ArticlePDF Available

Production of cytokines following brain injury: Beneficial and deleterious for the damaged tissue

Authors:
  • Dietrich-Bonhoeffer-Klinikum Neubrandenburg

Abstract and Figures

A profound inflammatory response is initiated immediately following traumatic brain injury (TBI) and is characterized by the release of several cytokines with pro- and anti-inflammatory functions. In order to elucidate which cytokines are released in the human brain in response to injury as well as in the peripheral compartment, IL-1, IL-6, IL-8, IL-10, TNF-alpha and TGF-beta were monitored in CSF and serum of severely brain-injured patients. Furthermore, we investigated the possible modulation of systemic reactions by IL-6 and the ability of IL-6 and IL-8 to promote the synthesis of nerve growth factor.
No caption available
… 
Content may be subject to copyright.
Molecular Psychiatry (1997) 2, 133–136
1997 Stockton Press All rights reserved 1359–4184/97 $12.00
was established in rats with the aim of reproducing and
CYTOKINES IN THE BRAIN
extending the results obtained in humans.
Production of cytokines
Materials and methods
following brain injury:
The patients included in this study had a severe trau-
matic brain injury and were treated according toa stan-
beneficial and deleterious
dardized protocol as previously described.
5–7
CSF and
serum samples were tested by ELISA for interleukin-
for the damaged tissue
(IL)-1, IL-6, IL-8, IL-10, TNF-
a
, transforming growth
factor-beta (TGF-
b
) and nerve growth factor (NGF) as
MC Morganti-Kossman
1
, PM Lenzlinger
1
,
described.
6–8
For the detection of IL-6 in rat serum and
V Hans
1
, P Stahel
2
, E Csuka
1
, E Ammann
1
,
CSF a bioassay was also utilized using 7TD1 cells. The
R Stocker
3
, O Trentz
3
and T Kossmann
3
integrity of the blood–brain barrier (BBB) was deter-
1
Department of Surgery, Division of Research, University
mined by calculating the daily CSF/serum albumin
Hospital Zu
¨
rich, 8091 Switzerland;
2
University of Alabama
quotient (Q
A
).
6
Astrocytes were isolated from newborn
at Birmingham, Department of Microbiology, Birmingham,
mouse brain and stimulated with CSF samples from
Alabama, USA;
3
Division of Trauma Surgery, University
brain-injured patients or with recombinant IL-6 and IL-
Hospital Zu
¨
rich, 8091 Switzerland
8.
6,7
The animal model of diffuse brain injury was
established according to Marmarou et al.
9
A total of 59
animals were used and five animals were killed at each
Keywords: brain injury; interleukins; tumor necrosis factor-
time point from 1 h up to 2 weeks after trauma. The
alpha; transforming growth factor-beta; nerve growth factor;
control group comprised six sham operated animals
astrocytes; acute-phase-response; blood–brain barrier
which were killed after 2 and 24 h.
A profound inflammatory response is initiated immedi-
ately following traumatic brain injury (TBI) and is
Results
characterized by the release of several cytokines with
pro- and anti-inflammatory functions. In order to eluci-
IL-6 and IL-8 were measured in CSF and serum of 22
date which cytokines are released in the human brain in
and 14 patients, respectively. IL-6 concentrations in
response to injury as well as in the peripheral compart-
CSF were found to be 40- to 100-fold higher compared
ment, IL-1, IL-6, IL-8, IL-10, TNF-a and TGF-b were moni-
to serum, during the entire study period.
6
IL-6 reached
tored in CSF and serum of severely brain-injured
the maximal concentration within the first days after
patients. Furthermore, we investigated the possible
trauma and remained elevated during the whole time
modulation of systemic reactions by IL-6 and the ability
of IL-6 and IL-8 to promote the synthesis of nerve
period (see Table 1 for the ranges of cytokines and NGF
growth factor.
in CSF and serum). Since IL-6 is also considered to
Introduction
be the major inducer of the acute phase response, the
concentrations of the acute phase proteins, C-reactive
A traumatic impact to the brain initiates an inflamma-
protein,
a
1-antitrypsin and fibrinogen were measured
tory response characterized by the activation of a num-
in serum. The increase of these proteins correlated
ber of cells and the release of immune mediators which
with the maximal levels of serum IL-6. Interestingly,
may contribute to deleterious secondary brain damage.
IL-6 levels in CSF and serum correlated with each
The kinetics of the release of cytokines and the cell
other when the BBB was dysfunctional as judged by
typesinvolved in cytokine production have been ident-
the Q
A
. On the contrary, no correlation was found
ified in several animal models.
1
As demonstrated in
between CSF- and serum-IL-6 during normal BBB con-
neurological diseases,chronic inflammation within the
ditions.
6
central nervous system (CNS) leads to demyelinating
lesions.
2
On the other hand, increased expression of
Table 1 Ranges of maximal cytokine and NGF concen-
cytokines within the injured brain has also been shown
trations in CSF and serum of brain-injured patients
to trigger neurotrophic factors such as the nerve growth
factor (NGF).
3
Therefore, the evaluation of cytokines
Cytokine or No. of Range of max. Range of max.
may offer further understanding of the pathophysiolog-
NGF patients levels in CSF levels in serum
ical events following TBI.
