Ergun, tumor necrosis factor-alpha induced Wang, X. Q., Peng, Y. P., Lu, J. H., Cao,
sis of neurological outcome, blood- R., and Ergungor, F. (2003). Prog- by brain injury in rat. J. Interferon B. B., and Qiu, Y. H. (2009). Neu-
brain barrier dysfunction, intracra- nostic value of interleukin-1 beta Cytokine Res. 19, 791795. roprotection of interleukin-6 against
nial neutrophil infiltration, and neu- levels after acute brain injury. Neu- Uzan, M., Tanriverdi, T., Baykara, O., NMDA attack and its signal trans-
ronal cell death in mice defi- rol. Res. 25, 871874. Kafadar, A., Sanus, G. Z., Tureci, E., duction by JAK and MAPK. Neu-
cient in genes for pro-inflammatory Taupin, V., Toulmond, S., Serrano, A., et al. (2005). Association between rosci. Lett. 450, 122126.
cytokines. J. Cereb. Blood Flow Benavides, J., and Zavala, F. (1993). interleukin-1 beta (IL-1beta) gene Weckbach, S., Perl, M., Heiland, T.,
Metab. 20, 369380. Increase in IL-6, IL-1 and TNF lev- polymorphism and outcome after Braumuller, S., Stahel, P. F., Flierl, M.
Statler, K. D., Jenkins, L. W., Dixon, C. els in rat brain following traumatic head injury: an early report. Acta A., et al. (2012). A new experimen-
E., Clark, R. S., Marion, D. W., and lesion. Influence of pre- and post- Neurochir. (Wien) 147, 715720; dis- tal polytrauma model in rats: mol-
Kochanek, P. M. (2001). The sim- traumatic treatment with Ro5 4864, cussion 720. ecular characterization of the early
ple model versus the super model: a peripheral-type (p site) benzodi- van Dongen, E. P., Ter Beek, H. T., inflammatory response. Mediators
translating experimental traumatic azepine ligand. J. Neuroimmunol. 42, Boezeman, E. H., Schepens, M. A., Inflamm. 2012, 890816.
brain injury research to the bedside. 177185. Langemeijer, H. J., and Aarts, L. P. Whalen, M. J., Carlos, T. M., Kochanek,
J. Neurotrauma 18, 11951206. Tehranian, R., Andell-Jonsson, S., Beni, (1998). Normal serum concentra- P. M., Wisniewski, S. R., Bell, M.
Stein, D. M., Lindell, A., Murdock, K. S. M., Yatsiv, I., Shohami, E., Bartfai, tions of S-100 protein and changes in J., Clark, R. S., et al. (2000).
R., Kufera, J. A., Menaker, J., Keled- T., et al. (2002). Improved recovery cerebrospinal fluid concentrations Interleukin-8 is increased in cere-
jian, K., et al. (2011). Relationship and delayed cytokine induction after of S-100 protein during and after brospinal fluid of children with

