ArticlePDF AvailableLiterature Review

Pharmacologic Neuroprotective Strategies in Neonatal Brain Injury

Authors:

Abstract and Figures

This article explains the mechanisms underlying choices of pharmacotherapy for hypoxic-ischemic insults of both preterm and term babies. Some preclinical data are strong enough that clinical trials are now underway. Challenges remain in deciding the best combination therapies for each age and insult.
Content may be subject to copyright.
Pharmacologic Neuroprotective
Strategies in Neonatal Brain
Injury
Sandra E. Juul, MD, PhD
a,
*, Donna M. Ferriero, MD, MS
b
MECHANISMS OF BRAIN INJURY: PRETERM VERSUS TERM
The two most common causes of neonatal brain injury in the United States are
extreme prematurity and hypoxic-ischemic encephalopathy (HIE). In the United
States, 1 in 8 babies is born before term (37–40 weeks), and 1.44% of babies
(56,000 per year) are born with a birth weight of 1250 g or less.
1
These small, preterm
babies are at high risk of death or neurodevelopmental impairment: approximately
20% die before hospital discharge, and 40% of survivors develop long-term intellec-
tual or physical impairment, including cerebral palsy (CP).
2–4
Care of preterm infants
accounts for more than half of pediatric health care dollars spent.
The brain rapidly increases in size, shape, and complexity during the second and
third trimesters.
5
Neurodevelopmental compromise can result from an interruption
of normal development or from damage to existing tissues. Brain development during
this period is vulnerable to hypoxia-ischemia (HI), oxidant stress, inflammation, exci-
totoxicity, and poor nutrition. These exposures can result in structural, biochemical,
Disclosure Statement: Neither author has anything to disclose.
a
Department of Pediatrics, University of Washington, 1959 Northeast Pacific Street,
Box 356320, Seattle, WA 98195, USA;
b
Neonatal Brain Disorders Laboratory, University of
California, San Francisco, 675 Nelson Rising Lane, Room 494, Box 0663, San Francisco, CA
94143, USA
* Corresponding author.
E-mail address: sjuul@uw.edu
KEYWORDS
Brain injury Hypoxic-ischemic encephalopathy Prematurity Preconditioning
KEY POINTS
There are many ways to achieve neuroprotection: preconditioning, salvaging, repair.
Hypothermia is now standard of care for term hypoxic-ischemic encephalopathy so
studies to investigate additional therapies will be added to that treatment.
Strategies that target multiple mechanisms and consider age-appropriate mechanisms
will be most beneficial.
Clin Perinatol 41 (2014) 119–131
http://dx.doi.org/10.1016/j.clp.2013.09.004 perinatology.theclinics.com
0095-5108/14/$ – see front matter Published by Elsevier Inc.
and cell-specific injury.
6
Preoligodendrocytes, which emerge and mature between 24
and 32 weeks of development, are particularly susceptible to injury, and damage to
these cells can result in white matter injury.
7
Although intracranial hemorrhage, peri-
ventricular leukomalacia, inflammatory conditions, and male gender are known risk
factors for poor outcomes, little is known about how to improve these outcomes.
HIE is estimated to contribute significantly to 23% of the 4 million neonatal deaths
that occur annually.
8
In the United States, HIE occurs in 1.5 to 2 live births per 1000,
with a higher incidence in premature infants.
9
Untreated, the sequelae of moderate to
severe HIE includes a 60% to 65% risk of mental retardation, CP, hydrocephalus, sei-
zures, or death. Perinatal inflammation is increasingly recognized as an important
contributor to neonatal HIE and poor neurodevelopmental outcomes
10
: the presence
of maternal fever alone increases the risk for CP, and chorioamnionitis further in-
creases the risks for brain injury in both preterm and term infants.
11,12
Timing of infec-
tion/inflammation relative to hypoxia is critical: it can be sensitizing (increase brain
injury) if it occurs acutely or after 72 hours, but may be protective if it occurs 24 hours
before hypoxia.
13
This differential response is not fully understood, but may depend on
activation of fetal/neonatal Toll-like receptors in the brain.
14,15
Understanding the
complex mechanisms of brain injury is essential to devising protective strategies.
THE INJURY CASCADE
Although the cellular targets of HI are different depending on age and severity of
insult, the basic cascade of injury occurs in a uniform way regardless of age and con-
tinues for a prolonged period of time. Cell death occurs in 2 main phases: primary
death from hypoxia and energy depletion, followed by reperfusion and increased
free radical (FR) formation, excitotoxicity, and nitric oxide production with secondary
energy failure and delayed death (Fig. 1). A tertiary phase was recently proposed, in
which factors can worsen outcome, predispose a newborn to further injury, or prevent
repair or regeneration after an initial insult to the brain.
16
Such mechanisms include
persistent inflammation and epigenetic changes, which cause a blockade of oligo-
dendrocyte maturation, impaired neurogenesis, impaired axonal growth, or altered
synaptogenesis.
The injury process begins with energy failure creating excitotoxicity, which is
caused by excessive glutamatergic activation that leads to progression of HI brain
injury. Glutamate plays a key role in development, affecting progenitor cell prolifera-
tion, differentiation, migration, and survival. Glutamate accumulates in the brain after
Fig. 1. The injury cascade as it occurs over time. Potential therapeutics are inserted during
the course of the cascade. See text for details on these agents. Epo, erythropoietin.
Juul & Ferriero
120
HI
17
from a variety of causes, including vesicular release from axons and reversal
of glutamate transporters. Glutamatergic receptors include N-methyl-D-aspartate
(NMDA), alpha-3-amino-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA), and
kainate. Developmental differences in glutamate receptor expression contribute to
the vulnerability of the immature brain (reviewed in Ref.
18
). NMDA receptor activation,
although important for synaptic plasticity and synaptogenesis, can increase intracel-
lular calcium, proapoptotic pathways via caspase-3 activation, FR formation, and
lipid peroxidation, resulting in profound and widespread injury to the developing
brain.