(pg ml
1
) (pg ml
1
)
The objective of our study was to differentiate
between intrathecal and peripheral production of vari-
TNF-
a
36 0.7–757 0–157
ous cytokines by analyzing the CSF and serum of
IL-1 36 0–6.58 0–36.2
patients following isolated, severe brain injury. The
IL-6 22 140–35500 0–1100
secretion of NGF and its dependence on cytokine pro-
IL-8 14 260–8000 0–2400
duction was also analyzed in the CSF of these patients
IL-10 28 0.5–440 1.8–98.5
and in vitro, using mouse astrocytes. An animal model
TGF-
b
20 0.02–5 0–136
NGF 22 0–12000 not done
of diffuse axonal injury, which represents the most
common type of damage in patients with brain injury,
4
Cytokine production after brain injury
MC Morganti-Kossmann
et al
134
The kinetics of IL-8 showed similarities with the IL- 6 and IL-8) seem to be produced within the brain
whereas the release of others in the CSF may account6 kinetics by being always more elevated in the CSF
compared to serum. In addition, maximal concen- for both penetration from the periphery as well as for
intrathecal synthesis (TNF-
a
, TGF-
b
, and IL-10). Theirtrations of IL-8 in the CSF correlated with severe dis-
turbance of the BBB.
8
release persists for a long time period although the
highest levels seem to be present early after injury.NGF was onlydetected in those patientswith highest
IL-6 and IL-8 concentrations in the CSF during the Cytokine production by cells of the CNS is supported
by several works performed either in vitro or in animalstudy period and reached a maximum of 12 ng ml
1
.
Both cytokines were found to be associated with the models. Moreover, much evidence exists on the ability
of cytokines to modulate not only the functions ofrelease of NGF in the CSF. Furthermore, human CSF
containing IL-6 or IL-8 incubated with cultured mouse immune cells but also of glial cells (reviewed in
Ref. 11).
11
astrocytes induced the production of NGF which was
reduced after preincubation with anti-cytokine anti- The penetration of IL-6 from CSF into serum may
occur as a result of BBB disruption or by an activebodies.
7,8
The animal model of diffuse axonal injury showed a transport mechanism when the BBB is normal.
12
Once
in the periphery, IL-6 may regulate the acute phasesimilar response in terms of IL-6 secretion in CSF and
serum. IL-6 was already detectable in CSF and serum response which was already described in brain-injured
patients.
13
The association of cytokines with the pro-2 h after injury showing a peak at 4 h with a mean con-
centration of 12883 pg ml
1
(n = 5) and returning to duction of NGF, confirmed by in vitro and in vivo stud-
ies
3,14
indicates that the acute inflammation may alsonormal by 16–24 h. Cytokine levels were always higher
in CSF than in serum as observed in humans. Sham promote the repair of the lesioned CNS in terms ofscar-
ring as well as of axonal regeneration (Figure 1 summa-operated animals used for controls only showed a
minor increase of IL-6 in serum but not in CSF. rizes the possible functions of IL-6 in brain-injured
patients).
15
However, persistent production of cyto-IL-10 was found in the CSF and serum of 28 patients.
Serum concentrations were predominantly higher in kines may also be deleterious for the damaged brain.
16
Cytokines have been identified in a number of central20 patients, CSF levels in seven and one showed simi-
lar levels in both fluids. Peak values of IL-10 in both nervous system disorders such as meningitis, HIV-
infection, multiple sclerosis and Alzheimer’s disease.compartments did not correlate with each other. When
compared to the pattern of IL-6 in the CSF, IL-10 These diseases are often accompanied by gradual loss
of motor, sensory or cognitive functions as a conse-appeared often simultaneously with the initial IL-6
peak and remained at low levels while IL-6 decreased. quence of destruction of neuronal tissue due to an
uncontrolled local inflammation.
17
In this regard, TNF-TNF-
a
was detected in 36 patients. Serum values
were found within the normal range (0–6.3 pg ml
1
)in
a
seems to have the major cytotoxic properties as com-
pared to the other cytokines and was shown to be syn-19 patients and were elevated in all the others
(maximum of 157.5 pg ml
1
). CSF concentrations thesized within the plaques in multiple sclerosis
brains.
1
reached 757 pg ml
1
in 29 individuals and were up to
100-fold higher than serum levels in 11 of them. In Therefore, it becomes of fundamental value that anti-
inflammatory mediators such as TGF-
b
and IL-10 orseven patients no TNF-
a
was detected in the CSF. Sev-
eral peaks characterized the patterns of TNF-
a
in both cytokine-antagonists are secreted in order to minimize
the consequences of a perpetuating inflammatoryfluids for each patient.
TGF-
b
was detected in the CSF and serum of 20 response.
18
IL-6 and IL-8 are two cytokines with plei-
otropic activities which besides their immunologicalpatients. The concentrations of TGF-
b
in serum
remained approximately normal, ranging from 9 and functions seem to play an important role in regener-
ation by increasing neuronal survival. The elucidation104 pg ml
1
however they increased continuously
toward the end of the study period. In CSF, the highest of the mechanisms underlying this reaction may offer
new therapeutic aproaches with the aim of decreasingpeaks which reached 5003pg ml
1
were found within
the first 3 days post-trauma. Penetration of this cyto- the incidence of secondary brain damage.
kine through a leakage of the BBB may occur and may
be reflected by the correlation found between the maxi-
Acknowledgements
mal Q
TGF-
b
and Q
A
. However, intrathecal synthesis of
These studies were supported by the Swiss National
TGF-
b
may also take place. In fact, by calculating the
Foundation No. 31.36375.92 and 31.42490.94. The
TGF-
b
index similarly to the IgG index,
10
we found that
authors gratefully acknowledge the work of the staff of
in nine patients this value was greater than the TGF-
b
the intensive care unit of the Division of Trauma Sur-
index calculated in normal individuals. Little or no IL-
gery at the University Hospital, Zu
¨
rich.
