ArticlePDF AvailableLiterature Review

Recent advances in neuroprotection for treating traumatic brain injury

Taylor & Francis
Expert Opinion on Investigational Drugs
Authors:
  • Thomas F. Frist, Jr. College of Medicine Belmont University

Abstract

The fact that traumatic brain injury is the leading cause of death and disability in the most active population (< 45 years of age) of industrialised countries underscores the need for intensified efforts to define and implement effective neuroprotective strategies. However, despite progressively growing knowledge on the mechanisms involved in the pathobiology of traumatic brain injury and promising preclinical findings, most of the neuroprotection trials have failed to deliver the expected level of beneficial effects. Some of the possible reasons underlying the lack of success of these clinical trials are addressed in this review, which describes some of the most promising and/or controversial ongoing clinical trials from their pathophysiological basis. In addition, new neurobiological findings and their consequence for novel neuroprotective approaches are discussed.
Review
10.1517/13543784.15.11.1371 © 2006 Informa UK Ltd ISSN 1354-3784 1371
Central & Peripheral Nervous Systems
Recent advances in
neuroprotection for treating
traumatic brain injury
Ibolja Cernak
Johns Hopkins University Applied Physics Laboratory, Biomedicine Business Area, 11100 Johns Hopkins
Road, Laurel, MD 20723-6099, USA
The fact that traumatic brain injury is the leading cause of death and dis-
ability in the most active population (< 45 years of age) of industrialised
countries underscores the need for intensified efforts to define and imple-
ment effective neuroprotective strategies. However, despite progressively
growing knowledge on the mechanisms involved in the pathobiology of
traumatic brain injury and promising preclinical findings, most of the neuro-
protection trials have failed to deliver the expected level of beneficial effects.
Some of the possible reasons underlying the lack of success of these clinical
trials are addressed in this review, which describes some of the most pro-
mising and/or controversial ongoing clinical trials from their patho-
physiological basis. In addition, new neurobiological findings and their
consequence for novel neuroprotective approaches are discussed.
Keywords: nanotechnology, neuroprotection, traumatic brain injury
Expert Opin. Investig. Drugs (2006) 15(11):1371-1381
1. Introduction
Traumatic brain injury (TBI) has frequently been called ‘silent epidemic’ of the
modern world referring to the fact that TBI is the leading cause of death and
disability in the most active population (< 45 years of age) of industrialised coun-
tries
[1]. Moreover, recent data report that the incidence of TBI in the USA is close
to 1.4 million people/year and, among them, an estimated 80,000 – 90,000 experi-
ence the onset of long-term disability
[2]. These findings underscore the need for
intensified efforts to define and implement effective neuroprotective strategies.
Indeed, there is an increasing awareness of what the implications of TBI and its
long-term consequences for society in economic, social and human are. This
resulted in 58 currently ongoing clinical trials concerning preventive measures,
diagnosis, treatment and rehabilitation of TBI that are registered at the Clinical
Trials website
[201], a service of the US NIH and developed by the National Library
of Medicine. Interestingly, although numerous experimental studies showed pro-
mising neuroprotective effects in experimental models of TBI, almost all of the clin-
ical trials have failed to demonstrate significant success in treating brain-injured
patients
[3]. One of the reasons underlying the relatively small success of clinical tri-
als is the complexity of the pathobiology of TBI; that is, TBI inflicts not only direct
mechanical damage to tissue but initiates complex biochemical changes resulting in
delayed neural cell loss (
Figure 1) [4]. It has been established that secondary injury
mechanisms via intricate interactions play an essential role in chronic neurological
disability. The lack of success of previous therapeutic efforts targeting only one indi-
vidual factor may be explained by the multifactorial nature of TBI. Based on the
concept of delayed biochemical injury, many drugs have proven to be effective in
1. Introduction
2. Clinical trials and high hopes
3. Experimental promises
4. Conclusion
5. Expert opinion
Recent advances in neuroprotection for treating traumatic brain injury
1372 Expert Opin. Investig. Drugs (2006) 15(11)
experimental neurotrauma [4]; however, no pharmacotherapy
has yet been found to be effective in clinical head injury
[3].
The development of clinically relevant experimental mod-
els of TBI (which successfully replicate the most important
changes reported in clinical head injuries such as brain
oedema, elevated intracerebral pressure, reduced cortical per-
fusion, decreased cerebral blood flow, neuroendocrine and
metabolic changes, and coma as well as mechanisms under-
lying neuronal cell death and resulting neurological deficits
following TBI) represent a valuable tool for developing novel
therapeutic approaches for brain injuries
[5]. Indeed, a better
knowledge of the pathophysiological mechanisms of head
injuries prompted clinical researchers to design and imple-
ment various therapeutic approaches aiming to modify the
harmful mechanisms and improve the outcome. This review
describes some of the most promising and/or controversial
ongoing clinical trials and explains their pathophysiological
basis. In addition, new neurobiological findings and their
relevance for novel neuroprotective approaches are discussed.
2. Clinical trials and high hopes
2.1 Dexanabinol
Numerous clinical trials have been conducted in the past
without significant effects (see Section 1)
[4,6,7]. Among the
trials, which failed to deliver the expected level of neuro-
protection, were the evaluation of corticosteroids
[8], barbitu-
rates
[9], glutamate antagonists [10], free radical scavengers [11]
and Ca
2+
channel blockers [12,13]. One of the clinical trials
with high expectations tested the efficacy of dexanabinol, a
synthetic cannabinoid analogue
[14] with strong neuro-
protective potential demonstrated in experimental models of
TBI
[15,16]. Experimental studies using various models of TBI
demonstrated that dexanabinol targeted various
pathophysiological mechanisms involved in TBI-induced
neurological deficits; it reduced glutamate excitoxicity, free
radical production and inflammatory response
[17] among
others. After the Phase I trial showed that dexanabinol
200 mg was safe in healthy volunteers, Phase II results indi-
cated improved control of intracranial pressure
[18] and a
Phase III trial was instigated
[19]. It was a big disappointment
when the final results showed no improvements in the control
of intracranial pressure or quality of life in the group of dex-
anabinol-treated patients versus placebo-treated group and
the subgroup analysis reported no indication of differential
treatment effects
[19].
2.2 Magnesium salts
Although the mechanisms of neuroprotection by magnesium
are not fully understood, they include a reduction of
presynaptic release of glutamate and blocking the glutama-
tergic N-methyl-
D-aspartate receptors, as well as blockage of
Ca
2+
entry via voltage-gated Ca
2+
channels and potentiation
of adenosine action among others
[20]. Preclinical studies
using experimental models of TBI (which replicated con-
tusion, diffuse axonal injury, oedema and subarachnoid haem-
orrhage
[21,22]) demonstrated significant beneficial effects of
magnesium. Combined with findings of limited clinical stud-
ies involving patients with brain injury
[23,24], the results of
these studies suggest that magnesium salts (magnesium sulfate
and magnesium chloride) are promising neuroprotective
agents for clinical use
[4]. Surprisingly, the Phase III clinical
trial evaluating the effects of magnesium sulfate for brain
injury and organised by the University of Washington showed
a negative outcome, with an increase of 25% in mortality in
the treated group compared with placebo controls (Temkin
NR, personal communication). Together with the results of a
large-scale, randomised clinical trial using magnesium sulfate
Figure 1. Schematic presentation of the most essential mechanisms underlying neurological deficits caused by TBI.
ICP: Intracranial pressure; TBI: Traumatic brain injury.
Necrosis Apoptosis
Neuronal cell death
Increased ICP
TBI
Impaired cerebral
vasoregulation
Mechanical tissue
destruction
Secondary injury
mechanisms
Brain oedema
Decreased cerebral
perfusion
Cernak
Expert Opin. Investig. Drugs (2006) 15(11) 1373
in acute stroke [25] and some previous clinical studies [26,27],
these findings have tempered the initial enthusiasm for a
neuroprotective benefit of magnesium
[28]. Currently, there is
an ongoing clinical trial evaluating putative neuroprotective
effects of magnesium sulfate in premature infants (Thomas
Jefferson University).
Nevertheless, further data on magnesiums neuroprotective
role in brain injury emerged with new studies. Chan et al.
[29]
reported that magnesium sulfate increased tissue oxygenation
and reduced hypoxia in patients undergoing temporary surgi-
cal artery occlusion. Moreover, van den Bergh et al. and the
MASH (Magnesium and Acetylsalicylic Acid in Subarachnoid
Hemorrhage) Study Group
[30] demonstrated that magnesium
sulfate therapy significantly reduced the frequency of delayed
cerebral ischaemia in patients with subarachnoidal haem-
orrhage. Recent experimental studies also report beneficial
effects of magnesium in TBI, thus further contributing to the
understanding of magnesium-related neuroprotection
[31,32].
Hopefully, further experimental studies and additional strictly
defined and controlled clinical trials will clarify whether mag-
nesium is neuroprotective in TBI and, if it is, under which
conditions and in which population.
It is noteworthy that both dexanabinol and magnesium are
multi-potential agents targeting several pivotal mechanisms
that result in long-term post-traumatic neurological deficits.
Why have these trials failed to deliver the highly expected
neuroprotection contrary to preclinical experimental studies?
There are multiple potential reasons for these failures
[33,34].
The first is the heterogeneity of the TBI patient population.
Indeed, these patients often have various degrees of tissue
damage, contusion, ischaemia, brain oedema, diffuse axonal
injury, and presence or absence of subarachnoidal haem-
orrhage. This amplifies the necessity of a stringent stratifica-
tion of patients into well-defined subgroups. Moreover,
clinical trials generally include patients with severe TBI, which
excludes the opportunity to analyse the effects of a treatment
on graded brain injury. Second, subjects included in clinical
trials are of both genders, whereas accumulating evidence
shows a distinct gender-dependent pathobiology of TBI
[35].
There are other aspects that could also significantly affect the
success of clinical trials; brain penetration, safety, tolerability
of the compound, and the interface between the pharma-
ceutical industry and academics are among the most
important factors
[36].
On the other hand, experimental studies standardise their
injury model to reproduce graded, usually moderate to severe,
neurological deficits in which positive therapeutic effects can be
statistically confirmed using the minimal number of animals.
Animals with severe injury that are not expected to benefit
from the treatment and those with mild injury that would
probably not show statistically observable benefit from the
drug are routinely excluded from research studies
[37]. More-
over, although mild TBI is the most frequently seen form of the
head injury in the human population, often underdiagnosed
and undertreated, animal ethics committees do not approve a
large experimental study of mild injury when a benefit has
already been demonstrated for moderate injury. All of these dif-
ferences in designing and analysing preclinical versus clinical
studies significantly contribute to obvious discrepancies in
study or trial outcomes.