Oxidative stress is an important component of early injury, along with excitotoxicity,
to the neonatal brain resulting from the excess formation of FRs (reactive oxygen spe-
cies and reactive nitrogen species) under pathologic conditions. These FRs include
superoxide anion (O
2
), hydroxyl radical (OH), singlet oxygen (
1
O
2
), and hydrogen
peroxide (H
2
O
2
). FRs target lipids, protein, and DNA, causing damage to these cellular
components and initiating a cascade that results in cell death.
19
These deleterious biological events trigger inflammatory processes that initially are
harmful and later may be beneficial to the repair processes that occur after injury. The
inflammatory response and cytokine production that accompany infection may play a
large role in cell damage and loss. Local microglia are activated early and produce
proinflammatory cytokines such as tumor necrosis factor (TNF)-alpha, interleukin
(IL)-1b, and IL-6, as well as glutamate, FRs, and nitric oxide (NO), and are the main
immunocompetent cells in the immature brain. Depending on the stimulus, molecular
context, and timing, these cells acquire various phenotypes, which are critical to the
outcome of the injury.
20
Cell death occurs throughout the cascade, moving from a purely necrotic type to
apoptosis with a continuum of phenotypes emerging in the developing brain (the
apoptosis-necrosis continuum). Throughout this process, mechanistic interactions
between cell death and hybrid forms of cell death occur, such as programmed or regu-
lated necrosis or necroptosis.
21
The mechanisms behind programmed necrosis in
neonatal brain injury are still being investigated but are regulated by the inflammatory
processes, especially the TNF receptor superfamily, which is activated early in injury.
There are many pharmacologic agents that can affect these injury phases (Table 1).
This article highlights some of the important future therapeutics.
Table 1
Best pharmacologic candidates for impact on injury phases
Best Candidates
Antenatal Postnatal
BH4 Melatonin
Melatonin Epo
nNOS inhibitors NAC
Xenon Xenon
Allopurinol Allopurinol
Vitamin C and E Vitamin C and E
Resveratrol Resveratrol
NAC Memantine
— Topiramate
Abbreviations: BH4, tetrahydrobiopterin; NAC, N-acetyl-L-cysteine; nNOS, neuronal nitric oxide
synthase.
Strategies in Neonatal Brain Injury 121
TARGETING THE INJURY RESPONSE
Antiexcitotoxic Agents
The earliest pharmacologic strategies to protect the newborn brain were designed to
block the initial phases of injury, excitotoxicity, and oxidative stress. Many of these
agents failed because it is impossible to block normal developmental processes,
like glutamatergic signaling, without harming the brain. Therefore, therapies designed
to block the NMDA receptor resulted in increased, rather than decreased, cell death.
22
However, some agents, like magnesium sulfate, used to stop preterm labor, seemed
to have beneficial effects even though they blocked the NMDA receptor. Recent clin-
ical trials have supported the use of antenatal magnesium.
23
In 16 hospitals in
Australia and New Zealand, 1062 women with fetuses younger than 30 weeks’ gesta-
tion were given a loading infusion of 16 mmol followed by 8 mmol MgSO
4
for up to
24 hours. Substantial gross motor dysfunction (3.4% vs 6.6%; relative risk (RR),
0.51; 95% confidence interval [CI], 0.29–0.91) and combined death or substantial
gross motor dysfunction were significantly reduced in the MgSO
4
group, although
there were no significant differences in mortality or CP in survivors. In the United
States, antenatal MgSO
4
did not reduce the risk of the composite outcome of CP or
death, much like the Australian study, but was seen to reduce moderate to severe
CP without increasing the risk of death.
23
However, the number needed to treat
was high at 56 (95% CI, 34–164).
Another potential antiexcitotoxic agent is xenon, a noble gas used as an anesthetic
agent. It has action against the NMDA receptor and has been shown to be an effective
agent against hypoxic-ischemic insult both to cortical neurons in vitro and in several
in vivo models.
24
Xenon lacks the dopamine-releasing properties that are present in
other NMDA antagonists and does not cause increased apoptotic cell death as
seen with other NMDA antagonists. Perhaps the most promising aspect of xenon
pharmacology is that it increases the translational efficiency of hypoxia inducible
factor-1 (HIF-1) through a mammalian target of rapamycin (mTOR) pathway
25
that
has resulted in its potential application in HIE for both postinjury treatment and as a
preconditioner. The prolonged increase in expression of HIF-1a by xenon causes
upregulation of cytoprotective proteins such as erythropoietin (Epo), vascular endo-
thelial growth factor (VEGF), and glucose transporter 1 protein.
25
When given with
sevoflurane during labor to rats, it preconditions the fetal brain against a subsequent
HI insult.
26
When given to neonatal rats in combination with hypothermia, it improves
both functional and structural outcomes, even when hypothermia is delayed, and the
effect is sustained through adulthood.
27
Antioxidants
Therapies designed to reduce oxidative stress have proved to be efficacious both in
the preterm and term injury states. Allopurinol was originally shown to be neuroprotec-
tive in postnatal day 7 rats after HI
28
but in humans was not seen to improve short-term
or long-term outcomes in a small trial after birth asphyxia.
29
It was postulated that the
drug needed to be given before reperfusion injury set in, so trials are now underway to
evaluate efficacy when given to mothers who have fetuses suspected of intrauterine
hypoxia. In a randomized, double-blind, placebo-controlled multicenter study that is
now in progress, intravenous allopurinol is being given antenatally with the primary
outcome being serum brain damage markers (S100b) and oxidative stress markers
(isoprostanes and so forth) in umbilical cord blood; secondary outcome measures
are neonatal mortality, serious composite neonatal morbidity, and long-term neuro-
logic outcome.
30
There is now a randomized, placebo-controlled, double-blinded
Juul & Ferriero
122
parallel group comparison study of hypothermia and allopurinol ongoing (the European
ALBINO Trial). Allopurinol is being given twice: 30 minutes after birth and then 12 hours
later, in addition to hypothermia in moderate to severe HIE. Outcomes will be assessed
at 2 years of life.
There are many antioxidants that have been investigated in both preterm and term
HI injury. Scavengers such as melatonin and vitamin E have shown promise. Lipid
peroxidation inhibitors such as the lazaroids, gingko biloba, and caffeic acid, and
FR reducers such as ebselen and Epo, have produced some amelioration of injury.