1 was detected in both CSF and serum of 36 patients.
References
Discussion
1 Morganti-Kossmann MC, Kossmann T. The immunology
The results described show an ongoing inflammatory
of brain injury. In: Rothwell N (ed.) Immune Responses
response within the brain as a result of trauma and very
in the Nervous System. Bios Scientific Publishers: Oxford,
UK, 1995, pp 159–187.
distinct patterns for each cytokine. Some of them (IL-
Cytokine production after brain injury
MC Morganti-Kossmann
et al
135
Figure 1 Possible systemic and local effects of IL-6 released after brain injury.
2 Hofman FM, Hinton DR, Johnson K, Merril JE. Tumor 8 Kossmann T, Stahel P, Lenzlinger P, Redl H, Dubs R,
Trentz O, Schlag G, Morganti-Kossmann MC. Interleukin-necrosis factor identified in multiple sclerosis brain. J Exp
Med 1989; 170: 607–612. 8 released into the cerebrospinal fluid after brain injury is
associated with blood brain barrier dysfunction and nerve3 Spranger M, Lindholm D, Bandtlow C, Heumann R,
Gnahn H, Naeher-Noe M, Thoenen H. Regulation of nerve growth factor production. J Cerebr Blood F Met (in press).
9 Marmarou A, Abd-Elfattah F, van den Brink W, Campbellgrowth factor synthesis in the rat central nervous system:
comparison between the effects of interleukin-1 and vari- J, Kita H, Demetriadou K. A new model of diffuse brain
injury in rats. J Neurosurg 1994; 80: 291–300.ous growth factors in astrocyte cultures and in vivo. Eur
J Neurosci 1990; 2: 69–76. 10 Tibbling G, Link H. Principles of albumin and IgG analy-
ses in neurological disorders. III. Evaluation of IgG syn-4 Gennarelli TA, Spielman GM, Langfit TW, Gildenberg PL,
Harrington T, Jane JA, Mashall LF, Miller JF, Pitts LH. thesis within the centralnervous system in multiple scler-
osis. Scand J Clin Lab Invest 1977; 37: 397–401.Influence of the type of intracranial lesion on outcome
from severe head injury. J Neurosurg 1992; 56: 26–32. 11 Benveniste E. The role of cytokines in multiple
sclerosis/autoimmune encephalitis and other neurological5 Stocker R, Bernays R, Kossmann T, Imhof HG. Monitoring
and treatment of acute head injury. In: Goris RJA, Trentz O disorders. In: Agarwal B, Puri R (eds). Human Cytokines,
Their Role in Research and Therapy. Blackwell Science:(eds). The Integrated Approach to Trauma Care. Springer
Verlag: Berlin, 1995, pp 197–210. Boston, 1995, pp 195–216.
12 Banks WA, Kastin AJ, Broadwell RD. Passage of cytokines6 Kossmann T, Hans V, Imhof H-G, Stocker R, Grob P,
Trentz O, Morganti-Kossmann MC. Intrathecal and serum across the blood brain barrier. Neuroimmunomodulation
1995; 2: 241–248.interleukin-6 and the acute phase response in patients
with severe traumatic brain injuries. Shock 1995; 4: 13 Young AB, Ott LG, Beard D, Dempsy RJ, Tibbs PA,
McClain CJ. The acute-phase response of the brain injured311–317.
7 Kossmann T, Hans V, Imhof H-G, Trentz O, Morganti- patient. J Neurosurg 1988, 69: 375–380.
14 Lindholm D, Hengerer B, Zafra F, Thoenen H. Trans-Kossmann MC. Interleukin-6 released in human cerebro-
spinal fluid following traumatic brain injury may trigger forming growth factor-
b
1 stimulates the expression of
nerve growth factor in the rat CNS. Devel Neurosci 1990;nerve growth factor production in astrocytes. Brain Res
1996; 713: 143–152. 1: 9–12.
Cytokine production after brain injury
MC Morganti-Kossmann
et al
136
15 Logan A, Oliver JJ, Berry M. Growth factors in CNS repair 18 Rothwell NJ, Relton JK. Involvement of cytokines in acute
and regeneration. Prog Growth Factor Res 1995; 5: 379– neurodegeneration in the CNS. Neurosci Biobehav Rev
405.
1993; 17: 217–227.
16 Campbell IL, Abraham CR, Masliah E, Kemper P, Inglis
JD, Oldstone MBA, Mucke L. Neurological disease in
Correspondence: MC Morganti-Kossmann, PhD, Department
transgenic mice by cerebral overexpression of interleukin-
of Surgery, Division of Research, University Hospital Zu
¨
rich,
6. Proc Natl Acad Sci USA 1993; 90: 10061–10065.
CH-8091 Zu
¨
rich, Switzerland. E-mail: KossmannKchi.usz.ch
17 Morganti-Kossmann MC, Kossmann T, Wahl SM. Cyto-
kines and neuropathology. Trends Pharmacol Sci 1992;
13: 286–291.