2.3 Hypothermia
Currently, there are two ongoing clinical trials evaluating the
use of hypothermia in brain-injured patients that are reg-
istered
[101]. The first trial, NABIS:H (National Acute Brain
Injury Study: Hypothermia), was verified by the University of
Texas Health Science Center in September 2005. The
NABIS:H is a randomised clinical trial that will include 240
patients with severe brain injury aged 16 – 45 years and ran-
domised to standard treatment at normothermia or to stand-
ard treatment with moderate hypothermia (32.5 – 34°C for
48 h). Hypothermia will be induced to < 35.5°C by < 2.5 h
after severe TBI, reaching 33°C by 4 h after injury and main-
tained for 48 h. The primary outcome measures will be the
dichotomised Glasgow Outcome Scale (GOS; good outcome:
good recovery or moderate disability; poor outcome: severe
disability, vegetative or dead) and measured by blinded assess-
ment of patients at 6 months postinjury. The second study,
Therapeutic Hypothermia for Severe Traumatic Brain Injury
in Japan, is a Phase III multi-centre trial that was verified by
Yamaguchi University Hospital in August 2005. This
randomised clinical trial includes patients with severe TBI
(Glasgow Coma Scale [GCS] 4 – 8) of both genders and aged
15–69years. The participants are randomly assigned into
either the mild hypothermia (32 – 34°C) and anti-
hyperthermia (35 – 37°C) group. Hypothermia is induced
within 6 h after TBI and the body temperature is kept at these
levels for 72 h. The end point parameters (GOS and total
medical expenses) are evaluated at 6 months postinjury.
Experimental
[38,39] and pilot clinical [40,41] studies have
demonstrated the neuroprotective effects of moderate hypo-
thermia in brain ischaemia and TBI. The multifactorial
mechanisms through which hypothermia may protect the
brain include reducting metabolic rate, blocking of excito-
toxic mechanisms, Ca
2+
antagonism, preserving protein syn-
thesis, regulating cerebral blood flow, reducing brain
thermopooling, decreasing oedema development and modu-
lating inflammatory response
[42]. Due to highly encouraging
preclinical findings, the results of the multi-centre NABISH-1
(National Acute Brain Injury Study using Hypothermia 1)
trial evaluated the effects of brain cooling on TBI outcome
and have been awaited with great enthusiasm. However, the
final report of the NABISH-1 study, which was published in
the New England Journal of Medicine
[43], brought great dis-
appointment showing that moderate hypothermia failed to
improve the outcome of brain-injured patients. Briefly, a total
of 392 patients (16 – 65 years of age) with severe head injury
were enrolled, 193 of whom were assigned to standard treat-
ment and 199 to standard treatment plus hypothermia.
Moderate hypothermia (33°C) was initiated within 6 h of
Recent advances in neuroprotection for treating traumatic brain injury
1374 Expert Opin. Investig. Drugs (2006) 15(11)
injury and maintained for 48 h via surface cooling. The effi-
cacy of the treatment was measured by dichotomised GOS at
6 months postinjury. The analysis of results showed no signif-
icant difference in mortality. The lack of success was
explained by the differences in the percentages of patients
who were hypothermic on admission and in the protocols of
re-warming, imbalances in randomisation and wide variability
in management between the high- and low-enrolment centres
[43,44]. As moderate hypothermia has been shown to be one of
the most powerful neuroprotective methods in experimental
models of TBI and stroke, its lack to induce neuroprotection
in the clinical environment is a puzzling paradox. This may be
the reason for new clinical trials, which intend to use more
homogenous and strictly defined clinical protocols.
2.4 Decompressive craniectomy
High intracranial pressure (ICP) is one of the most frequent
causes of death and disability after severe TBI
[45,46].
Increased brain water content (i.e., brain oedema, increased
cerebral blood volumes and/or mass lesions) has been shown
as a key contributor to elevated ICP. Clinically relevant ICP
increase is generally defined as ICP above a threshold of
15 20 mmHg measured within subdural, intraventricular,
extradural or intraparenchymal compartments. Very often,
when high ICP is not associated with removable intracranial
mass (as in the case with diffuse brain swelling after TBI),
drug therapies are frequently unsuccessful to regulate
ICP
[47]. Currently, most trauma centres approach high ICP
using the recommendations of the Guidelines for the
Management of Severe Traumatic Brain Injury
[48]. These
guidelines include general maneuvers such as head elevation,
sedation, resuscitation and so on; CSF drainage, moderate
hypocapnia (i.e., partial pressure of CO
2
[pCO
2
] of
30–35mmHg) and mannitol administration as the first-line
therapeutic measures, and finally, the ‘second tier’ procedures,
which are indicated when the high ICP is refractory to previ-
ous measures. Decompressive craniectomy (DC), a method
to convert the confined-space skull into an open one by
removing a variable size of a bone
[49,50], besides high-dose
barbiturates and severe hyperhyperventilation (pCO
2
of
< 30 mmHg), belong to this group.
The DECRA (Early Decompressive Craniectomy in
Patients with Severe Traumatic Brain Injury) trial is a
multi-centre randomised trial verified by the National Trauma
Research Institute of Australia in September 2005. The goal
of this trial is to evaluate the effect of early DC on neuro-
logical function in patients with severe TBI. The DECRA
trial includes patients with severe TBI of both genders and
aged 15 – 60 years who admitted to the hospital within the
first 72 h from injury. Severe brain injury for the purpose of
inclusion criteria is defined as: GCS < 9 with computed tomo-
graphy (CT) scan showing evidence of brain oedema (grade
DII plus some evidence of DIII or DIV); GCS > 8 established
before intubation together with brain oedema graded as DIII
or DIV, and a CT scan showing basal cistern compression
with midline shift; or refractory ICP existing for > 15 min
continuously or cumulatively over 1 h, despite the best
conventional management (as defined in the study protocol).
Primary end point measures include GOS measured 6 months
after injury, whereas secondary outcomes are defined as mean
and maximum ICP measured hourly, in-hospital mortality
rate during 6 and 12 months, and number of days spent in the
intensive care unit of the hospital. The intended sample size is
499 patients from 40 centres. The RESCUE-ICP (Ran-
domised Evaluation of Surgery with Craniectomy for Uncon-
trollable Elevation of Intracranial Pressure) trial is another
currently ongoing clinical trial although it is not registered
[201]. The trial will include 400 patients of 10 – 65 years of age
with severe TBI and admitted in the specialised intensive care
units of 40 centres. A standard protocol is proposed to main-
tain ICP < 25 mmHg by applying treatment in 2 stages. After
these stages are unsuccessful in controlling ICP, the patients
will be randomised to continuation of medical management
versus DC
[42].
Interestingly, despite numerous small series and case reports
suggesting neuroprotective effects of DC in severe TBI
patients, a comprehensive review of the available literature
failed to show that the routine use of secondary DC sig-
nificantly improves the outcome in adults with severe TBI
and refractory high ICP
[42]. On the other hand, a small pro-
spective, randomised clinical trial of secondary DC, which
included 27 children (< 18 years of age) with severe TBI
(14 with medical management alone and 13 with DC and
medical treatment) demonstrated that DC effectively reduced
refractory ICP, decreased mortality and improved functional
outcome
[51]. This discrepancy may be caused by a funda-
mental difference between injury mechanisms in young com-
pared with adult brain. Indeed, it has been shown that
children more frequently develop diffuse cerebral swelling
after TBI compared with adults
[52], and that the
TBI-induced overactivation of ongoing physiological pro-
grammed neuronal death (apoptosis) is unique for the devel-
oping brain
[53]. Defining and taking into account the
age-dependent differences in the pathomechanisms of TBI are
necessary to develop new neuroprotective approaches, and
design and analyse clinical trials.
As DC is widely used in treating the civilian and military
adult population with severe head injury, regardless of the
obvious contradiction in the related literature, further
well-designed randomised multi-centre studies are needed
to clarify the potential values of DC. Hopefully, the
ongoing DECRA and RESCUE-ICP trials will take into
account some of the lessons learned in previous non-con-
trolled studies and recommendations made on the basis of
that knowledge
[42].
2.5 Therapy against brain oedema
Management of post-traumatic brain oedema remains largely
empirical and it has not changed in 50 years. Brain oedema,
a potentially devastating complication of TBI, is defined as an
Cernak
Expert Opin. Investig. Drugs (2006) 15(11) 1375
increased volume of the brain caused by pathological fluid
accumulation and has been established having significant role
in ICP increase, impairments in cerebral blood flow and
decreased cerebrospinal fluid
[54]. The exact mechanisms
underlying brain oedema formation following TBI are not
entirely clarified. Accumulating evidence suggest importance
of the blood–brain barrier (BBB) permeability
[55], the
NMDA receptors
[56], bradykinin [57], the matrix
metalloproteases (MMPs) of a family of proteases
[58],
aquaporin 4 (AQP4) of a family of membrane water-channel
proteins
[59,60] and neuropeptides (such as substance P [61,62])
in TBI-elicited brain oedema development.
Treatments used in clinical management of post-traumatic
brain oedema include mannitol
[63], hypertonic saline
(HTS)
[64] and steroids among others [65]. The two main
mechanisms of mannitol’s action are osmotic and haemo-
dynamic. As mannitol does not cross the intact BBB, it estab-
lishes a concentration gradient across the BBB forcing the
movement of water from the oedematous brain to the intra-
vascular space; this process is followed by fast renal excretion
of mannitol and water. The haemodynamic effects of manni-
tol are based on a reduction in blood viscosity, which conse-
quently leads to increased cerebral blood flow and decrease in
cerebral blood volume via passive vasoconstriction
[65].
Although mannitol 20% is routinely used to reduce brain
bulk and intracranial pressure, its use has never been subjected
to a randomised, placebo-controlled trial analysing its effects
on post-TBI outcome. It is also noteworthy that mannitol
causes diuresis leading to secondary effects (such as systemic
hypovolaemia and hypotension) and adverse changes in serum
and urinary Na
+
, K
+
and osmolarity, which can be deleterious
to the brain-injured patient.