Nitric oxide synthase inhibitors such as aminoguanidine, L-omega-nitro-arginine-
methyl-ester (L-NAME), 7 nitroindazole, and newer derivatives are still being
investigated.
31,32
The most promising of these agents seems to be melatonin, which shows efficacy in
both preterm and term injury. Melatonin has many targets along the injury cascade,
including oxidative stress, inflammation, apoptosis, mitochondrial failure, as well as
nuclear effects. The benefit is in the lack of significant side effects in children and
term neonates. A recent observational study showed that melatonin levels are deficient
in preterm and term newborn infants, and it is now being trialed daily for 7 days after
premature birth to identify whether it will reduce the risk of prematurity-associated
brain injury (MINT; ISRCTN15119574). Another investigation is underway in premature
and full-term babies to identify optimal treatment doses (MELIP, NCT01340417; and
MIND, NCT01340417), and there is a study to determine the effects of maternal sup-
plementation on outcome in term infants (PREMELIP; identification number pending).
An Australian study evaluating melatonin to prevent brain injury in unborn growth-
restricted babies is ongoing in which mothers receive melatonin during pregnancy
and oxidative stress is monitored in maternal serum, placenta, and umbilical cord
blood. A composite neonatal outcome will be evaluated (NCT01695070).
Dietary manipulations may also prove promising in neuroprotection. Pomegranate
juice is rich in polyphenols that can protect the neonatal mouse brain against an HI
insult when given to mothers in their drinking water.
33
Even when given after the insult
to neonatal animals, there is substantial protection in hippocampus, cortex, and stria-
tum.
34
Omega-3 polyunsaturated fatty acid supplementation can reduce brain dam-
age and improve long-term neurologic outcomes even 5 weeks after an HI insult to
rodents. The effect is best appreciated in microglia, where nuclear factor kappa B acti-
vation and release of inflammatory mediators are inhibited, thus providing an antiin-
flammatory effect as well.
35
Antiinflammatory Agents
As mentioned earlier, melatonin has multiple targets in the injury cascade and is a
perfect candidate for manipulating inflammation. In several animal models, small
and large, preterm and term, it has shown efficacy against excitotoxic lesions as
well as HI. A recent study in rodents revealed that melatonin preserved white matter
and learning disabilities after ibotenate lesions to the postnatal day 5 brain and was
equally efficacious against IL-1binjections.
36
In a fetal sheep model at E90, cord oc-
clusion produced substantial white matter injury that was blocked with melatonin.
37
In
term brain models of HI, melatonin markedly decreased microglial activation and pre-
served myelination.
38
The most promising study to date in piglets revealed that mela-
tonin, in combination with hypothermia, provided substantial improvement compared
with hypothermia in preserving brain function measured by amplitude-integrated elec-
troencephalogram, and reduced cell death in the thalamus, the region most affected
by the asphyxia insult.
39
There is now a collaborative study between Hopital Robert
Debre and St Thomas Hospital (Kings College London) as a proof of concept in
Strategies in Neonatal Brain Injury 123
neuroprotection study. It will be a double-blinded randomized trial of premature new-
borns of less than 28 weeks’ gestational age. Babies will be randomized to placebo,
low-dose melatonin, or high-dose melatonin, and outcome will be assessed by mag-
netic resonance imaging and neurodevelopmental outcome at 24 months (MINT Trial,
ISRCTN15119574).
GROWTH FACTORS AS NEUROPROTECTANTS
Many growth factors have essential roles during fetal and postnatal brain develop-
ment. Although the effects of some, such as brain-derived neurotrophic factor
(BDNF), are largely restricted to the brain, others such as Epo, VEGF, granulocyte col-
ony–stimulating factor (GCSF), and insulinlike growth factor 1 (IGF-1) have important
somatic effects in addition to their roles in neurodevelopment. All of the factors listed
earlier have been evaluated as neuroprotectant therapies for adult and neonatal brain
injury. At this time, Epo is the best studied for this purpose, and is the closest to clinical
use. The pleiotropic nature of these growth factors makes it essential to test meticu-
lously for safety before clinical use, particularly because very high doses are often
required for neuroprotection, given that these large molecules do not readily cross
the blood-brain barrier.
Epo
Epo and its receptor (EpoR) are expressed in the developing central nervous system
(CNS), and are required for normal brain development.
40
Acute exposure to hypoxia
upregulates the expression of EpoR on oligodendrocytes and neurons, without a
commensurate increase in Epo expression.
41
The presence of unbound cell surface
EpoR drives cells of neuronal and oligodendrocyte lineage to apoptosis, whereas
ligand-bound EpoR activates survival signaling pathways. With Epo binding, EpoR di-
merizes to activate antiapoptotic pathways via phosphorylation of JAK2, phosphory-
lation, and activation of mitogen-activated protein kinase (MAPK), extracellular related
kinase (ERK1/2), as well as the phosphatidylinositaol 3-kinase (PI3K/Akt) pathway and
signal transducer and transcriptional activator 5 (STAT5), which are critical in cell sur-
vival.
42
Epo also stimulates production of BDNF. Epo signaling inhibits early mecha-
nisms of brain injury by its antiinflammatory,
43,44
antiexcitotoxic,
45
antioxidant,
46,47
and antiapoptotic effects on neurons and oligodendrocytes. Repair of brain injury is
also enhanced in the presence of Epo because of its positive effects on neurogenesis
and angiogenesis, which are essential for plasticity and remodeling.
48,49
Epo effects
are dose dependent, with multiple doses being more effective than single doses.
50,51
Epo reduces neuronal loss and learning impairment following brain injury,
52
and, even
when initiated as late as 48 to 72 hours after injury, there is evidence of improved
behavioral outcomes, enhanced neurogenesis, increased axonal sprouting, and
reduced white matter injury in animal models of brain injury.