... In animal models of midline fluid impaction, the levels of typical proinflammatory cytokines [such as interleukin (IL)-1β and tumour necrosis factor (TNF)-α] in the cortex peaked at 3-9 h [10]. Clinical studies have observed elevated levels of IL-6, IL-8, IL-10, TNF-α, and chemokine CC ligand-2 (CCL2) within the initial two days following TBI [11,12]. The elevation of these cytokines signifies immune cell activation and provides evidence for the correlation between activated immune responses and brain pathology. ...
Article
Full-text available
Background The intricate regulatory relationship between mitochondrial dysfunction, apoptosis, and immune cells remains largely elusive following traumatic brain injury (TBI). Methods The GSE45997 dataset from the Gene Expression Omnibus database and utilized GEO2R to screen for differentially expressed genes (DEGs). Functional enrichment analyses were performed. Mitochondrial gene data from the MitoCarta3.0 database were combined with the DEGs to identify mitochondria-related DEGs (MitoDEGs). The hub MitoDEGs related to apoptosis were further screened. Animal models of TBI were established to investigate the mechanisms underlying mitochondrial dysfunction regulation of apoptosis. Furthermore, we explored the relationship between MitoDEGs/hub MitoDEGs and immune cells using the Spearman correlation method. Results Fifty-seven MitoDEGs were significantly enriched in pathways related to fatty acid degradation and metabolism. We identified three upregulated hub MitoDEGs, namely Dnm1l, Mcl1 and Casp3, were associated with apoptosis. In the animal experiments, we observed significant expression levels of microtubule-associated protein 1 light chain 3 beta (LC3B) surrounding the injury site. Most LC3B-expressing cells exhibited positive staining for Beclin 1 and colocalization analysis revealed the simultaneous presence of Beclin 1 and caspase-3. The Western blot analysis further unveiled a significant upregulation of cleaved caspase-3 levels and LC3B II/LC3B I ratio after TBI. Moreover, the quantity of myeloid cell leukaemia-1 immunoreactive cells was notably higher than that in the control group. Spearman correlation analysis demonstrated strong associations between plasma cells, marginal zone B cells, native CD4 T cells, monocytes, and MitoDEGs/hub MitoDEGs. Conclusions This study sheds light on enhanced fatty acid metabolism following mitochondrial dysfunction and its potential association with apoptosis and immune cell activation, thereby providing new mechanistic insights into the acute phase of TBI.
... This activation of the gut-lung axis constitutes the "third hit, " culminating in the onset or exacerbation of ALI. ABI: Acute brain injury; ALI: Acute lung injury; ARDS: Acute respiratory distress syndrome; CAP: Cholinergic anti-inflammatory pathway; E/NE: Epinephrine/norepinephrine; HPA: Hypothalamic-pituitary-adrenal; MV: Mechanical ventilation increased levels of IL-6, IL-10, IL-8, TNF-α and c-c motif chemokine ligand 2 within the first two days post-TBI before gradually return to normal over a period of several weeks [50][51][52]. This cytokine cataract has been shown to induce astrogliosis and stimulate further microglial activation and axonal dysfunction, indicating an inseparable association between activated immunity and acute ABI [53]. ...
Article
Full-text available
It has been convincingly demonstrated in recent years that isolated acute brain injury (ABI) may cause severe dysfunction of peripheral extracranial organs and systems. Of all potential target organs and systems, the lung appears to be the most vulnerable to damage after ABI. The pathophysiology of the bidirectional brain–lung interactions is multifactorial and involves inflammatory cascades, immune suppression, and dysfunction of the autonomic system. Indeed, the systemic effects of inflammatory mediators in patients with ABI create a systemic inflammatory environment (“first hit”) that makes extracranial organs vulnerable to secondary procedures that enhance inflammation, such as mechanical ventilation (MV), surgery, and infections (“second hit”). Moreover, accumulating evidence supports the knowledge that gut microbiota constitutes a critical superorganism and an organ on its own, potentially modifying various physiological functions of the host. Furthermore, experimental and clinical data suggest the existence of a communication network among the brain, gastrointestinal tract, and its microbiome, which appears to regulate immune responses, gastrointestinal function, brain function, behavior, and stress responses, also named the “gut-microbiome–brain axis.” Additionally, recent research evidence has highlighted a crucial interplay between the intestinal microbiota and the lungs, referred to as the “gut-lung axis,” in which alterations during critical illness could result in bacterial translocation, sustained inflammation, lung injury, and pulmonary fibrosis. In the present work, we aimed to further elucidate the pathophysiology of acute lung injury (ALI) in patients with ABI by attempting to develop the “double-hit” theory, proposing the “triple-hit” hypothesis, focused on the influence of the gut–lung axis on the lung. Particularly, we propose, in addition to sympathetic hyperactivity, blast theory, and double-hit theory, that dysbiosis and intestinal dysfunction in the context of ABI alter the gut–lung axis, resulting in the development or further aggravation of existing ALI, which constitutes the “third hit.”
... The literature reports that bTBI animal models can increase pro-inflammatory somnogenic cytokines in the brain, such as IL-1α, IL-1β, and IL-6 that could contribute to sleep dysregulation (Heyburn et al., 2023). Additionally, patients with TBI have elevated serum levels of pro-inflammatory cytokines (Morganti-Kossman et al., 1997;Morganti-Kossmann et al., 2002), including IL-1β (Tasçı et al., 2003). These findings are consistent with studies of increased sleepiness after TBI in humans and level of TBI severity with IL-1β potentially increasing sleepiness after injury (Watson et al., 2007). ...