www.frontiersin.org March 2013 | Volume 4 | Article 18 | 17


Woodcock and Morganti-Kossmann Traumatic brain injury, biomarkers, cytokines

severe head injury. Crit. Care Med. Woodroofe, M. N., Sarna, G. S., Wad- of interleukin-1 receptor antago- endothelial cells. J. Cell. Physiol. 226,
28, 929934. hwa, M., Hayes, G. M., Lough- nist. J. Cereb. Blood Flow Metab. 18, 16941701.
Williams, A. J., Wei, H. H., Dave, lin, A. J., Tinker, A., et al. 840847. Ziebell, J. M., Bye, N., Semple, B.
J. R., and Tortella, F. C. (2007). (1991). Detection of interleukin- Yao, C., Williams, A. J., Ottens, A. K., D., Kossmann, T., and Morganti-
Acute and delayed neuroinflamma- 1 and interleukin-6 in adult rat Lu, X. C., Liu, M. C., Hayes, R. Kossmann, M. C. (2011). Attenuated
tory response following experimen- brain, following mechanical injury, L., et al. (2009). P43/pro-EMAPII: neurological deficit, cell death and
tal penetrating ballistic brain injury by in vivo microdialysis: evidence a potential biomarker for discrim- lesion volume in Fas-mutant mice is
in the rat. J. Neuroinflammation 4, of a role for microglia in cytokine inating traumatic versus ischemic associated with altered neuroinflam-
17. production. J. Neuroimmunol. 33, brain injury. J. Neurotrauma 26, mation following traumatic brain
Winnerkvist, A., Anderson, R. E., Hans- 227236. 12951305. injury. Brain Res. 1414, 94105.
son, L. O., Rosengren, L., Estrera, A. Wu, D., and Pardridge, W. M. (1999). Yao, C., Williams, A. J., Ottens, A. K.,
E., Huynh, T. T., et al. (2007). Multi- Neuroprotection with noninvasive May Lu, X. C., Chen, R., Wang, Conflict of Interest Statement: The
level somatosensory evoked poten- neurotrophin delivery to the brain. K. K., et al. (2008). Detection authors declare that the research was
tials and cerebrospinal proteins: Proc. Natl. Acad. Sci. U.S.A. 96, of protein biomarkers using high- conducted in the absence of any com-
indicators of spinal cord injury in 254259. throughput immunoblotting fol- mercial or financial relationships that
thoracoabdominal aortic aneurysm Xie, K. (2001). Interleukin-8 and lowing focal ischemic or penetrating could be construed as a potential con-
surgery. Eur. J. Cardiothorac. Surg. human cancer biology. Cytokine ballistic-like brain injuries in rats. flict of interest.
31, 637642. Growth Factor Rev. 12, 375391. Brain Inj. 22, 723732.
Winter, C. D., Iannotti, F., Pringle, Xu, J., Fan, G., Chen, S., Wu, Y., Xu, X. Yatsiv, I., Morganti-Kossmann, M. C., Received: 26 November 2012; accepted:
A. K., Trikkas, C., Clough, G. M., and Hsu, C. Y. (1998). Methyl- Perez, D., Dinarello, C. A., Novick, 10 February 2013; published online: 04
F., and Church, M. K. (2002). A prednisolone inhibition of TNF- D., Rubinstein, M., et al. (2002). Ele- March 2013.
microdialysis method for the recov- alpha expression and NF-kB acti- vated intracranial IL-18 in humans Citation: Woodcock T and Morganti-
ery of IL-1beta, IL-6 and nerve vation after spinal cord injury in and mice after traumatic brain Kossmann MC (2013) The role of
growth factor from human brain rats. Brain Res. Mol. Brain Res. 59, injury and evidence of neuropro- markers of inflammation in traumatic
in vivo. J. Neurosci. Methods 119, 135142. tective effects of IL-18-binding pro- brain injury. Front. Neurol. 4:18. doi:
4550. Yan, E. B., Hellewell, S. C., Bellan- tein after experimental closed head 10.3389/fneur.2013.00018
Winter, C. D., Pringle, A. K., Clough, der, B. M., Agyapomaa, D. A., and injury. J. Cereb. Blood Flow Metab. This article was submitted to Frontiers in
G. F., and Church, M. K. (2004). Morganti-Kossmann, M. C. (2011). 22, 971978. Neurotrauma, a specialty of Frontiers in
Raised parenchymal interleukin-6 Post-traumatic hypoxia exacerbates Zeitzer, M. B., and Brooks, J. M. Neurology.
levels correlate with improved out- neurological deficit, neuroinflam- (2008). In the line of fire: traumatic Copyright 2013 Woodcock and
come after traumatic brain injury. mation and cerebral metabolism in brain injury among Iraq War vet- Morganti-Kossmann. This is an open-
Brain 127, 315320. rats with diffuse traumatic brain erans. AAOHN J. 56, 347353; quiz access article distributed under the terms
Woiciechowsky, C., Schoning, B., injury. J. Neuroinflammation 8, 147. 354345. of the Creative Commons Attribution
Cobanov, J., Lanksch, W. R., Volk, H. Yang, G. Y., Liu, X. H., Kadoya, C., Zhang, L., Li, H. Y., Li, H., Zhao, J., License, which permits use, distribution
D., and Docke, W. D. (2002). Early Zhao, Y. J., Mao, Y., Davidson, B. Su, L., Zhang, Y., et al. (2011). and reproduction in other forums, pro-
IL-6 plasma concentrations corre- L., et al. (1998). Attenuation of Lipopolysaccharide activated vided the original authors and source
late with severity of brain injury ischemic inflammatory response in phosphatidylcholine-specific phos- are credited and subject to any copy-
and pneumonia in brain-injured mouse brain using an adenovi- pholipase C and induced IL-8 and right notices concerning any third-party
patients. J. Trauma 52, 339345. ral vector to induce overexpression MCP-1 production in vascular graphics etc.

Frontiers in Neurology | Neurotrauma March 2013 | Volume 4 | Article 18 | 18

Vous aimerez peut-être aussi