53,54
Epo is now under investigation for both term and preterm brain injury. The antiin-
flammatory, antiexcitotoxic, and antioxidant effects are relevant to brain injury in
both age groups. The specific effects of Epo in preoligodendrocytes may be most rele-
vant to the white matter injury that characterizes preterm brain injury. Treatment
approaches to acute brain injury in term infants (HIE) and preterm infants (intraventric-
ular hemorrhage [IVH]) should differ from preventative strategies in preterm infants. In
the former, a shorter duration of high-dose Epo is most appropriate, whereas for the
latter, a more prolonged treatment strategy that continues during the period of oligo-
dendrocyte vulnerability is most likely to succeed. In addition to the specific cellular
effects on neurons and oligodendrocytes, this more prolonged treatment also
Juul & Ferriero
124
decreases the availability and potential toxicity of free iron, caused by the erythropoi-
etic effects of Epo, by increasing iron utilization.
Translation to Clinical Trials
Epo does not cross the placenta, so prenatal treatment is not an option. It is approved
by the US Food and Drug Administration and has a robust safety profile in neonates,
with more than 3000 neonates randomized to placebo-controlled trials testing its eryth-
ropoietic effects.
55
Doses required for neuroprotection are higher than those used for
prevention and treatment of anemia, because only a small fraction of circulating Epo
crosses the blood brain barrier. In animal models of neonatal brain injury, Epo doses
of 1000 to 5000 U/kg result in sustained neuroprotection, improving both short-term
and long-term structure and function.
56
Phase I/II trials have been done to establish
safety and translational pharmacokinetics of Epo in preterm
57,58
and term neonates.
59
These studies suggest that 1000 U/kg/dose provides an area under the curve (AUC)
most similar to a neuroprotective dose of 5000 U/kg in rodents (Table 2). The optimal
dose and duration of treatment is likely to differ for treatment of HIE compared with pre-
venting or treating brain injury in preterm infants, and is not yet known. Note that the
pharmacokinetics in preterm and term asphyxiated infants are different, with a longer
half-life noted at higher doses in infants with HIE. Phase II and III studies are now un-
derway for neuroprotection of both extreme prematurity and HIE in term infants
(Box 1). In the United States, a multicenter randomized controlled trial of preterm
Epo neuroprotection is beginning (PENUT trial, NCT01378273). This study will use
1000 U/kg for 6 doses followed by 400 U/kg 3 times a week until 33 weeks of gestation.
A Swiss trial has used 3000 U/kg for 3 doses in the first weeks of life. Enrollment is com-
plete for this trial, with follow-up underway. The BRITE study, comparing Darbepoetin,
Epo, and placebo, is showing improved outcomes in preterm neonates receiving either
Darbepoetin or Epo. Erythropoietic agents (Epo and darbepoetin) are also being stud-
ied in combination with hypothermia for the treatment of HIE in term infants. Pilot
studies have shown safety and early signs of benefit, and larger studies are planned
or ongoing in the United States, France (NCT01732146), and China.
BDNF
BDNF is an important growth factor during fetal brain formation, particularly in the hip-
pocampus, cerebral cortex, basal forebrain, and cerebellum. It is also active in adult
neurogenesis. BDNF binds primarily to receptor tyrosine kinase B and activates
MAPK and Ca
21
-calmodulin kinase II (CAMKII), which regulate cAMP-responsive
Table 2
Epo pharmacokinetics
Epo Dose (U/kg) AUC C
max
T
1/2
P7 Rodents SC 5000 117,677 6224 8.4
P7 Rodents IP 5000 140,331 10,015 6.7
Preterm infants <1000 g 500 31,412 2780 8078 538 5.4 0.6
1000 81,498 7067 14,017 1293 7.1 0.7
2500 317,881 22,941 46,467 2987 8.7 1.4
Term HIE infants 500 50,306 67,426 7046 814 7.2 1.9
1000 131,054 17,083 13,780 2674 15.0 4.5
2500 328,002 61,945 33,316 7377 18.7 4.7
Abbreviations: AUC, area under the curve; C
max
, maximum plasma concentration of the drug; IP,
intraperitoneal; P7, postnatal day 7; SC, subcutaneously; T
1/2
, half life.
Strategies in Neonatal Brain Injury 125
element binding (CREB) and synapsin transcription. Neuroprotection from glutamate
toxicity is mediated through PI3K and the Ras/MAPK signaling pathways, and involves
an increase in B-cell lymphoma 2 (bcl-2) proteins.
60
Both exercise and caffeine in-
crease BDNF secretion, thereby increasing recognition memory and neurogenesis.
VEGF
VEGF is a growth factor that is stimulated by HIF-1 and stimulates vasculogenesis and
angiogenesis, essential processes needed for brain development and brain repair. In
addition, VEGF has specific neurotrophic and neuroprotective effects in adult and
neonatal models of hypoxic-ischemic brain injury.
61
These effects also involve activa-
tion of Akt and extracellular receptor kinase (ERKs). Tight regulation of this factor is
needed because both overexpression and underexpression can contribute to disease.
GCSF
GCSF is a hematopoietic glycoprotein that stimulates the clonal maturation of neutro-
phil progenitors and increases many functional activities of mature neutrophils. In
addition to these hematopoietic effects, GCSF and its receptor are expressed on
neuronal cells in a variety of brain regions.
62
GCSF has shown neuroprotection in
several models of brain injury, and is well tolerated at high doses. GCSF has been
Box 1
Clinical trials of neuroprotective agents
Phase 1 and 2 trials
Xenon and cooling therapy in babies at high risk of brain injury following poor condition at
birth: randomized pilot study. Bristol, United Kingdom
Phase 2 and 3 trials
Safety and efficacy of topiramate in neonates with HIE treated with hypothermia (NeoNATI),
Florence, Italy. NCT01241019
Neonatal Epo and therapeutic hypothermia; short-term outcome study (NEAT O Study).
University of California, San Francisco, multicenter, Thrasher funded
Darbepoetin administration in newborns undergoing cooling for encephalopathy (DANCE
Study) Utah, multicenter, Thrasher funded. NCT01471015
Phase 3 trials
A multicenter randomized controlled trial of therapeutic hypothermia plus magnesium
sulfate (MgSO
4
) versus therapeutic hypothermia plus placebo in the management of term
and near-term babies with HIE. Turkey. NCT01646619
Optimizing cooling strategies at less than 6 hours of age for neonatal HIE. A National
Institute of Child Health and Human Development (NICHD)–funded project. NCT01192776
Phase III study of efficacy of high-dose Epo to prevent HIE sequelae in term newborns. Paris,
France. NCT01732146
Evaluation of systemic hypothermia initiated after 6 hours of age in infants of greater than
or equal to 36 weeks’ gestation with HIE: a bayesian evaluation. An NICHD-funded project.