Article
Full-text available
Traumatic brain injury (TBI) can induce dysregulation of sleep. Sleep disturbances include hypersomnia and hyposomnia, sleep fragmentation, difficulty falling asleep, and altered electroencephalograms. TBI results in inflammation and altered hemodynamics, such as changes in blood brain barrier permeability and cerebral blood flow. Both inflammation and altered hemodynamics, which are known sleep regulators, contribute to sleep impairments post-TBI. TBIs are heterogenous in cause and biomechanics, which leads to different molecular and symptomatic outcomes. Animal models of TBI have been developed to model the heterogeneity of TBIs observed in the clinic. This review discusses the intricate relationship between sleep, inflammation, and hemodynamics in pre-clinical rodent models of TBI.
... Key cytokines such as IL-1β, TGFβ, TNF, IL-6, and IL-10 are consistently elevated after TBI, directly and indirectly increasing neuronal hyperexcitability and contributing to seizures. For instance, IL-1β is raised in TBI patients prone to epilepsy, and it has been shown to decrease seizure threshold in transgenic mice [96][97][98][99][100][101]. Similarly, TGFβ signaling triggers seizures and neuronal hyperexcitability, and its inhibition can reduce the severity of PTS [102][103][104][105]. IL-6, another critical cytokine, is elevated after TBI and associated with an increased susceptibility to seizures [106][107][108][109][110][111][112][113][114][115]. ...
Article
Full-text available
This paper presents an in-depth exploration of Post-Traumatic Epilepsy (PTE), a complex neurological disorder following traumatic brain injury (TBI), characterized by recurrent, unprovoked seizures. With TBI being a global health concern, understanding PTE is crucial for effective diagnosis, management, and prognosis. This study aims to provide a comprehensive overview of the epidemiology, risk factors, and emerging biomarkers of PTE, thereby informing clinical practice and guiding future research. The epidemiological aspect of the study reveals PTE as a significant contributor to acquired epilepsies, with varying incidence influenced by injury severity, age, and intracranial pathologies. The paper delves into the multifactorial nature of PTE risk factors, encompassing clinical, demographic, and genetic elements. Key insights include the association of injury severity, intracranial hemorrhages, and early seizures with increased PTE risk, and the roles of age, gender, and genetic predispositions. Advancements in neuroimaging, electroencephalography, and molecular biology are presented, highlighting their roles in identifying potential PTE biomarkers. These biomarkers, ranging from radiological signs to electroencephalography EEG patterns and molecular indicators, hold promise for enhancing PTE pathogenesis understanding, early diagnosis, and therapeutic guidance. The paper also discusses the critical roles of astrocytes and microglia in PTE, emphasizing the significance of neuroinflammation in PTE development. The insights from this review suggest potential therapeutic targets in neuroinflammation pathways. In conclusion, this paper synthesizes current knowledge in the field, emphasizing the need for continued research and a multidisciplinary approach to effectively manage PTE. Future research directions include longitudinal studies for a better understanding of TBI and PTE outcomes, and the development of targeted interventions based on individualized risk profiles. This research contributes significantly to the broader understanding of epilepsy and TBI.
... 10,11 TBI induces and modulates circulating levels of selected cytokines, chemokines, and alarmins that activate secondary injury cascades and cause blood-brain barrier (BBB) breakdown, cytotoxic and vasogenic edema, excessive immune cell infiltration, and neuronal apoptosis. 12 Collectively, certain cytokines-small proteins that modulate cell-cell communication and immune reactions (e.g., interleukins [ILs], tumor necrosis factors [TNF]), chemokines-a subclass of cytokines that recruits immune cells toward lesions (e.g., macrophage-associated proteins), and alarmins-damage-associated molecular patterns that trigger and amplify inflammatory cascades (''danger signals''), 13,14 constitute key signaling molecules that bridge primary and secondary TBI, with potentially dynamic roles in TBI outcome. ...