Hypertonic saline solutions have been suggested as an alter-
native to mannitol for the treatment of brain oedema
[65]. It
has been demonstrated that through multiple and probably
interacting mechanisms (such as optimisation of systemic and
cerebral haemodynamics, modulation of cerebral vasospasm
and changes in neuroendocrine-immune parameters
[64])
HTSs reduce brain oedema and ICP. The adverse effects of
HTS are similar to other osmotic agents: osmotic demyelina-
tion syndrome, renal insufficiency and rebound intracranial
hypertension
[64]. Despite promising clinical data on hyper-
tonic saline
[66,67], controversy remains about the role of HTS
in the treatment of TBI-induced oedema. The results of a
larger multi-centre trial
[68] conducted in Australia showed no
improved neurological outcome in patients with severe brain
trauma and hypotension who received hypertonic saline for
pre-hospital resuscitation compared with conventional fluid
management. Further experimental and clinical studies are
necessary to fully define the precise mechanisms of actions,
optimise administration regimens and confirm the role of
HTS in brain oedema management.
As glucoroticoids (GCs) have been shown to ameliorate
vasogenic oedema accompanying tumour, inflammatory
conditions and other disorders linked to increased BBB
permeability
[69], they were incorporated as a standard part of
TBI-induced brain oedema management over the course of
40 years. Interestingly, numerous clinical trials conducted dur-
ing this period of time showed no proof of steroid efficacy in
improving post-traumatic outcome in patients with severe
head injury
[70]. The recent MRC CRASH (Corticosteroid
Randomisation After Significant Head injury) international
standard randomised controlled trial evaluated the effect of
corticosteroids on death and disability after head injury in
10,008 randomly allocated adults. Data at 6 months showed a
significantly increased risk of death in the GC than in the
placebo group and no evidence that the effect of GCs changed
with injury severity or time since injury
[8]. These results
further support the recommendation that GCs should not be
used for reducing ICP or improving outcome after severe TBI.
The broad array of recent experimental studies suggests
novel therapeutic possibilities in preventing and/or reducing
brain oedema after TBI: inhibition of the complement
cascade by N-acetylheparin in thrombin-induced brain dam-
age
[71], modulation of AQP-4 channels by magnesium [31,72]
or blocking bradykinin B
2
receptor [73] among others. Con-
tinuing research is needed to enhance the understanding of
the pathobiology of cerebral oedema and identify new
approaches against TBI-induced brain oedema.
2.6 Progesterone
Numerous experimental studies using various models of TBI
demonstrated that progesterone, a neurosteroid that is natu-
rally found in the brains of both men and women, has signifi-
cant neuroprotective properties
[74]. Progesterone confers its
neuroprotective action most possibly via multiple mecha-
nisms
[75]. This hormone has been shown to reduce neuronal
injury caused by glutamate, FeSO
4
and Aβ toxicity; decrease
post-traumatic brain oedema
[76] and anxiety [77]; reverse the
early postinjury dysfunction in mitochondrial respiration and
reduce hippocampal neuronal loss after TBI
[78]; has anti-
oxidant effects
[79]; and attenuates mechanisms of apoptotic
cell death
[80] and so on.
A recent study
[81] has shown that acute TBI per se does not
induce an endogenous release of progesterone and that stable
progesterone concentrations can be rapidly achieved by
continuous intravenous infusion in patients with acute to
severe TBI. Pharmacokinetic alterations observed in pro-
gesterone levels after TBI were not gender dependent
[81].
These results demonstrate a feasibility of progesterone treat-
ment in TBI patients. ProTECT (Progesterone for Traumatic
Brain Injury, Experimental Clinical Treatment) was a sin-
gle-centre, Phase II, double-blind, randomised, placebo-con-
trolled trial organised by Emory University to analyse the
effects of progesterone treatment on the outcome of moderate
and severe blunt TBI. The study started in May 2002, ended
in January 2006 and included patients of both genders, of
18 years of age with moderate to severe blunt head trauma
(GCS: 4 – 12) that occurred within 11 h. The preliminary
results seem to be promising.
Recent advances in neuroprotection for treating traumatic brain injury
1376 Expert Opin. Investig. Drugs (2006) 15(11)
3. Experimental promises
3.1 Neurotransplantation
As the brain has limited capacity for self repair, increasing
research efforts address the possibility of reducing func-
tional deficits caused by brain injury or disease by trans-
planting new cells or manipulating endogenous progenitor
cells that can replace those with irreversible damage
[82].
Tissue derived from the fetal CNS has been broadly evalu-
ated as a material for transplantation due to its pre-
disposition for long-term survival and ability to regain full
functionality
[83]; However, alternative sources of human
neurons for the purpose of transplantation have been devel-
oped, which include immortalised cell lines and cultured
stem cells
[84] because of ethical restrictions, lack of availa-
bility and immunological limitations. Postmitotic human
neurons (NT2N cells) derived from the human embryonal
tetracarcinoma (NT2 cell) line have demonstrated many of
the vital characteristics of normal developing and mature
human neurons
[85]. After the satisfying safety trial, Kondzi-
olka et al.
[86] implanted the NT2N cells stereotactically
into the brain of patients with motor deficits caused by
stroke. Despite the lack of a significant benefit in motor
function as determined by the primary efficacy measure
(i.e., European Stroke Scale at 6 months post-treatment), a
considerable improvement was observed in some patients
that manifested in improved quality of life. Stem cells are
undifferentiated cells that lack antigen markers typical of
mature cells and display significant proliferation potential,
whereas progenitor cells have a limited capacity for self
renewal and a more restricted lineage potential than that of
stem cell
[82]. Ideally, the transplanted cells should be able to
generate most or all subtypes of neurons and glial cells; cur-
rently, the embryonic stem cells are the best characterised
cells with such a feature
[87]. Although neurotransplantation
has great therapeutic potential, restoration of brain func-
tion after TBI requires more than just cellular replacement;
further knowledge on graft function, transplant-induced
molecular changes underlying behavioural recovery and
influence of environmental factors are necessary to achieve
a clinically relevant therapy for TBI. Currently, the Uni-
versity of Texas Health Science Center is conducting a
Phase I clinical trial aiming to determine if bone marrow
harvest-containing mesenchymal stem cells and haemato-
poietic stem cells, bone marrow precursor cell separation
and re-infusion are safe in children after severe TBI. The
study started in April 2006 and includes children of
5 – 14 years of age on the day of injury, with TBI occurring
< 24 h prior to injury and with a GCS of 5 – 8. Using chil-
dren as the primary population is justified by facts that chil-
dren are more likely to have isolated TBI that is more
diffuse and less likely to be secondary to extra-axial fluid
collections and that children have a greater neurological
plasticity compared with adults.
3.2 Cell-cycle inhibition
Upregulation of cell-cycle proteins has been shown in both
mitotic and postmitotic neural cells after brain injury in adult
animals. They induce proliferation in mitotic cells (such as
astroglia and microglia) but they stimulate caspase-dependent
apoptosis in postmitotic cells (such as neurons)
[88]. Recent
studies showed that early central administration of the cell-cycle
inhibitor flavopiridol after experimental TBI significantly
reduced lesion volume, scar formation and neuronal cell death
as well as promoting near complete behavioural recovery
[89].
Moreover, cell-cycle protein upregulation has been significantly
reduced, etoposide-induced neuronal cell death attenuated and
astrocyte proliferation decreased by using structurally different
cell-cycle inhibitors (flavopiridol, roscovitine and olomoucine)
in primary neuronal or astrocyte cultures. Flavopiridol also
reduced proliferation/activation of microglia in a con-
centration-dependent manner. It is noteworthy that central
administration of flavopiridol has been successful in improving
functional outcome in a dose-dependent manner after fluid
percussion induced brain injury in rats, whereas delayed sys-
temic administration of flavopiridol significantly decreased
brain lesion volume and attenuated oedema development after
TBI
[90]. Accumulating information [91] provides further sup-
port for the therapeutic potential of cell-cycle inhibitors for the
treatment of TBI, suggesting reduction of neuronal cell death,
inhibition of glial proliferation and attenuation of microglial
activation as the most likely protective mechanisms.
3.3 Small neuropeptides
Ample evidence shows neuroprotective effects of the tri-
peptide thyrotropin-releasing hormone (TRH) across multi-
ple experimental models of TBI
[92]. On the basis of the active
metabolic product of TRH, cyclo-His-Pro, a number of novel
cyclic dipeptides have been developed out of which four com-
pounds were found to reduce cell death after trophic with-
drawal or traumatic injury in primary neuronal cultures, and
two of these protected against glutamate or β-amyloid neuro-
toxicity
[93]. Interestingly, all of the compounds significantly
improved functional (motor and cognitive) outcome after
controlled cortical impact injury in mice and reduced brain
lesion volumes. The mechanisms of these small neuropeptides
are still not fully clarified. As a dipeptide, it would be
expected that these compounds confer their effects via recep-
tor system(s); however, the receptor-binding studies did not
show significant receptor binding of the prototype 35b to 50
classical receptors, channels or transporters
[93]. The mecha-
nisms suggested
[93] include attenuation of both apoptotic and
necrotic cell death pathways, reduction of intracellular Ca
2+
accumulation after injury, stabilisation of mitochondrial
membrane potential and a decrease in cytochrome c release.
Additional data have been presented showing that compound
35b, which is being developed for a clinical trial, decreased
injury-induced transcriptional changes for a number of genes
(and proteins) such as cell-cycle genes, aquaporins and
Cernak
Expert Opin. Investig. Drugs (2006) 15(11) 1377
cathepsins [94]. The fact that small neuropeptides upregulate
multiple endogenous neuroprotective factors and reduce
induction of multiple classic secondary injury mechanisms
suggests that they may have potential usefulness in clinical
head injury. Nevertheless, there is still a possibility that these
compounds confer their effects through at least one receptor
system; such an unidentified receptor–drug interaction could
cause multiple side effects and hence diminish the value of
this therapeutic approach for clinical use.
3.4 Anti-inflammatory strategies
Neuroinflammation has been shown to play a significant role in
the pathobiology of TBI. This complex process includes intricate
interactions between cytokines/chemokines, metalloproteases,
NO and the arachidonic acid cascade among others
[95,96].
Indeed, inflammatory mediators (such as IL-1, IL-6 and
TNF-α) have been demonstrated to play important roles in
delayed CNS damage. Moreover, administration of a COX-2
inhibitor after TBI improved functional outcome in rats suggest-
ing the importance of eicosanoids in post-traumatic neurological
deficits
[97,98]. Injury-induced neuronal cell loss was reduced
using a C5a receptor antagonist, whereas secondary injury mech-
anisms after TBI were attenuated by the lack of C3 or C5 com-
plement
[99]. Furthermore, neuropeptide (substance P) release
has been shown to significantly contribute to TBI-induced
neurogenic inflammation
[61,100], whereas substance P antagonist
significantly improved functional outcome in experimental ani-
mals with TBI
[62]. These findings suggest that factors of neuro-
genic inflammation are promising targets for the development of
novel and effective therapeutics for TBI.