NCT00614744
Preterm Epo Neuroprotection (PENUT) Trial. A multicenter, randomized, placebo-controlled
phase III 940–subject trial of Epo for the neuroprotection of extremely low gestational age
neonates. An NINDS-funded project. NCT01378273
Melatonin as a Novel Neuroprotectant in Preterm Infants Trial (MINT). UK trial funded by
Medical Research Council. ISRCTN15119574
Juul & Ferriero
126
reported to have antiapoptotic, antiinflammatory, antiexcitotoxic, and neurotrophic
properties with demonstrated improved long-term outcomes.
63
It is currently being
evaluated in a phase II clinical trial for adult ischemic stroke (NCT00132470).
64
IGF-1
IGF-1 has an important role in normal brain development, promoting neuronal growth,
cellular proliferation, and differentiation in vitro, when injected directly into brain, or
when given intranasally.
65
It has also been found to have neuroprotective effects,
improving long-term function after hypoxic-ischemic brain injury. However, its clinical
application to neurologic disorders is limited by its large molecular size, poor central
uptake, and mitogenic potential.
Drug Delivery
When considering drugs for neuroprotection, drug delivery is an important consider-
ation. For example, if a drug crosses the placenta and can be tolerated by the mother,
prenatal treatment is an option, such as with xenon and melatonin. The ability to give
the drug intravenously is important, given that many critically ill neonates cannot take
oral medications. Some medications, such as melatonin, are presently only available
as oral formulations. Inhalational agents such as xenon have the problem that, if a sig-
nificant oxygen requirement exists, the ability to deliver neuroprotective concentra-
tions (30%–50%) may be limited.
Box 2
Gaps in knowledge
1. Cytokine response
Innate immunity differs in newborns compared with adults
Th1 preponderance in newborns
Th2 in adults
When does it change?
How is this affected by prematurity?
How does this affect brain injury/repair?
2. Microglial response
Three activation states of CNS microglia
M1: classic activation (tissue defense, proinflammatory)
M2: alternative activation (repair, antiinflammatory, fibrosis, matrix reconstruction)
M3: acquired deactivation (immunosuppression, phagocytosis of apoptotic cells)
How are these states regulated?
Can this response be harnessed for healing?
Does it differ for preterm versus term infants?
3. Preconditioning
Hypoxia
Inflammation
What are the molecular mechanisms?
Can these mechanisms be harnessed to improve outcomes?
Strategies in Neonatal Brain Injury 127
Small neuroactive peptides may have a therapeutic advantage compared with
larger molecules such as Epo or GCSF, which are larger glycoproteins that do not
cross the blood-brain barrier well. Modified neuropeptides and mimetic peptides
have been designed to overcome these barriers, and are in various stages of testing.
Another novel approach is to use alternative delivery methods, targeting the inflam-
matory system. Polyamidoamine dendrimers have been shown to localize in activated
microglia and astrocytes in the brains of newborn rabbits with CP, but not healthy con-
trols.
66
This nanotechnology approach has been used with excellent results to deliver
dendrimer-bound N-acetyl-L-cysteine (NAC) to the target inflammatory cells to sup-
press neuroinflammation, using much lower concentrations than are needed with sys-
temic dosing.
Combination Therapies
As more information is gained about mechanisms of brain injury and neuroprotection,
combination therapies may be applied. As mentioned earlier, some preclinical combi-
nation studies are already underway, for example, hypothermia plus xenon, and hypo-
thermia plus Epo for the treatment of term HIE.
Gaps in Knowledge
There remain gaps in knowledge (Box 2). All of these must be considered in the
context of the developing neonate, because immune function, cell populations, and
specific vulnerabilities and response to injury change over time. As clinicians become
more knowledgeable in these areas, new approaches to neuroprotection may become
apparent.
REFERENCES
1. Hamilton BE, Hoyert DL, Martin JA, et al. Annual summary of vital statistics:
2010-2011. Pediatrics 2013;131:548–58.
2. O’Shea TM, Allred EN, Dammann O, et al. The ELGAN study of the brain and
related disorders in extremely low gestational age newborns. Early Hum Dev
2009;85:719–25.
3. Stoll BJ, Hansen NI, Bell EF, et al. Neonatal outcomes of extremely preterm in-
fants from the NICHD Neonatal Research Network. Pediatrics 2010;126:443–56.
4. Gargus RA, Vohr BR, Tyson JE, et al. Unimpaired outcomes for extremely low
birth weight infants at 18 to 22 months. Pediatrics 2009;124:112–21.
5. Lodygensky GA, Vasung L, Sizonenko SV, et al. Neuroimaging of cortical devel-
opment and brain connectivity in human newborns and animal models. J Anat
2010;217:418–28.
6. Volpe JJ. Brain injury in premature infants: a complex amalgam of destructive
and developmental disturbances. Lancet Neurol 2009;8:110–24.
7. Back SA, Riddle A, McClure MM. Maturation-dependent vulnerability of peri-
natal white matter in premature birth. Stroke 2007;38:724–30.
8. Black RE, Cousens S, Johnson HL, et al. Global, regional, and national causes
of child mortality in 2008: a systematic analysis. Lancet 2010;375:1969–87.
9. Kurinczuk JJ, White-Koning M, Badawi N. Epidemiology of neonatal encepha-
lopathy and hypoxic-ischaemic encephalopathy. Early Hum Dev 2010;86:
329–38.
10. van Vliet EO, de Kieviet JF, Oosterlaan J, et al. Perinatal infections and neurode-
velopmental outcome in very preterm and very low-birth-weight infants: a meta-
analysis. JAMA Pediatr 2013;167:662–8.
Juul & Ferriero
128
11. Badawi N, Kurinczuk JJ, Keogh JM, et al. Intrapartum risk factors for newborn en-
cephalopathy: the Western Australian case-control study. BMJ 1998;317:1554–8.