Article
Full-text available
The relationship between systemic inflammation and secondary injury in traumatic brain injury (TBI) is complex. We investigated associations between inflammatory markers and clinical confirmation of TBI diagnosis and prognosis. The prospective TRACK-TBI Pilot (Transforming Research and Clinical Knowledge in Traumatic Brain Injury Pilot) study enrolled TBI patients triaged to head computed tomography (CT) and received blood draw within 24 h of injury. Healthy controls (HCs) and orthopedic controls (OCs) were included. Thirty-one inflammatory markers were analyzed from plasma. Area under the receiver operating characteristic curve (AUC) was used to evaluate discriminatory ability. AUC >0.7 was considered acceptable. Criteria included: TBI diagnosis (vs. OC/HC); moderate/severe vs. mild TBI (Glasgow Coma Scale; GCS); radiographic TBI (CT positive vs. CT negative); 3- and 6-month Glasgow Outcome Scale-Extended (GOSE) dichotomized to death/greater relative disability versus less relative disability (GOSE 1–4/5–8); and incomplete versus full recovery (GOSE <8/ = 8). One-hundred sixty TBI subjects, 28 OCs, and 18 HCs were included. Markers discriminating TBI/OC: HMGB-1 (AUC = 0.835), IL-1b (0.795), IL-16 (0.784), IL-7 (0.742), and TARC (0.731). Markers discriminating GCS 3–12/13–15: IL-6 (AUC = 0.747), CRP (0.726), IL-15 (0.720), and SAA (0.716). Markers discriminating CT positive/CT negative: SAA (AUC = 0.767), IL-6 (0.757), CRP (0.733), and IL-15 (0.724). At 3 months, IL-15 (AUC = 0.738) and IL-2 (0.705) discriminated GOSE 5–8/1–4. At 6 months, IL-15 discriminated GOSE 1–4/5–8 (AUC = 0.704) and GOSE <8/ = 8 (0.711); SAA discriminated GOSE 1–4/5–8 (0.704). We identified a profile of acute circulating inflammatory proteins with potential relevance for TBI diagnosis, severity differentiation, and prognosis. IL-15 and serum amyloid A are priority markers with acceptable discrimination across multiple diagnostic and outcome categories. Validation in larger prospective cohorts is needed. ClinicalTrials.gov Registration: NCT01565551
Article
Cognitive deficits are a common comorbidity with neurological disorders and normal aging. Inflammation is associated with multiple diseases including classical neurodegenerative dementias such as Alzheimer's disease (AD) and autoimmune disorders such as multiple sclerosis (MS), in which over half of all patients experience some form of cognitive deficits. Other degenerative diseases of the central nervous system (CNS) including frontotemporal lobe dementia (FTLD), and Parkinson's disease (PD) as well as traumatic brain injury (TBI) and psychological disorders like major depressive disorder (MDD), and even normal aging all have cytokine-associated reductions in cognitive function. Thus, there is likely commonality between these secondary cognitive deficits and inflammation. Neurological disorders are increasingly associated with substantial neuroinflammation, in which CNS-resident cells secrete cytokines and chemokines such as tumor necrosis factor (TNF)α and interleukins (ILs) including IL-1β and IL-6. CNS-resident cells also respond to a wide variety of cytokines and chemokines, which can have both direct effects on neurons by changing the expression of ion channels and perturbing electrical properties, as well as indirect effects through glia-glia and immune-glia cross-talk. There is significant overlap in these cytokine and chemokine expression profiles across diseases, with TNFα and IL-6 strongly associated with cognitive deficits in multiple disorders. Here, we review the involvement of various cytokines and chemokines in AD, MS, FTLD, PD, TBI, MDD, and normal aging in the absence of dementia. We propose that the neuropsychiatric phenotypes observed in these disorders may be at least partially attributable to a dysregulation of immunity resulting in pathological cytokine and chemokine expression from both CNS-resident and non-resident cells.
Article
Full-text available
Adipose tissue has recently been recognized as an important endocrine organ that plays a crucial role in energy metabolism and in the immune response in many metabolic tissues. With this regard, emerging evidence indicates that an important crosstalk exists between the adipose tissue and the brain. However, the contribution of adipose tissue to the development of age‐related diseases, including Alzheimer's disease, remains poorly defined. New studies suggest that the adipose tissue modulates brain function through a range of endogenous biologically active factors known as adipokines, which can cross the blood–brain barrier to reach the target areas in the brain or to regulate the function of the blood–brain barrier. In this review, we discuss the effects of several adipokines on the physiology of the blood–brain barrier, their contribution to the development of Alzheimer's disease and their therapeutic potential. LINKED ARTICLES This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc
Article
Damage to the axonal white matter tracts within the brain is a key cause of neurological impairment and long-term disability following traumatic brain injury (TBI). Understanding how axonal injury develops following TBI requires gyrencephalic models that undergo shear strain and tissue deformation similar to the clinical situation and investigation of the effects of post-injury insults like hypoxia. The aim of this study was to determine the effect of post-traumatic hypoxia on axonal injury and inflammation in a sheep model of TBI. Fourteen male Marino sheep were allocated to receive a single TBI via a modified humane captive bolt animal stunner, or sham surgery, followed by either a 15 minute period of hypoxia or maintenance of normoxia. Head kinematics were measured in injured animals. Brains were assessed for axonal damage, microglia and astrocyte accumulation and inflammatory cytokine expression at 4 hrs following injury. Early axonal injury was characterised by calpain activation, with significantly increased SNTF immunoreactivity, a proteolytic fragment of alpha-II spectrin, but not with impaired axonal transport, as measured by amyloid precursor protein (APP) immunoreactivity. Early axonal injury was associated with an increase in GFAP levels within the CSF, but not with increases in IBA1 or GFAP+ve cells, nor in levels of TNFα, IL1β or IL6 within the cerebrospinal fluid or white matter. No additive effect of post-injury hypoxia was noted on axonal injury or inflammation. This study provides further support that axonal injury post-TBI is driven by different pathophysiological mechanisms, and detection requires specific markers targeting multiple injury mechanisms. Treatment may also need to be tailored for injury severity and timing post-injury to target the correct injury pathway.
Article
Full-text available
Traumatic brain injury usually results in neuronal loss and cognitive deficits. Promoting endogenous neurogenesis has been considered as a viable treatment option to improve functional recovery after TBI. However, neural stem/progenitor cells (NSPCs) in neurogenic regions are often unable to migrate and differentiate into mature neurons at the injury site. Transglutaminase 2 (TGM2) has been identified as a crucial component of neurogenic niche, and significantly dysregulated after TBI. Therefore, we speculate that TGM2 may play an important role in neurogenesis after TBI, and strategies targeting TGM2 to promote endogenous neural regeneration may be applied in TBI therapy. Using a tamoxifen-induced Tgm2 conditional knockout mouse line and a mouse model of stab wound injury, we investigated the role and mechanism of TGM2 in regulating hippocampal neurogenesis after TBI. We found that Tgm2 was highly expressed in adult NSPCs and up-regulated after TBI. Conditional deletion of Tgm2 resulted in the impaired proliferation and differentiation of NSPCs, while Tgm2 overexpression enhanced the abilities of self-renewal, proliferation, differentiation, and migration of NSPCs after TBI. Importantly, injection of lentivirus overexpressing TGM2 significantly promoted hippocampal neurogenesis after TBI. Therefore, TGM2 is a key regulator of hippocampal neurogenesis and a pivotal therapeutic target for intervention following TBI.