4. Conclusion
It has now been accepted that TBI results in a multifactorial
secondary injury cascade that leads to neuronal cell death and
related long-term neurological deficits. Neuroprotective strat-
egies face challenge to identify and simultaneously target the
most important mechanisms involved in the secondary injury
cascade. Hence the most recent therapeutic candidates are
multifactorial in nature. These compounds show a greater
potential to become a ‘magic bullet’ drug compared with a
combination of drugs. The deficiencies of polypharmacy can
be explained by undesirable interactions of the individual
drugs and that multiple drugs have multiple side effects.
Current neuroscience applications concerning TBI are
focused on basic molecular and cellular mechanisms, some-
times without necessary consideration of their potential clinical
implications or analysing the time-line for possible translation
of the gained knowledge into clinical arena. Hence, therapeutic
approaches in early- and even late-stage clinical trials are often
repetitions of previous ones. There is an awareness of mistakes
in clinical design and data analysis, which could underlie nega-
tive outcome of TBI-related clinical trials; the new, higher
stringency design suggested by many scientists should increase
the reliability of information given by clinical trials.
5. Expert opinion
Despite immense research efforts and persisting enthusiasm,
the magic cocktail that would prevent delayed brain damage
and/or replace or regenerate the irreversibly injured cells after
TBI has yet to be found. Although knowledge on the patho-
biology of TBI (including mechanisms of neuronal cell
death, neurogenic inflammation and brain oedema and so
on) are progressively increasing, the quest for the Holy Grail
of neuroprotective therapies that would offer relief to an
enormous population of people suffering from the con-
sequences of TBI is still unsuccessful. There is an obvious
imbalance between extraordinary intellectual potential
invested into this research generating a vast amount of infor-
mation concerning brain injury and its therapy, and the lack
of success of clinical trials evaluating neuroprotective
strategies in TBI. Such a discrepancy suggests a need for ‘out
of the box’ thinking and fundamentally new approaches in
designing neuroprotective strategies.
Nanotechnology
[101] and molecular self assembly applied
to repair injured neuronal structures might offer such an
approach. Ellis-Behnke
[102] used a synthetic nanomaterial to
establish a permissive environment for axonal regrowth
in vivo. This impressive self-assembling protein created a
scaffold-like tissue-bridging structure and provided a frame-
work for partial axonal re-innervation with regenerative
potential both in young and adult animals. Moreover,
polymethylmethacrylate-covered silicon chips have also been
used as a nerve-regenerating scaffold with encouraging results
[103]. Carbon nanotubes made of rolled layers of graphite
have also been shown as potential devices that are able to
improve neural signal transfer by supporting dendrite elonga-
tion and cell adhesion
[104]. Design of functionalised nano-
particles that can be administered systemically and improve
drug delivery across the BBB is another clinically relevant
area of nanotechnology
[101]. Neuropeptides (such as
enkephalins) and the NMDA receptor antagonist
MRZ 2/576 have been absorbed onto the surface of
poly(butylcyanoacrylate) nanoparticles with polysorbate 80
coating; this coating adsorbs apolipoproteins B and -E from
the blood, which enables the uptake of nanoparticles by brain
capillary endothelial cells through receptor-mediated endo-
cytosis
[105]. There are numerous challenges related to the use
of nanotechnology in neuroscience. Structural complexity
and heterogeneity, the highly anatomically restrictive nature
of the CNS and the extremely intricate cellular interactions
cause a unique set of obstacles
[101]. Nevertheless, the applica-
tions of nanotechnology enable physical interaction with
neural cells at cellular and subcellular levels, with a potential
to establish functional interaction at a systemic level and
facilitate direct physiological effects. Taking together, nano-
technology offers enormous opportunities for further under-
standing of the pathobiology of TBI and (as such) might
represent one of the ‘out of the box’ research directions for
developing novel therapies.
Recent advances in neuroprotection for treating traumatic brain injury
1378 Expert Opin. Investig. Drugs (2006) 15(11)
Bibliography
1. BRUNS J Jr, HAUSER WA: The
epidemiology of traumatic brain injury:
areview. Epilepsia (2003)
44(Suppl. 10):2-10.
2. CENTER FOR DISEASE CONTROL
AND PREVENTION: Incidence rates of
hospitalization related to traumatic brain
injury-12 states, 2002. MMWR Morb.
Mortal. Wkly Rep. (2006) 55(8):201-204.
3. TOLIAS CM, BULLOCK MR: Critical
appraisal of neuroprotection trials in head
injury: what have we learned? NeuroRx
(2004) 1(1):71-79.
4. VINK R, VAN DEN HEUVEL C:
Recent advances in the development of
multifactorial therapies for the treatment of
traumatic brain injury. Expert Opin.
Investig. Drugs (2004) 13(10):1263-1274.
5. CERNAK I: Animal models of head
trauma. NeuroRx (2005) 2(3):410-422.
•• Comprehensive review of the experimental
models of TBI, describing the pros and
cons of particular animal models.
6. FADEN AI: Neuroprotection and traumatic
brain injury: the search continues.
Arch. Neurol. (2001) 58(10):1553-1555.
7. NARAYAN RK, MICHEL ME,
ANSELL B et al.: Clinical trials in head
injury. J. Neurotrauma (2002)
19(5):503-557.
8. EDWARDS P, ARANGO M, BALICA L
et al.: Final results of MRC CRASH, a
randomised placebo-controlled trial of
intravenous corticosteroid in adults with
head injury-outcomes at 6 months. Lancet
(2005) 365(9475):1957-1959.
9. ROBERTS I: Barbiturates for acute
traumatic brain injury. Cochrane Database
Syst. Rev. (2000) 2:CD000033.
10. MORRIS SH, KNEVETT S,
LERNER EG, BINDMAN LJ: Group I
mGluR agonist DHPG facilitates the
induction of LTP in rat prelimbic cortex
in vitro. J. Neurophysiol. (1999)
82(4):1927-1933.
11. YOUNG B, RUNGE JW, WAXMAN KS
et al.: Effects of pegorgotein on neurologic
outcome of patients with severe head injury.
A multicenter, randomized controlled trial.
JAMA (1996) 276(7):538-543.
12. THE EUROPEAN STUDY GROUP ON
NIMODIPINE IN SEVERE HEAD
INJURY: A multicenter trial of the efficacy
of nimodipine on outcome after severe head
injury. J. Neurosurg. (1994) 80(5):797-804.
13. LANGHAM J, GOLDFRAD C,
TEASDALE G, SHAW D, ROWAN K:
Calcium channel blockers for acute
traumatic brain injury. Cochrane Database
Syst. Rev. (2003) 4:CD000565.
14. BIEGON A, JOSEPH AB: Development of
HU-211 as a neuroprotectant for ischemic
brain damage. Neurol. Res. (1995)
17(4):275-280.
15. BAYIR H, CLARK RS, KOCHANEK PM:
Promising strategies to minimize
secondary brain injury after head trauma.
Crit. Care Med. (2003)
31(Suppl. 1):S112-S117.
16. SHOHAMI E, NOVIKOV M, BASS R:
Long-term effect of HU-211, a novel
non-competitive NMDA antagonist, on
motor and memory functions after closed
head injury in the rat. Brain Res. (1995)
674(1):55-62.
17. SHOHAMI E, GALLILY R,
MECHOULAM R, BASS R,
BEN-HUR T: Cytokine production in the
brain following closed head injury:
dexanabinol (HU-211) is a novel TNF-α
inhibitor and an effective neuroprotectant.
J. Neuroimmunol. (1997) 72(2):169-177.
18. KNOLLER N, LEVI L, SHOSHAN I
et al.: Dexanabinol (HU-211) in the
treatment of severe closed head injury: a
randomized, placebo-controlled, Phase II
clinical trial. Crit. Care Med. (2002)
30(3):584-554.
19. MAAS A, MURRAY G, HENNEY HI
et al.: Efficacy and safety of dexanabinol in
severe traumatic brain injury: results of
Phase III randomised, placebo-controlled,
clinical trial. Lancet Neurol. (2006) 5:38-45.
20. VINK R, CERNAK I: Regulation of
intracellular free magnesium in central
nervous system injury. Front. Biosci. (2000)
5:D656-D665.
21. VINK R, O’CONNOR CA, NIMMO AJ,
HEATH DL: Magnesium attenuates
persistent functional deficits following
diffuse traumatic brain injury in rats.
Neurosci. Lett. (2003) 336(1):41-44.
22. HEATH DL, VINK R: Magnesium
sulphate improves neurologic outcome
following severe closed head injury in rats.
Neurosci. Lett. (1997) 228(3):175-178.
23. MUIR KW: New experimental and clinical
data on the efficacy of pharmacological
magnesium infusions in cerebral infarcts.
Magnes. Res. (1998) 11(1):43-56.
24. MAUSKOP A, ALTURA BT,
CRACCO RQ, ALTURA BM: Intravenous
magnesium sulfate rapidly alleviates
headaches of various types. Headache (1996)
36(3):154-160.
25. MUIR KW, LEES KR, FORD I, DAVIS S,
INTRAVENOUS MAGNESIUM
EFFICACY IN STROKE (IMAGES)
STUDY INVESTIGATORS: Magnesium
for acute stroke (Intravenous Magnesium
Efficacy in Stroke trial): randomized
controlled trial. Lancet (2004)
36(9407):439-435.
26. BOET R, MEE E: Magnesium sulfate in
the management of patients with Fisher
Grade 3 subarachnoid hemorrhage: a pilot
study. Neurosurgery (2000) 47(3):602-606
(Discussion 606-607).
27. VEYNA RS, SEYFRIED D, BURKE DG
et al.: Magnesium sulfate therapy after
aneurysmal subarachnoid hemorrhage.
J. Neurosurg. (2002) 96(3):510-514.
28. McKEE JA, BREWER RP, MACY GE
et al.: Magnesium neuroprotection is
limited in humans with acute brain injury.
Neurocrit. Care (2005) 2(3):342-351.
29. CHAN MT, BOET R, NG SC,
POON WS, GIN T: Magnesium sulfate for
brain protection during temporary cerebral
artery occlusion. Acta Neurochir. Suppl.
(2005) 95:107-111.