12. Wu YW, Croen LA, Shah SJ, et al. Cerebral palsy in a term population: risk fac-
tors and neuroimaging findings. Pediatrics 2006;118:690–7.
13. Eklind S, Mallard C, Arvidsson P, et al. Lipopolysaccharide induces both a pri-
mary and a secondary phase of sensitization in the developing rat brain. Pediatr
Res 2005;58:112–6.
14. Vontell R, Supramaniam V, Thornton C, et al. Toll-like receptor 3 expression in
glia and neurons alters in response to white matter injury in preterm infants.
Dev Neurosci 2013;35(2–3):130–9.
15. Stridh L, Ek CJ, Wang X, et al. Regulation of toll-like receptors in the choroid
plexus in the immature brain after systemic inflammatory stimuli. Transl Stroke
Res 2013;4:220–7.
16. Fleiss B, Gressens P. Tertiary mechanisms of brain damage: a new hope for
treatment of cerebral palsy? Lancet Neurol 2012;11:556–66.
17. Gucuyener K, Atalay Y, Aral YZ, et al. Excitatory amino acids and taurine levels
in cerebrospinal fluid of hypoxic ischemic encephalopathy in newborn. Clin
Neurol Neurosurg 1999;101:171–4.
18. Jensen FE. Developmental factors regulating susceptibility to perinatal brain
injury and seizures. Curr Opin Pediatr 2006;18:628–33.
19. Buonocore G, Perrone S, Bracci R. Free radicals and brain damage in the
newborn. Biol Neonate 2001;79:180–6.
20. Hagberg H, Gressens P, Mallard C. Inflammation during fetal and neonatal life:
implications for neurologic and neuropsychiatric disease in children and adults.
Ann Neurol 2012;71:444–57.
21. Chavez-Valdez R, Martin LJ, Northington FJ. Programmed necrosis: a prominent
mechanism of cell death following neonatal brain injury. Neurol Res Int 2012;
2012:257563.
22. Ikonomidou C, Turski L. Why did NMDA receptor antagonists fail clinical trials for
stroke and traumatic brain injury? Lancet Neurol 2002;1:383–6.
23. Costantine MM, Drever N. Antenatal exposure to magnesium sulfate and neuro-
protection in preterm infants. Obstet Gynecol Clin North Am 2011;38:351–66, xi.
24. Wilhelm W, Hammadeh ME, White PF, et al. General anesthesia versus moni-
tored anesthesia care with remifentanil for assisted reproductive technologies:
effect on pregnancy rate. J Clin Anesth 2002;14:1–5.
25. Ma D, Lim T, Xu J, et al. Xenon preconditioning protects against renal ischemic-
reperfusion injury via HIF-1alpha activation. J Am Soc Nephrol 2009;20:713–20.
26. Yang T, Zhuang L, Rei Fidalgo AM, et al. Xenon and sevoflurane provide anal-
gesia during labor and fetal brain protection in a perinatal rat model of hypox-
ia-ischemia. PLoS One 2012;7:e37020.
27. Thoresen M, Hobbs CE, Wood T, et al. Cooling combined with immediate or de-
layed xenon inhalation provides equivalent long-term neuroprotection after
neonatal hypoxia-ischemia. J Cereb Blood Flow Metab 2009;29:707–14.
28. Palmer C, Towfighi J, Roberts RL, et al. Allopurinol administered after inducing
hypoxia-ischemia reduces brain injury in 7-day-old rats. Pediatr Res 1993;33:
405–11.
29. Benders MJ, Bos AF, Rademaker CM, et al. Early postnatal allopurinol does not
improve short term outcome after severe birth asphyxia. Arch Dis Child Fetal
Neonatal Ed 2006;91:F163–5.
30. Kaandorp JJ, Benders MJ, Rademaker CM, et al. Antenatal allopurinol for
reduction of birth asphyxia induced brain damage (ALLO-Trial); a randomized
Strategies in Neonatal Brain Injury 129
double blind placebo controlled multicenter study. BMC Pregnancy Childbirth
2010;10:8.
31. Buonocore G, Groenendaal F. Anti-oxidant strategies. Semin Fetal Neonatal
Med 2007;12:287–95.
32. Yu L, Derrick M, Ji H, et al. Neuronal nitric oxide synthase inhibition prevents ce-
rebral palsy following hypoxia-ischemia in fetal rabbits: comparison between
JI-8 and 7-nitroindazole. Dev Neurosci 2011;33:312–9.
33. Loren DJ, Seeram NP, Schulman RN, et al. Maternal dietary supplementation
with pomegranate juice is neuroprotective in an animal model of neonatal
hypoxic-ischemic brain injury. Pediatr Res 2005;57:858–64.
34. West T, Atzeva M, Holtzman DM. Pomegranate polyphenols and resveratrol pro-
tect the neonatal brain against hypoxic-ischemic injury. Dev Neurosci 2007;29:
363–72.
35. Zhang W, Hu X, Yang W, et al. Omega-3 polyunsaturated fatty acid supplemen-
tation confers long-term neuroprotection against neonatal hypoxic-ischemic
brain injury through anti-inflammatory actions. Stroke 2010;41:2341–7.
36. Ramanantsoa N, Fleiss B, Bouslama M, et al. Bench to cribside: the path for
developing a neuroprotectant. Translational Stroke Research 2013;4:258–77.
37. Welin AK, Svedin P, Lapatto R, et al. Melatonin reduces inflammation and cell
death in white matter in the mid-gestation fetal sheep following umbilical cord
occlusion. Pediatr Res 2007;61:153–8.
38. Villapol S, Fau S, Renolleau S, et al. Melatonin promotes myelination by decreasing
white matter inflammation after neonatal stroke. Pediatr Res 2011;69:51–5.
39. Robertson NJ, Faulkner S, Fleiss B, et al. Melatonin augments hypothermic neu-
roprotection in a perinatal asphyxia model. Brain 2013;136:90–105.
40. Yu X, Shacka JJ, Eells JB, et al. Erythropoietin receptor signalling is required for
normal brain development. Development 2002;129:505–16.