Article
Full-text available
Frozen brain specimens from patients with multiple sclerosis (MS) and other neurologic diseases were analyzed using immunocytochemical techniques for the presence of TNF. In brain lesions in MS, and subacute sclerosing panencephalitis, TNF+ cells were demonstrated. At the lesion site in MS, TNF+ staining is associated with both astrocytes and macrophages. These observations were not made in Alzheimer's disease or normal brain tissue. The presence of TNF in MS lesions suggests a significant role for cytokines and the immune response in disease progression.
Article
Full-text available
Recent studies attempting to define the outcome from severe head injury have implied, directly or indirectly, that the severity of injury (as determined by the Glasgow Coma Scale (GCS)) is the sole determinant of outcome. Little attention has been focused on the type of lesion that causes the low GCS score, and there exists an unstated hypothesis that the lesion type is not an important determinant of outcome. No attempt has been made to determine whether patients who have the same GCS score caused by different lesions have the same or different outcomes. Since this is impossible to test without a large number of cases, data were obtained from seven head-injury centers on patients who fulfilled the Glasgow criteria for severe head injury (GCS ≤ 8 for at least 6 hours). Patients were categorized according to a simple classification system comprising seven lesion types, each of which was further subdivided into two GCS score ranges (3 to 5 and 6 to 8). Of 1107 patients, the overall mortality was 41%, but ranged from 9% to 74% among the different lesion categories. Conversely, 26% had good recovery (at 3 months), but among the different lesion groups the range was 6% to 68%. Acute subdural hematoma with GCS scores of 3 to 5 was uniformly the worst problem (74% mortality and 8% good recovery), whereas diffuse injury coma of 6 to 24 hours with GCS scores of 6 to 8 had 9% mortality and 68% incidence of good recovery. Results of this study demonstrate marked heterogeneity within this severe head-injury group and point out that patients with the same GCS score have markedly different outcomes, depending on the causative lesion. The type of lesion is thus as important a factor in determining outcome as is the GCS score, and both must be considered when describing severely head-injured patients.
Article
Full-text available
Cytokines are thought to be important mediators in physiologic and pathophysiologic processes affecting the central nervous system (CNS). To explore this hypothesis, transgenic mice were generated in which the cytokine interleukin 6 (IL-6), under the regulatory control of the glial fibrillary acidic protein gene promoter, was overexpressed in the CNS. A number of transgenic founder mice and their offspring exhibited a neurologic syndrome the severity of which correlated with the levels of cerebral IL-6 expression. Transgenic mice with high levels of IL-6 expression developed severe neurologic disease characterized by runting, tremor, ataxia, and seizure. Neuropathologic manifestations included neuro-degeneration, astrocytosis, angiogenesis, and induction of acute-phase-protein production. These findings indicate that cytokines such as IL-6 can have a direct pathogenic role in inflammatory, infectious, and neurodegenerative CNS diseases.
Article
A total of 116 patients were subgrouped according to the presence of neurological disorder and abnormal S-albumin, S-IgG, S-haptoglobin and S-CRP. Abnormal serum protein concentrations were registered in 21 % of patients with neurological symptoms but no signs of organic neurological disorder, and in 58% of patients with various neurological disorders except multiple sclerosis. Increased S-IgG and S-haptoglobin were most common. The CSF/S albumin ratio is proposed to be a more sensitive and adequate parameter for the demonstration of a blood-brain damage than CSF-protein or CSF-albumin. In patients with serum protein abnormalities the CSF IgG/protein and CSF IgG/albumin ratios were increased in 28% and 44% of the patients without organic neurological disease and in 40% and 51% of the patients with neurological disorders except multiple sclerosis. The IgG-index = (CSF/S IgG ratio)/(CSF/S albumin ratio) was elevated only in 8% and 15%. The IgG-index is a better measure for IgG synthesis within the CNS than the other two.
Article
Inflammatory processes in the brain require the cooperation of immunocompetent cells and glial cells, which communicate by secreting bidirectional mediators. Resident cells within the nervous system can synthesize and secrete inflammatory cytokines, as well as neuropeptides, contributing to the response within the CNS to injury or immunological challenge. Although the mechanisms of cell activation and immune interaction are poorly understood, accumulating evidence implicates these pathways in neuropathogenesis, as described here by Sharon Wahl and colleagues. For example, in the acquired immune deficiency syndrome (AIDS), HIV-1-induced nervous system dysfunction and dementia are associated with the presence of infiltrating leukocytes and the release of inflammatory cytokines. Defining the pathways of cytokine dysregulation and neurotoxicity invoked by the infiltrating leukocytes, as well as the contribution of the neural cells themselves, may help to identify mechanisms of intervention in this and other debilitating CNS diseases.