30. VAN DEN BERGH WM, ALGRA A,
VAN KOOTEN F, RINKEL GJ,
MASH STUDY GROUP: Magnesium
sulfate in aneurysmal subarachnoid
hemorrhage: a randomized controlled trial.
Stroke (2005) 36(5):1011-1015.
31. GHABRIEL MN, THOMAS A, VINK R:
Magnesium restores altered aquaporin-4
immunoreactivity following traumatic
brain injury to a pre-injury state.
Acta Neurochir. Suppl. (2006) 96:402-406.
32. HOANE MR: Treatment with magnesium
improves reference memory while reducing
GFAP expression following traumatic brain
injury. J. Neurotrauma (2005)
22(7):783-792.
33. TEASDALE GM, MAAS A,
IANNOTTI F, OHMAN J,
UNTERBERG A: Challenges in translating
the efficacy of neuroprotective agents in
experimental models into knowledge of
clinical benefits in head injured patients.
Acta Neurochir. Suppl. (Wien) (1999)
73:111-116.
34. MAAS AI: Neuroprotective agents in
traumatic brain injury. Expert Opin.
Investig. Drugs (2001) 10(4):753-767.
Cernak
Expert Opin. Investig. Drugs (2006) 15(11) 1379
35. O’CONNOR CA, CERNAK I, VINK R:
Interaction between anesthesia, gender, and
functional outcome task following diffuse
traumatic brain injury in rats.
J. Neurotrauma (2003) 20(6):533-541.
36. DOPPENBERG EM, CHOI SC,
BULLOCK R: Clinical trials in traumatic
brain injury. What can we learn from
previous studies? Ann. NY Acad. Sci. (1997)
825:305-322.
37. FADEN AI: Neuroprotection and traumatic
brain injury: theoretical option or realistic
proposition. Curr. Opin. Neurol. (2002)
15(6):707-712.
38. ZHAO H, YENARI MA, SAPOLSKY RM,
STEINBERG GK: Mild postischemic
hypothermia prolongs the time window for
gene therapy by inhibiting cytochrome C
release. Stroke (2004) 35(2):572-577.
39. INAMASU J, SUGA S, SATO S et al.:
Postischemic hypothermia attenuates
apoptotic cell death in transient focal
ischemia in rats. Acta Neurochir. Suppl.
(2000) 76:525-527.
40. HASHIGUCHI N, SHIOZAKI T,
OGURA H et al.: Mild hypothermia
reduces expression of heat shock protein 60
in leukocytes from severely head-injured
patients. J. Trauma (2003)
55(6):1054-1060.
41. ADELSON PD, RAGHEB J,
MUIZELAAR JP et al.: Phase II clinical
trial of moderate hypothermia after severe
traumatic brain injury in children.
Neurosurgery (2004) 56:740-754.
42. SAHUQUILLO J, ARIKAN F:
Decompressive craniectomy for the
treatment of refractory high intracranial
pressure in traumatic brain injury.
Cochrane Database Syst. Rev. (2006)
1:CD003983.
•• Detailed analysis of accumulated
experience, success and failure of
decompressive craniectomy for the
treatment of TBI.
43. CLIFTON GL, MILLER ER, CHOI SC
et al.: Lack of effect of induction of
hypothermia after acute brain injury.
N. Engl. J. Med. (2001) 344(8):556-563.
44. SEPPELT I: Hypothermia does not improve
outcome from traumatic brain injury.
Crit. Care Resusc. (2005) 7(3):233-237.
45. SIGNORINI DF, ANDREWS PJ,
JONES PA, WARDLAW JM, MILLER JD:
Adding insult to injury: the prognostic
value of early secondary insults for survival
after traumatic brain injury. J. Neurol.
Neurosurg. Psychiatry (1999) 66(1):26-31.
46. CZOSNYKA M, GUAZZO E,
WHITEHOUSE M et al.: Significance of
intracranial pressure waveform analysis after
head injury. Acta Neurochir. (Wien) (1996)
138(5):531-541 (Discussion 541-532).
47. JOURDAN C, CONVERT J,
MOTTOLESE C et al.: Evaluation of the
clinical benefit of decompression
hemicraniectomy in intracranial
hypertension not controlled by medical
treatment. Neurochirurgie (1993)
39(5):304-310.
48. BRAIN TRAUMA FOUNDATION:
Guidelines for the Management of Severe
Head Injury. Brain Trauma Foundation,
New York, USA (1995).
49. COOPER PR, ROVIT RL,
RANSOHOFF J: Hemicraniectomy in the
treatment of acute subdural hematoma:
a re-appraisal hemicraniectomy in the
treatment of acute subdural hematoma.
Surg. Neurol. (1976) 5:25-28.
50. MAKINO H, YAMAURA A: Assessment of
outcome following large decompressive
craniectomy in management of serious
cerebral contusion. A review of 207 cases.
Acta Neurochir. Suppl. (1976)
28(1):193-194.
51. TAYLOR A, BUTT W, ROSENFELD J
et al.: A randomized trial of very early
decompressive craniectomy in children with
traumatic brain injury and sustained
intracranial hypertension. Childs Nerv. Syst.
(2001) 17(3):154-162.
52. BERGER MS, PITTS LH, LOVELY M,
EDWARDS MS, BARTKOWSKI HM:
Outcome from severe head injury in
children and adolescents. J. Neurosurg.
(1985) 62(2):194-199.
53. FELDERHOFF-MUESER U,
IKONOMIDOU C: Mechanisms of
neurodegeneration after paediatric brain
injury. Curr. Opin. Neurol. (2000)
13(2):141-145.
54. KIMELBERG HK: Current concepts of
brain edema. Review of laboratory
investigations. J. Neurosurg. (1995)
83(6):1051-1059.
55. JOO F, KLATZO I: Role of cerebral
endothelium in brain oedema. Neurol. Res.
(1989) 11(2):67-75.
56. DEMPSEY RJ, BASKAYA MK,
DOGAN A: Attenuation of brain edema,
blood–brain barrier breakdown, and injury
volume by ifenprodil, a polyamine-site
N-methyl-
D-aspartate receptor antagonist,
after experimental traumatic brain injury in
rats. Neurosurgery (2000) 47(2):399-404.
57. ZAUSINGER S, LUMENTA DB,
PRUNEAU D et al.: Therapeutical
efficacy of novel non-peptide
bradykinin B
2
receptor antagonist on
brain edema formation and ischemic
tissue damage in focal cerebral ischemia.
Acta Neurochir. Suppl. (2003) 86:205-207.
58. SHIGEMORI Y, KATAYAMA Y, MORI T,
MAEDA T, KAWAMATA T: Matrix
metalloproteinase-9 is associated with
blood-brain barrier opening and brain
edema formation after cortical contusion in
rats. Acta Neurochi. Suppl. (2006)
96:130-133.
59. AMIRY-MOGHADDAM M,
OTSUKA T, HURN PD et al.:
An α-syntrophin-dependent pool of AQP4
in astroglial end-feet confers bidirectional
water flow between blood and brain.
Proc. Natl. Acad. Sci. USA (2003)
100(4):2106-2111.
60. KLEINDIENST A, FAZZINA G,
AMORINI AM et al.: Modulation of
AQP4 expression by the protein kinase C
activator, phorbol myristate acetate,
decreaes ischemia-induced brain edema.
Acta Neurochir. Suppl. (2006) 96:393-397.
61. NIMMO AJ, CERNAK I, HEATH DL
et al.: Neurogenic inflammation is
associated with development of edema and
functional deficits following traumatic brain
injury in rats. Neuropeptides (2004)
38(1):40-47.
62. VINK R, DONKIN JJ, CRUZ MI,
NIMMO AJ, CERNAK I: A substance P
antagonist increases brain intracellular free
magnesium concentration after diffuse
traumatic brain injury in rats. J. Am.
Coll. Nutr. (2004) 23(5):538S-540S.
63. WAKAI A, ROBERTS I,
SCHIERHOUT G: Mannitol for acute
traumatic brain injury. Cochrane Database
Syst. Rev. (2005) 4:CD001049.
64. DOYLE JA, DAVIS DP, HOYT DB:
The use of hypertonic saline in the
treatment of traumatic brain injury.
J. Trauma (2001) 50(2):367-383.
65. RABINSTEIN AA: Treatment of cerebral
edema. Neurologist (2006) 12(2):59-73.
66. FRESHMAN SP, BATTISTELLA FD,
MATTEUCCI M, WISNER DH:
Hypertonic saline (7.5%) versus mannitol: a
comparison for treatment of acute head
injuries. J. Trauma (1993) 35(3):344-348.
Recent advances in neuroprotection for treating traumatic brain injury
1380 Expert Opin. Investig. Drugs (2006) 15(11)
67. PINTO FC, CAPONE-NETO A,
PRIST R, E SILVA MR,
POLI-DE-FIGUEIREDO LF:
Volume replacement with lactated Ringers
or 3% hypertonic saline solution during
combined experimental hemorrhagic shock
and traumatic brain injury. J. Trauma
(2006) 60(4):758-763
(Discussion 763-754).
68. COOPER DJ, MYLES PS,
McDERMOTT FT et al.: Prehospital
hypertonic saline resuscitation of patients
with hypotension and severe traumatic
brain injury: a randomized controlled trial.
JAMA (2004) 291(11):1350-1357.
69. FRENCH LA, GALICICH JH: The use of
steroids for control of cerebral edema.
Clin. Neurosurg. (1964) 10:212-223.
70. GHAJAR J, HESDORFFER DC: Steroids
CRASH out of head-injury treatment.
Lancet Neurol. (2004) 3(12):708.
71. GONG Y, XI GH, KEEP RF,
HOFF JT, HUA Y: Complement
inhibition attenuates brain edema and
neurological deficits induced by thrombin.
Acta Neurochir. Suppl. (2005) 95:389-392.
72. WANG F, FENG XC, LI YM, YANG H,
MA TH: Aquaporins as potential drug
targets. Acta Pharmacol. Sin. (2006)
27(4):395-340.
73. KLASNER B, LUMENTA DB,
PRUNEAU D, ZAUSINGER S,
PLESNILA N: Therapeutic window of
bradykinin B
2
receptor inhibition after focal
cerebral ischemia in rats. Neurochem. Int.
(2006) 49(5):442-447.
74. SINGH M: Progesterone-induced
neuroprotection. Endocrine (2006)
29(2):271-274.
75. STEIN DG: The case for progesterone.
Ann. NY Acad. Sci. (2005) 1052:152-169.
76. O’CONNOR CA, CERNAK I, VINK R:
Both estrogen and progesterone attenuate
edema formation following diffuse
traumatic brain injury in rats. Brain Res.