41. Mazur M, Miller RH, Robinson S. Postnatal erythropoietin treatment mitigates
neural cell loss after systemic prenatal hypoxic-ischemic injury. J Neurosurg Pe-
diatr 2010;6:206–21.
42. Digicaylioglu M, Lipton SA. Erythropoietin-mediated neuroprotection involves
cross-talk between Jak2 and NF-kappaB signalling cascades. Nature 2001;
412:641–7.
43. Sun Y, Calvert JW, Zhang JH. Neonatal hypoxia/ischemia is associated with
decreased inflammatory mediators after erythropoietin administration. Stroke
2005;36:1672–8.
44. Juul SE, Beyer RP, Bammler TK, et al. Microarray analysis of high-dose recom-
binant erythropoietin treatment of unilateral brain injury in neonatal mouse hip-
pocampus. Pediatr Res 2009;65:485–92.
45. Zacharias R, Schmidt M, Kny J, et al. Dose-dependent effects of erythropoietin
in propofol anesthetized neonatal rats. Brain Res 2010;1343:14–9.
46. Kumral A, Tugyan K, Gonenc S, et al. Protective effects of erythropoietin against
ethanol-induced apoptotic neurodegeneration and oxidative stress in the devel-
oping C57BL/6 mouse brain. Brain Res Dev Brain Res 2005;160:146–56.
47. Chattopadhyay A, Choudhury TD, Bandyopadhyay D, et al. Protective effect of
erythropoietin on the oxidative damage of erythrocyte membrane by hydroxyl
radical. Biochem Pharmacol 2000;59:419–25.
48. Iwai M, Cao G, Yin W, et al. Erythropoietin promotes neuronal replacement
through revascularization and neurogenesis after neonatal hypoxia/ischemia
in rats. Stroke 2007;38:2795–803.
Juul & Ferriero
130
49. Osredkar D, Sall JW, Bickler PE, et al. Erythropoietin promotes hippocampal
neurogenesis in in vitro models of neonatal stroke. Neurobiol Dis 2010;38:
259–65.
50. Kellert BA, McPherson RJ, Juul SE. A comparison of high-dose recombinant
erythropoietin treatment regimens in brain-injured neonatal rats. Pediatr Res
2007;61:451–5.
51. Gonzalez FF, Abel R, Almli CR, et al. Erythropoietin sustains cognitive function
and brain volume after neonatal stroke. Dev Neurosci 2009;31:403–11.
52. Demers EJ, McPherson RJ, Juul SE. Erythropoietin protects dopaminergic neu-
rons and improves neurobehavioral outcomes in juvenile rats after neonatal hyp-
oxia-ischemia. Pediatr Res 2005;58:297–301.
53. Iwai M, Stetler RA, Xing J, et al. Enhanced oligodendrogenesis and recovery of
neurological function by erythropoietin after neonatal hypoxic/ischemic brain
injury. Stroke 2010;41:1032–7.
54. Reitmeir R, Kilic E, Kilic U, et al. Post-acute delivery of erythropoietin induces
stroke recovery by promoting perilesional tissue remodelling and contralesional
pyramidal tract plasticity. Brain 2011;134:84–99.
55. Juul S. Erythropoietin in anemia of prematurity. J Matern Fetal Neonatal Med
2012;25:80–4.
56. van der Kooij MA, Groenendaal F, Kavelaars A, et al. Neuroprotective properties
and mechanisms of erythropoietin in in vitro and in vivo experimental models for
hypoxia/ischemia. Brain Res Rev 2008;59:22–33.
57. Juul SE, McPherson RJ, Bauer LA, et al. A phase I/II trial of high-dose erythro-
poietin in extremely low birth weight infants: pharmacokinetics and safety. Pedi-
atrics 2008;122:383–91.
58. Fauchere JC, Dame C, Vonthein R, et al. An approach to using recombinant
erythropoietin for neuroprotection in very preterm infants. Pediatrics 2008;122:
375–82.
59. Wu YW, Bauer LA, Ballard RA, et al. Erythropoietin for neuroprotection in
neonatal encephalopathy: safety and pharmacokinetics. Pediatrics 2012;130:
683–91.
60. Almeida RD, Manadas BJ, Melo CV, et al. Neuroprotection by BDNF against
glutamate-induced apoptotic cell death is mediated by ERK and PI3-kinase
pathways. Cell Death Differ 2005;12:1329–43.
61. Feng Y, Rhodes PG, Bhatt AJ. Neuroprotective effects of vascular endothelial
growth factor following hypoxic ischemic brain injury in neonatal rats. Pediatr
Res 2008;64:370–4.
62. Xiao BG, Lu CZ, Link H. Cell biology and clinical promise of G-CSF: immunomo-
dulation and neuroprotection. J Cell Mol Med 2007;11:1272–90.
63. Fathali N, Lekic T, Zhang JH, et al. Long-term evaluation of granulocyte-colony
stimulating factor on hypoxic-ischemic brain damage in infant rats. Intensive
Care Med 2010;36:1602–8.
64. Schabitz WR, Laage R, Vogt G, et al. AXIS: a trial of intravenous granulocyte
colony-stimulating factor in acute ischemic stroke. Stroke 2010;41:2545–51.
65. Popken GJ, Hodge RD, Ye P, et al. In vivo effects of insulin-like growth factor-I
(IGF-I) on prenatal and early postnatal development of the central nervous sys-
tem. Eur J Neurosci 2004;19:2056–68.
66. Kannan S, Dai H, Navath RS, et al. Dendrimer-based postnatal therapy for neu-
roinflammation and cerebral palsy in a rabbit model. Sci Transl Med 2012;4:
130ra46.
Strategies in Neonatal Brain Injury 131
... Firstly, we determined secretoneurin concentrations in preterm-born infants at different time points in the neonatal period and put them in reference with available data from term-born neonates [13]. Based on the knowledge that secretoneurin is upregulated by hypoxia and is involved in inflammatory processes [14]-factors which play crucial roles in the injury cascade of neonatal brain injury [15]-we hypothesized that secretoneurin serum concentrations are elevated in preterm infants suffering from brain injury. To explore this hypothesis, we secondly compared secretoneurin concentrations in preterm infants with and without this diagnosis on imaging modalities. ...