Article
The acute response to injury and infection is manifested by increased synthesis of acute-phase proteins by the liver, an increased white blood cell count, fever, a negative nitrogen balance, and altered serum mineral levels (zinc, iron, and copper). This response is thought to be partially mediated by cytokines such as interleukin-1, but has not been well studied in head-injured patients. In this study, 25 patients were studied for evidence of the acute-phase response extending from hospital admission up to 21 days postinjury. The patients were divided into two groups to determine if severity of injury influenced the response. Group 1 consisted of nine patients with admission peak 24-hour Glasgow Coma Scale (GCS) scores of 4 or less; Group 2 consisted of 16 patients with admission peak 24-hour GCS scores of 8 or greater. All patients demonstrated some evidence of the acute-phase response. Serum alpha-1 acid glycoprotein, ceruloplasmin, and C-reactive protein levels were elevated on admission and throughout the study. Serum albumin and zinc levels were depressed on admission; zinc levels gradually normalized by Day 21 in both groups, but hypoalbuminemia was observed throughout the study period. Serum copper levels were normal on admission but increased to above normal in both groups by Day 11 postinjury. Urinary urea nitrogen excretion was elevated in both groups and peaked on Day 7 for Group 1 and Day 11 for Group 2 patients. The patients with admission GCS scores equal to or less than 4 had overall higher temperatures than were seen in those with GCS scores greater than or equal to 8 (p = 0.009). All patients but one had an elevated white blood cell count on admission. It is concluded that brain-injured patients with admission GCS scores of 3 to 4 and 8 to 14 demonstrate an acute-phase response which lasts for at least 3 weeks postinjury. It is speculated that this response is at least partially mediated by increased intraventricular interleukin-1 activity.
Article
This report describes the development of an experimental head injury model capable of producing diffuse brain injury in the rodent. A total of 161 anesthetized adult rats were injured utilizing a simple weight-drop device consisting of a segmented brass weight free-falling through a Plexiglas guide tube. Skull fracture was prevented by cementing a small stainless-steel disc on the calvaria. Two groups of rats were tested: Group 1, consisting of 54 rats, to establish fracture threshold; and Group 2, consisting of 107 animals, to determine the primary cause of death at severe injury levels. Data from Group 1 animals showed that a 450-gm weight falling from a 2-m height (0.9 kg-m) resulted in a mortality rate of 44% with a low incidence (12.5%) of skull fracture. Impact was followed by apnea, convulsions, and moderate hypertension. The surviving rats developed decortication flexion deformity of the forelimbs, with behavioral depression and loss of muscle tone. Data from Group 2 animals suggested that the cause of death was due to central respiratory depression; the mortality rate decreased markedly in animals mechanically ventilated during the impact. Analysis of mathematical models showed that this mass-height combination resulted in a brain acceleration of 900 G and a brain compression gradient of 0.28 mm. It is concluded that this simple model is capable of producing a graded brain injury in the rodent without a massive hypertensive surge or excessive brain-stem damage.
Article
Cytokines, (particularly interleukins and growth factors) are synthesised in the brain, and induced by brain damage. Interleukin-I appears to directly mediate ischaemic and excitotoxic brain damage, whereas growth factors (e.g., bFGF, NGF), and the phospholipid binding protein lipocortin-1 exhibit neuroprotective actions. Central administration of recombinant interleukin-1 receptor antagonist markedly attenuates damage induced by focal cerebral ischaemia, or pharmacological activation of NMDA receptors in the rat brain. The mechanisms of action of these cytokines on neurodegeneration are unknown, but indirect evidence has implicated corticotropin releasing factor, arachidonic acid, and nitric oxide. In vitro effects of interleukin-1, growth factors, and lipocortin-1 have been reported on intracellular calcium homeostasis, which is critically important in neurodegeneration. Pharmacological modulation of the expression and/or actions of cytokines in the brain may be of considerable therapeutic benefit in the treatment of acute neurodegeneration.
Article
Patients with severe traumatic brain injury (TBI) show a profound acute-phase response. Because interleukin-6 (IL-6) is an important mediator of these pathophysiological changes, IL-6 levels were monitored in the cerebrospinal fluid (CSF) and serum of 20 patients with severe isolated TBI. All patients received indwelling ventricular catheters for intracranial pressure monitoring and for release of CSF when intracranial pressure exceeded 15 mmHg. CSF and blood samples were drawn daily for up to 14 days. The CSF/serum albumin ratio (QA) served as a parameter of blood brain barrier dysfunction. Differential blood counts as well as the acute-phase proteins C-reactive protein, alpha 1-antitrypsin, and fibrinogen were recorded. IL-6 was detected in all CSF samples and reached values of up to 31,000 pg/mL, while serum levels remained significantly lower (alpha < or = .01) and never exceeded 1,100 pg/mL the entire study period. A correlation between CSF and serum IL-6 was found initially after the trauma and corresponded to a severe dysfunction of the blood brain barrier (r = .637, p = .001). Maximum IL-6 concentrations in serum correlated with peak levels of acute-phase proteins (C-reactive protein, alpha 1-antitrypsin, and fibrinogen). With regard to blood cell count, an initial leukocytosis combined with a borderline lymphocytopenia was observed. Thrombocytes decreased to a subnormal level during the first few days, but reached supranormal numbers by the end of the study period. Our results show that the increase of IL-6 levels in CSF and serum is followed by a profound acute-phase response in patients with TBI. Because cytokine concentrations are significantly lower in serum compared with CSF, we hypothesize that IL-6 produced in the central nervous system may play a role in initiating the acute-phase response.