(2005) 1062(1-2):171-174.
77. CUTLER SM, VANLANDINGHAM JW,
MURPHY AZ, STEIN DG: Slow-release
and injected progesterone treatments
enhance acute recovery after traumatic brain
injury. Pharmacol. Biochem. Behav. (2006)
24:24.
78. ROBERTSON CL, PUSKAR A,
HOFFMAN GE et al.: Physiologic
progesterone reduces mitochondrial
dysfunction and hippocampal cell loss
after traumatic brain injury in female rats.
Exp. Neurol. (2006) 197(1):235-243.
79. ROOF RL, HOFFMAN SW, STEIN DG:
Progesterone protects against lipid
peroxidation following traumatic brain
injury in rats. Mol. Chem. Neuropathol.
(1997) 31(1):1-11.
80. DJEBAILI M, HOFFMAN SW,
STEIN DG: Allopregnanolone and
progesterone decrease cell death and
cognitive deficits after a contusion of the rat
pre-frontal cortex. Neuroscience (2004)
123(2):349-359.
81. WRIGHT DW, RITCHIE JC,
MULLINS RE, KELLERMANN AL,
DENSON DD: Steady-state serum
concentrations of progesterone following
continuous intravenous infusion in patients
with acute moderate to severe traumatic
brain injury. J. Clin. Pharmacol. (2005)
45(6):640-648.
82. SHOUTEN JW, FULP CT, ROYO NC
et al.: A review and rationale for the use of
cellular transplantation as a therapeutic
strategy for traumatic brain injury.
J. Neurotrauma (2004) 21(11):1501-1538.
•• Explanation of molecular and physiological
concepts of neurotransplantation, and
the challenges and benefits related to
this technique.
83. JOHANSSON B, GRABOWSKI M:
Functional recovery after brain infarction:
plasticity and neural transplantation.
Brain Pathol. (1994) 4:85-95.
84. BARINAGA M: Fetal neuron grafts pave
the way for stem cell therapies. Science
(2000) 287:1421-1422.
85. TROJANOSWKI JQ, KLEPPNER SR,
HARTLEY RS et al.: Transfectable and
transplantable postmitotic human neurons:
a potential ‘platform’ for gene therapy of
nervous system diseases. Exp. Neurol. (1997)
144(1):92-97.
86. KONDZIOLKA D, STEINBERG GK,
WECHSLER L et al.: Neurotransplantation
for patients with subcortical motor stroke: a
Phase II randomized trial. J. Neurosurg.
(2005) 10:38-45.
87. ZHANG S-C: Embryonic stem cells for
neural replacement therapy: prospects and
challenges. J. Hematother. Stem Cell Res.
(2003) 12:625-634.
88. NGUYEN MD, MUSHYNSKI WE,
JULIEN JP: Cycling at the interface
between neurodevelopment and
neurodegeneration. Cell Death Differ.
(2002) 9(12):1294-1306.
89. DI GIOVANNI S, MOVSESYAN V,
AHMED F et al.: Cell cycle inhibition
provides neuroprotection and reduces glial
proliferation and scar formation after
traumatic brain injury. Proc. Natl. Acad.
Sci. USA (2005) 102(23):8333-8338.
New perspectives of cell-cycle inhibition in
preventing or attenuating neuronal cell
death and scar formation following TBI.
90. CERNAK I, STOICA B, BYRNES KR,
DI GIOVANNI S, FADEN AI: Role of the
cell cycle in the pathobiology of central
nervous system trauma. Cell Cycle (2005)
4(9):1286-1293.
91. VERDAGUER E, JORDA EG,
ALVIRA D et al.: Inhibition of multiple
pathways accounts for the antiapoptotic
effects of flavopiridol on potassium
withdrawal-induced apoptosis in neurons.
J. Mol. Neurosci. (2005) 26(1):71-84.
92. FADEN AI: Neuropeptides and central
nervous system injury. Clinical
implications. Arch. Neurol. (1986)
43(5):501-504.
93. FADEN AI, KNOBLACH SM,
CERNAK I et al.: Novel diketopiperazine
enhances motor and cognitive recovery after
traumatic brain injury in rats and shows
neuroprotection in vitro and in vivo.
J. Cereb. Blood Flow Metab. (2003)
23(3):342-354.
94. FADEN AI, KNOBLACH SM,
MOVSESYAN VA, LEA PMT,
CERNAK I: Novel neuroprotective
tripeptides and dipeptides. Ann. NY
Acad. Sci. (2005) 1053:472-481.
95. MINAMI M, KATAYAMA T, SATOH M:
Brain cytokines and chemokines: roles in
ischemic injury and pain. J. Pharmacol. Sci.
(2006) 100(5):461-470.
96. LUCAS SM, ROTHWELL NJ,
GIBSON RM: The role of inflammation in
CNS injury and disease. Br. J. Pharmacol.
(2006) 147(Suppl. 1):S232-S240.
97. CERNAK I, O’CONNOR C, VINK R:
Inhibition of cyclooxygenase 2 by
nimesulide improves cognitive outcome
more than motor outcome following
diffuse traumatic brain injury in rats.
Exp. Brain Res. (2002) 147(2):193-199.
98. KAWANO T, ANRATHER J, ZHOU P
et al.: Prostaglandin E2 EP1 receptors:
downstream effectors of COX-2
neurotoxicity. Nat. Med. (2006)
12(2):225-229.
99. SEWELL DL, NACEWICZ B, LIU F et al.:
Complement C3 and C5 play critical roles
Cernak
Expert Opin. Investig. Drugs (2006) 15(11) 1381
in traumatic brain cryoinjury: blocking
effects on neutrophil extravasation by C5a
receptor antagonist. J. Neuroimmunol.
(2004) 155:55-63.
100. VINK R, YOUNG A, BENNETT CJ et al.:
Neuropeptide release influences brain
edema formation after diffuse traumatic
brain injury. Acta Neurochir. Suppl. (2003)
86:257-260.
101. SILVA GA: Neuroscience nanotechnology:
progress, opportunities and challenges.
Nat. Rev. Neurosci. (2006) 7(1):65-74.
•• Excellent overview explaining the basic
principles of nanotechnology applications
for neuroscience, progress of their
development, challenges and opportunities
they offer to neurotrauma research and
therapy of TBI patients.
102. ELLIS-BEHNKE RG, LIANG YX,
YOU SW et al.: Nano neuro knitting:
peptide nanofiber scaffold for brain repair
and axon regeneration with functional
return of vision. Proc. Natl. Acad. Sci. USA
(2006) 103(13):5054-5059.
Exciting possibilities of nanotechnology
application in designing and
generating nanofibres to support and guide
axonal regeneration.
103. JOHANSSON F, CARLBERG P,
DANIELSEN N, MONTELIUS L,
KANJE M: Axonal outgrowth on
nano-imprinted patterns. Biomaterials
(2006) 27(8):1251-1258.
104. LOVAT V, PANTAROTTO D,
LAGOSTENA L et al.: Carbon nanotube
substrates boost neuronal electrical
signaling. Nano Lett. (2005)
5(6):1107-1110.
105. KREUTER J: Influence of the surface
properties on nanoparticle-mediated
transport of drugs to the brain.
J. Nanosci. Nanotechnol. (2004)
4(5):484-488.
Website
201. http://www.ClinicalTrials.gov
Clinical trials website.
Affiliation
Ibolja Cernak MD PhD, Senior Professional Staff
Johns Hopkins University Applied Physics
Laboratory, Biomedicine Business Area,
11100 Johns Hopkins Road, Laurel,
MD 20723-6099, USA
Tel: +1 443 778 2637; Fax: +1 443 778 5889;
E-mail: ibolja.cernak@jhuapl.edu
... The cortical undercut model of PTE was developed to mimic penetrating cortical injuries and has been used in both in vivo and ex vivo studies. Cortical undercut has been performed on rats, mice, and cats of varying age (Graber and Prince, 1999;2006;Chauvette et al., 2016). Typically, rats are anesthetized and placed into a stereotaxic frame before unilateral exposure of the area of interest, typically the sensorimotor cortex. ...
... Attention should be paid to avoid any damage to major blood supplies. When a more complete undercut is needed, a second transcortical cut can be made with the needle without rotation (Graber and Prince, 1999;2006). After a maturation period, animals are sacrificed, and coronal slices are obtained for electrophysiologic recordings and histology. ...
... Furthermore, there are many differences in brain anatomy and complexity between small animals and humans. There are also some analogies, i.e., cerebrovascular parameters (Cernak, 2006), but the ratio of white:gray matter differs tremendously, making interpretation of behavioral alterations more challenging (Cordeiro and Horn, 2015). Furthermore, human TBI tends to be much more heterogenous in both injury location and cause (e.g., fall, car accident, sports injury, military events, etc.) than controlled laboratory experiments. ...
Article
Full-text available
Post-traumatic epilepsy (PTE) is one of the most devastating long-term, network consequences of traumatic brain injury (TBI). There is currently no approved treatment that can prevent onset of spontaneous seizures associated with brain injury, and many cases of PTE are refractory to antiseizure medications. Post-traumatic epileptogenesis is an enduring process by which a normal brain exhibits hypersynchronous excitability after a head injury incident. Understanding the neural networks and molecular pathologies involved in epileptogenesis are key to preventing its development or modifying disease progression. In this article, we describe a critical appraisal of the current state of PTE research with an emphasis on experimental models, molecular mechanisms of post-traumatic epileptogenesis, potential biomarkers, and the burden of PTE-associated comorbidities. The goal of epilepsy research is to identify new therapeutic strategies that can prevent PTE development or interrupt the epileptogenic process and relieve associated neuropsychiatric comorbidities. Therefore, we also describe current preclinical and clinical data on the treatment of PTE sequelae. Differences in injury patterns, latency period, and biomarkers are outlined in the context of animal model validation, pathophysiology, seizure frequency, and behavior. Improving TBI recovery and preventing seizure onset are complex and challenging tasks; however, much progress has been made within this decade demonstrating disease modifying, anti-inflammatory, and neuroprotective strategies, suggesting this goal is pragmatic. Our understanding of PTE is continuously evolving, and improved preclinical models allow for accelerated testing of critically needed novel therapeutic interventions in military and civilian persons at high risk for PTE and its devastating comorbidities. Significance Statement Post-traumatic epilepsy is a chronic seizure condition after brain injury. With few models and limited understanding of the underlying progression of epileptogenesis, progress is extremely slow to find a preventative treatment for PTE. This study reviews the current state of modeling, pathology, biomarkers, and potential interventions for PTE and comorbidities. There’s new optimism in finding a drug therapy for preventing PTE in people at risk, such as after traumatic brain injury, concussion, and serious brain injuries, especially in military persons.
... Participants' performance and responses may have differed from those of nonrespondents, and the stigma associated with mental health problem (15) may have resulted in reluctance to report negative feeling and behaviors. Although self-reported data are typically considered a limitation by researchers, reports indicate that the ASRS has adequate sensitivity (68.7%), excellent specificity (99.5%), excellent accuracy (97.9%), and good kappa scores (0.76) (27). Lastly, it should be pointed out that although measures were taken to minimize distractions, this study was not conducted in a controlled testing environment. ...
... As many as 1 in 10 soldiers deployed to combat experience symptoms indicative of mild brain trauma (2), and many who suffer a TBI experience long-term symptoms. Following the initial mechanical damage caused by a TBI, a chain reaction of biochemical events occurs leading to delayed neuron damage (27). The cellular changes that occur after a TBI are in many ways like an accelerated form of brain aging (28). ...
... Sullivan et al (24) attained a correlation of 0.54 between FFQ estimates of omega-3 long-chain PUFAs and plasma fatty acids, while Lucus et al (25) obtained a correlation of 0.42 between FFQ estimates of EPA + DHA and red blood cell membrane measurements of the corresponding fatty acids.The participants in the current study had lower omega-3 HUFA scores (16.9% ) compared to the 20.2 % reported by Metherel et al(26) for young men and women from Ontario who were consuming 0.22% of energy as omega-3 polyunsaturated fat. The omega-3 HUFA score and the FFQ-based intake of EPA + DHA in the present study were also lower than the 28.8% omega-3 HUFA score and the 350 mg/day (SD 508) food record estimate for EPA + DHA reported by Fratesi et al(27) for older adults. Noteworthy in the study by Fratesi et al ...
... Li et al. (2017a) and Sun et al. (2016) showed that the MIPS technique is more sensitive than ICP monitoring when detecting brain lesions, demonstrating that the MIPS technique can achieve early detection in real-time. However, for TBI-induced CCH the ICP cannot increase immediately because of the effect of cerebral autoregulation (Cernak, 2006;Di Ieva, Schmitz & Cusimano, 2013). Therefore, this study aimed to investigate whether the MIPS method can perform an early diagnosis of TBI-induced CCH. ...
Article
Full-text available
Closed cerebral hemorrhage (CCH) is a common symptom in traumatic brain injury (TBI) patients who suffer intracranial hemorrhage with the dura mater remaining intact. The diagnosis of CCH patients prior to hospitalization and in the early stage of the disease can help patients get earlier treatments that improve outcomes. In this study, a noncontact, portable system for early TBI-induced CCH detection was constructed that measures the magnetic induction phase shift (MIPS), which is associated with the mean brain conductivity caused by the ratio between the liquid (blood/CSF and the intracranial tissues) change. To evaluate the performance of this system, a rabbit CCH model with two severity levels was established based on the horizontal biological impactor BIM-II, whose feasibility was verified by computed tomography images of three sections and three serial slices. There were two groups involved in the experiments (group 1 with 10 TBI rabbits were simulated by hammer hit with air pressure of 600 kPa by BIM-II and group 2 with 10 TBI rabbits were simulated with 650 kPa). The MIPS values of the two groups were obtained within 30 min before and after injury. In group 1, the MIPS values showed a constant downward trend with a minimum value of-11.17 ± 2.91 at the 30th min after 600 kPa impact by BIM-II. After the 650 kPa impact, the MIPS values in group 2 showed a constant downward trend until the 25th min, with a minimum value of-16.81 ± 2.10. Unlike group 1, the MIPS values showed an upward trend after that point. Before the injury, the MIPS values in both group 1 and group 2 did not obviously change within the 30 min measurement. Using a support vector machine at the same time point after injury, the classification accuracy of the two types of severity was shown to be beyond 90%. Combined with CCH pathological mechanisms, this system can not only achieve the detection of early functional changes in CCH but can also distinguish different severities of CCH.
... Traumatic brain injury (TBI) is the most common neurological disease, and as such, has become a significant public health problem [1][2][3]. Primary injuries trigger complex cascades of secondary injury processes, which can persist for hours, days, and even months following an injury. ...
Article
Although store-operated calcium entry (SOCE) has been implicated in several neurological disorders, the exact mechanism for its role in traumatic brain injury (TBI) has not been elucidated. In this study, we found that TBI upregulated the expression of a calcium sensor protein called stromal interactive molecule 2 (STIM2); however, the levels of its homologue, STIM1, were unaffected. Both STIM1 and STIM2 are crucial components of SOCE, both in vivo and in vitro. Using shRNA, we discovered that downregulation of STIM2, but not STIM1, significantly improved neuronal survival in both an in vitro and in vivo model of TBI, decreasing neuronal apoptosis, and preserving neurological function. This neuroprotection was associated with alleviating TBI-induced calcium overload and preserving mitochondrial function. Additionally, downregulation of STIM2 not only inhibited Ca(2+) release from the endoplasmic reticulum (ER), but also reduced SOCE-mediated Ca(2+) influx, decreased mitochondrial Ca(2+), restored mitochondrial morphology and improved mitochondrial function, including MMP, ROS production and ATP synthesis. These results indicate that inhibition of STIM2 can protect neurons from TBI by inhibiting calcium overload and preserving mitochondrial function. This suggests that STIM2 might be an effective interventional target for TBI. Copyright © 2015. Published by Elsevier B.V.
... Presently, there are no approved treatments that can be administered after traumatic brain injury (TBI) to mitigate the progression of brain damage and improve functional outcome. During the past several decades, thousands of gene expression studies have provided much insight into the pathogenesis of TBI, and led to therapeutic strategies to inhibit genes and cellular pathways associated with cell death but these have not translated into effective treatments [1][2][3][4]; to date, hundreds of clinical trials of brain injury have failed to advance past Phase 2 trials [5,6]. This failure is reflected in the clinical management of TBI patients; once they are stabilized, there is little other than supportive care that can be done to limit the cascades of neuronal inflammation, oxidative stress and cell death [7][8][9]. ...
Article
Full-text available
Cognitive deficits in survivors of traumatic brain injury (TBI) are associated with irreversible neurodegeneration in brain regions such as the hippocampus. Comparative gene expression analysis of dying and surviving neurons could provide insight into potential therapeutic targets. We used two pathway-specific PCR arrays (RT2 Profiler Apoptosis and Neurotrophins & Receptors PCR arrays) to identify and validate TBI-induced gene expression in dying (Fluoro-Jade-positive) or surviving (Fluoro-Jade- negative) pyramidal neurons obtained by laser capture microdissection (LCM). In the Apoptosis PCR array, dying neurons showed significant increases in expression of genes associated with cell death, inflammation, and endoplasmic reticulum (ER) stress compared with adjacent, surviving neurons. Pro-survival genes with pleiotropic functions were also significantly increased in dying neurons compared to surviving neurons, suggesting that even irreversibly injured neurons are able to mount a protective response. In the Neurotrophins & Receptors PCR array, which consists of genes that are normally expected to be expressed in both groups of hippocampal neurons, only a few genes were expressed at significantly different levels between dying and surviving neurons. Immunohistochemical analysis of selected, differentially expressed proteins supported the gene expression data. This is the first demonstration of pathway-focused PCR array profiling of identified populations of dying and surviving neurons in the brain after TBI. Combining precise laser microdissection of identifiable cells with pathway-focused PCR array analysis is a practical, low-cost alternative to microarrays that provided insight into neuroprotective signals that could be therapeutically targeted to ameliorate TBI-induced neurodegeneration.
... Following TBI infiltration of neutrophils, induction of nitric oxide synthase, release of free oxygen radicals and pro-inflammatory microglial cytokines contribute to the posttraumatic inflammatory response [50][51][52]. Accordingly, experimental and clinical trials seek for efficient anti-inflammatory therapies in the acute phase to reduce brain lesion [16,53,54]. One of these potential neuroprotective anti-inflammatory drugs is the investigated a-MSH (11)(12)(13). ...
Article
Full-text available
Following traumatic brain injury (TBI) neuroinflammatory processes promote neuronal cell loss. Alpha-melanocyte-stimulating hormone (α-MSH) is a neuropeptide with immunomodulatory properties, which may offer neuroprotection. Due to short half-life and pigmentary side-effects of α-MSH, the C-terminal tripeptide α-MSH(11–13) may be an anti-inflammatory alternative. The present study investigated the mRNA concentrations of the precursor hormone proopiomelanocortin (POMC) and of melanocortin receptors 1 and 4 (MC1R/MC4R) in naive mice and 15 min, 6, 12, 24, and 48 h after controlled cortical impact (CCI). Regulation of POMC and MC4R expression did not change after trauma, while MC1R levels increased over time with a 3-fold maximum at 12 h compared to naive brain tissue. The effect of α-MSH(11–13) on secondary lesion volume determined in cresyl violet stained sections (intraperitoneal injection 30 min after insult of 1 mg/kg α-MSH(11–13) or 0.9% NaCl) showed a considerable smaller trauma in α-MSH(11–13) injected mice. The expression of the inflammatory markers TNF-α and IL-1β as well as the total amount of Iba-1 positive cells were not reduced. However, cell branch counting of Iba-1 positive cells revealed a reduced activation of microglia. Furthermore, tripeptide injection reduced neuronal apoptosis analyzed by cleaved caspase-3 and NeuN staining. Based on the results single α-MSH(11–13) administration offers a promising neuroprotective property by modulation of inflammation and prevention of apoptosis after traumatic brain injury.
Article
Full-text available
Subconcussive impacts are very common in the sports world and can have many negative impacts on human function, including increased risk for cognitive decline and behavioral impairments such as chronic traumatic encephalopathy (CTE). The purpose of this article is to analyze the available literature on the effects of jugular vein compression applied by a cervical collar on cerebral structure and function in the setting of chronic impact exposure. This narrative review analyzed 17 articles on brain structure and function, published between 1992 and 2022. Our review of the 17 studies shows an overall neuroprotective effect of the external jugular vein compression applied by the cervical collar during insult to the head as compared to groups who did not wear a collar. These findings suggest a potential role of the cervical collar, in addition to helmets, in reducing the incidence of concussion-induced microtraumas and cascading secondary injury mechanisms. Though positive results are consistent throughout the studies, future studies with increased sample sizes are necessary to create precise estimates of the effects of the cervical collar. In addition, the analyzed studies mainly looked at the effects of the cervical collar on football players, soccer players, and Special Weapons and Tactics (SWAT) team members; thus, additional rigorous studies are needed to assess the impact of the cervical collar on other high-risk populations such as military and law-enforcement personnel, among others.
Article
Purpose: Homer1a is a member of the post-synaptic density protein family that plays an important role in neuronal synaptic activity and is extensively involved in neurological disorders. The aim of this study is to investigate the role of Homer1a in modulating neuronal survival using an in vitro traumatic neuronal injury model. Materials and methods: Neurons were extracted from rats and identifited. Then, the cells were treated with Homerla overexpression or interference vectors. Western blot was performed to evaluate the expression of Homerla, apoptosis-related proteins(caspase3, caspase8, caspase9, Fasl, Bax, and p53), autophagy-related proteins (LC3ll and Beclin1), and the activiation of PI3K/AKT/mTOM pathway. In addition, the cell viability and apoptosis rate were measured. Results: After transfection with overexpression or interference vectors, the mRNA and protein expression of Homer1a increased or decreased significantly, respectively. Upregulation of Homer1a significantly alleviated apoptosis and enhanced cell viability and autophagy after traumatic neuronal injury. Homer1a overexpression also significantly decreased the expression of the pro-apoptosis proteins caspase 3, caspase 8, caspase 9, Fasl, Bax, and p53 in neurons. Furthermore, neuron autophagy was increased after traumatic neuronal injury as demonstrated by the greater accumulation of autophagosomes and higher expression of LC3II and Beclin1 induced by Homer1a overexpression. In addition, Homer1a overexpression inhibited the activation of PI3K/AKT/mTOR signaling. Conclusion: These findings indicated that Homer1a potentially protects neurons from traumatic injury by regulating apoptosis and autophagy via the caspase and PI3K/AKT/mTOR signaling pathways and may be an effective intervention target in traumatic brain injury.
Article
Retrobulbar hemorrhage is an uncommon, vision-threatening complication of orbit and eyelid surgery. We review the incidence of retrobulbar hemorrhage, risk factors, orbital anatomy, pathophysiology, diagnosis and treatment options. Preoperative, intraoperative and postoperative measures to prevent retrobulbar hemorrhage are discussed, and a treatment algorithm is presented.
Article
Homer protein, a member of the post-synaptic density (PSD) protein family, plays an important role in the neuronal synaptic activity and is extensively involved in neurological disorders. The present study investigates the role of Homer1b/c in modulating neuronal survival by using an in vitro traumatic neuronal injury model, which was achieved by using a punch device that consisted of 28 stainless steel blades joined together and produced 28 parallel cuts. Downregulation of Homer1b/c by specific siRNA significantly (p < 0.05) alleviated the cytoplasmic calcium levels and neuron lactate dehydrogenase (LDH) release, and ultimately decreased apoptotic rate after traumatic neuronal injury compared with non-targeting siRNA control treatment in cultured rat cortical neurons. Moreover, the expression of mGluR1a was significantly (p < 0.05) reduced in the Homer1b/c siRNA transfected neurons after injury. Therefore, Homer1b/c not only modulated the mGluR1a-inositol 1, 4, 5-triphosphate receptors (IP3R)-Ca(2+) signal transduction pathway, but also regulated the expression of mGluR1a in mechanical neuronal injury. These findings indicate that the suppression of Homer1b/c expression potentially protects neurons from glutamate excitotoxicity after injury and might be an effective intervention target in traumatic brain injury.
Article
The protein kinase C activator, phorbol 12-myristate 13-acetate (PMA), is known to interact with aquaporin-4 (AQP4), a waterselective transporting protein abundant in astrocytes and ependymal cells, that has been found to decrease osmotically-induced swelling. The purpose of this study was to examine whether PMA given at different time points following focal ischemia induced by middle cerebral artery occlusion (MCAO) reduces brain edema by AQP4 modulation. Male Sprague-Dawley rats were randomly assigned to sham procedure, vehicle, or PMA infusion (230 μg/kg), starting either 60 minutes before, or 30 or 60 minutes after MCAO (each group n = 12). After a 2-hour period of ischemia and 2 hours of reperfusion, the animals were sacrificed for assessment of brain water content, sodium, and potassium concentrations. AQP4 expression was assessed by immunoblotting. Statistical analysis was performed by ANOVA followed by Tukey's post hoc test. PMA treatment significantly reduced brain water content concentration in the infarcted area when started before or 30 minutes postocclusion (p < 0.001, p = 0.022) and prevented the subsequent sodium shift (p < 0.05). Furthermore, PMA reduced ischemia-induced AQP4 up-regulation (p ≤ 0.05). Attenuation of the ischemia-induced AQP4 up-regulation by PMA suggests that the reduction in brain edema formation following PMA treatment was at least in part mediated by AQP4 modulation.
Article
Experimental research during the past decade has greatly increased our understanding of the pathophysiology of traumatic brain injury (TBI) and allowed us to develop neuroprotective pharmacological therapies. Encouraging results of experimental pharmacological interventions, however, have not been translated into successful clinical trials, to date. Traumatic brain injury is now believed to be a progressive degenerative disease characterized by cell loss. The limited capacity for self-repair of the brain suggests that functional recovery following TBI is likely to require cellular transplantation of exogenous cells to replace those lost to trauma. Recent advances in central nervous system transplantation techniques involve technical and experimental refinements and the analysis of the feasibility and efficacy of transplantation of a range of stem cells, progenitor cells and postmitotic cells. Cellular transplantation has begun to be evaluated in several models of experimental TBI, with promising results. The following is a compendium of these new and exciting studies, including a critical discussion of the rationale and caveats associated with cellular transplantation techniques in experimental TBI research. Further refinements in future research are likely to improve results from transplantation-based treatments for TBI.
Article
Chapter
We evaluated the effects of magnesium sulfate on brain tissue oxygen (PtO2) tension, carbon dioxide (PtCO2) tension and pH (pHt) in patients undergoing temporary artery occlusion for clipping of cerebral aneurysm. We studied 18 patients with aneurysmal subarachnoid hemorrhage. All patients received standard anesthetics using target controlled infusion of propofol (3 µg/ml) and remifentanil (10 ng/ml). After craniotomy, a calibrated multiparameter sensor (Neurotrend, Diametrics Medical, Minneapolis, MN) was inserted to measure PtO2, PtCO2 and pHt in tissue at risk of ischemia during temporary artery occlusion. Patients were then randomly allocated to receive either intravenous saline or magnesium 20 mmol over 10 min followed by an infusion 4 mmol/h. Plasma magnesium concentration, brain tissue gases and pHt were determined at baseline, 30 min after study drug infusion and 4 min after temporary clipping. Data were analyzed by factorial ANOVA with repeated measures. Intergroup difference was compared with unpaired t test. P value < 0.05 was considered significant. Patient characteristics, baseline brain tissue gases and pHt did not differ between groups. Magnesium infusion increased PtO2 by 34%. Following temporary artery occlusion, PtO2 and pHt decreased and PtCO2 increased in both groups. However, tissue hypoxia was less severe and the rate of PtO2 decline was slower in the magnesium group. Our data suggested that magnesium enhances tissue oxygenation and attenuates hypoxia during temporary artery occlusion.
Article
Objective. —To evaluate outcome of patients with severe closed head injury treated with pegorgotein, a scavenger of oxygen-derived free radicals.Design. —Randomized, parallel, placebo-controlled, third-party—blind, multicenter trial, with a blinded, multicenter follow-up protocol.Setting. —Twenty-nine centers in the United States.Patients. —A total of 463 patients with severe closed head injury and a Glasgow Coma Scale score of 8 or less after resuscitation and stabilization.Interventions. —Patients received a single intravenous dose of placebo, 10 000 U/kg of pegorgotein, or 20000 U/kg of pegorgotein within 8 hours after injury.Outcome Measures. —The primary endpoint was the Glasgow Outcome Scale (GOS) score at 3 months after brain injury with GOS data trichotomized into good, fair, or poor outcome. Secondary efficacy endpoints included the Disability Rating Scale (DRS) and mortality. A secondary analysis was performed using GOS scores dichotomized into favorable and unfavorable outcomes. In a follow-up protocol at 6 months, GOS and DRS scores were again determined.Results. —Of 463 patients randomized, 162 received placebo; 149, pegorgotein 10000 U/kg; and 152, pegorgotein 20000 U/kg. Treatment groups were comparable with respect to demographic characteristics, mechanism of injury, and time to treatment. Pegorgotein was well tolerated at both dose levels. At month 3, the trichotomized analysis found no significant statistical difference in neurologic outcome between the pegorgotein and the placebo groups. Although differences were not statistically significant, there were more favorable outcomes and no increase in the number of deaths or vegetative states among the patients given pegorgotein, more subjects had good or favorable outcomes with the 10000-U/kg dose than with the 20000-U/kg dose or placebo, and less disability was observed with the 10000-U/kg dose than with either the 20000-U/kg dose or placebo. No differences in mortality rate or cause of death were found between the 10000-U/kg and placebo groups at either month 3 or month 6.The only statistically significant difference between the groups was a decreased incidence of adult respiratory distress syndrome in the 10000-U/kg group as compared with the placebo group (P<.015).Conclusions. —In this clinical trial of 463 patients with severe head injury, no statistically significant difference in neurologic outcome or mortality was observed between patients treated with pegorgotein and those receiving placebo.
Article
Hypertonic saline (7.5% NaCl = HS) was compared with 20% mannitol (MAN), for the treatment of increased intracranial pressure (ICP), in a large animal model of head injury. Sheep were instrumented for hemodynamic and ICP monitoring and fluid administration. Elevated ICP (20-25 mm Hg) was produced by inflating an epidural balloon for 1 hour. Animals were then given a bolus of 250 mL of either HS (n = 7) or MAN (n = 7) and monitored for 2 hours. No significant differences in hemodynamic variables were noted between groups. The ICP decreased to the same degree in both groups during the 2 hours of observation (HS = 11 +/- 3.8 mm Hg; MAN = 8 +/- 2 mm Hg). Brain water contents were also similar (HS = 3.68 +/- 0.09 mL H2O/g dry wt; MAN = 3.83 +/- 0.08 mL H2O/g dry wt). The 7.5% NaCl was equally effective in treating elevated ICP caused by a space-occupying lesion when compared with 20% mannitol. Hypertonic saline has the additional benefit of rapid cardiovascular resuscitation of associated hemorrhagic shock with small-volume infusion.