... As secretoneurin expression is induced by cellular hypoxia [14], these episodes are a potential stimulus for its upregulation. In addition, secretoneurin is known to be involved in inflammatory processes [14], which play a crucial role in later phases of preterm brain injury and can be associated with unfavourable outcomes [15]. ...
Article
Full-text available
Neurodevelopmental impairment is a significant complication among survivors of preterm birth. To improve outcomes, reliable biomarkers for early detection of brain injury and prognostic assessment are required. Secretoneurin is a promising early biomarker of brain injury in adults and full-term neonates suffering from perinatal asphyxia. Data on preterm infants is currently lacking. The aim of this pilot study was to determine secretoneurin concentrations in preterm infants in the neonatal period, and to assess secretoneurin's potential as a biomarker of preterm brain injury. We included 38 very preterm infants (VPI) born at <32 weeks' gestation in the study. Secretoneurin concentrations were measured in serum samples obtained from the umbilical cord, at 48 hours and 3 weeks of life. Outcome measures included repeated cerebral ultrasonography, magnetic resonance imaging at term-equivalent age, general movements assessment, and neurodevelopmental assessment at a corrected age of 2 years by the Bayley Scales of Infant and Toddler Development, third edition (Bayley-III). In comparison to a term-born reference population, VPI had lower secretoneurin serum concentrations in umbilical cord blood and blood collected at 48 hours of life. When measured at 3 weeks of life, concentrations correlated with gestational age at birth. Secretoneurin concentrations did not differ between VPI with an imaging-based diagnosis of brain injury and those without, but when measured in umbilical cord blood and at 3 weeks of life correlated with and were predictive of Bayley-III motor and cognitive scale scores. Secretoneurin levels in VPI differ from term-born neonates. Secretoneurin seems unsuitable as a diagnostic biomarker of preterm brain injury, but bears some prognostic potential and is worthy of further investigation as a blood-based biomarker of preterm brain injury.
... Treatments used for experimental HI target specific pathological injury processes, including excitotoxicity, oxidative stress, mitochondrial dysfunction and neuroinflammation [21]. Due to its success in protecting the developing brain following HI [19,22], HT became the standard therapy used for treating HIE patients. ...
Article
Full-text available
Neonatal hypoxic-ischemic (HI) encephalopathy (HIE) in term newborns is a leading cause of mortality and chronic disability. Hypothermia (HT) is the only clinically available therapeutic intervention; however, its neuroprotective effects are limited. Lactoferrin (LF) is the major whey protein in milk presenting iron-binding, anti-inflammatory and anti-apoptotic properties and has been shown to protect very immature brains against HI damage. We hypothesized that combining early oral administration of LF with whole body hypothermia could enhance neuroprotection in a HIE rat model. Pregnant Wistar rats were fed an LF-supplemented diet (1 mg/kg) or a control diet from (P6). At P7, the male and female pups had the right common carotid artery occluded followed by hypoxia (8% O2 for 60′) (HI). Immediately after hypoxia, hypothermia (target temperature of 32.5–33.5 °C) was performed (5 h duration) using Criticool®. The animals were divided according to diet, injury and thermal condition. At P8 (24 h after HI), the brain neurochemical profile was assessed using magnetic resonance spectroscopy (1H-MRS) and a hyperintense T2W signal was used to measure the brain lesions. The mRNA levels of the genes related to glutamatergic excitotoxicity, energy metabolism and inflammation were assessed in the right hippocampus. The cell markers and apoptosis expression were assessed using immunofluorescence in the right hippocampus. HI decreased the energy metabolites and increased lactate. The neuronal–astrocytic coupling impairments observed in the HI groups were reversed mainly by HT. LF had an important effect on astrocyte function, decreasing the levels of the genes related to glutamatergic excitotoxicity and restoring the mRNA levels of the genes related to metabolic support. When combined, LF and HT presented a synergistic effect and prevented lactate accumulation, decreased inflammation and reduced brain damage, pointing out the benefits of combining these therapies. Overall, we showed that through distinct mechanisms lactoferrin can enhance neuroprotection induced by HT following neonatal brain hypoxia-ischemia.
... Allopurinol, a xanthine oxidase inhibitor and free radical scavenger, has been shown to be neuroprotective following neonatal hypoxic-ischemic brain injury in many small and large animal studies 36 . Allopurinol reduces oxidative stress and therefore early administration appears to be extremely important regarding its activity following neonatal hypoxic-ischemic brain injury 10 . Currently, a randomized-controlled clinical trial (ALBINO study, clinical trials gov: NCT03162653) is investigating the additional neuroprotective effects of allopurinol in combination with therapeutic hypothermia; pre-clinical data is still lacking to prove additional neuroprotection of allopurinol in combination with therapeutic hypothermia. ...
Article
Full-text available
Intrapartum hypoxia–ischemia leading to neonatal encephalopathy (NE) results in significant neonatal mortality and morbidity worldwide, with > 85% of cases occurring in low- and middle-income countries (LMIC). Therapeutic hypothermia (HT) is currently the only available safe and effective treatment of HIE in high-income countries (HIC); however, it has shown limited safety or efficacy in LMIC. Therefore, other therapies are urgently required. We aimed to compare the treatment effects of putative neuroprotective drug candidates following neonatal hypoxic-ischemic (HI) brain injury in an established P7 rat Vannucci model. We conducted the first multi-drug randomized controlled preclinical screening trial, investigating 25 potential therapeutic agents using a standardized experimental setting in which P7 rat pups were exposed to unilateral HI brain injury. The brains were analysed for unilateral hemispheric brain area loss after 7 days survival. Twenty animal experiments were performed. Eight of the 25 therapeutic agents significantly reduced brain area loss with the strongest treatment effect for Caffeine, Sonic Hedgehog Agonist (SAG) and Allopurinol, followed by Melatonin, Clemastine, ß-Hydroxybutyrate, Omegaven, and Iodide. The probability of efficacy was superior to that of HT for Caffeine, SAG, Allopurinol, Melatonin, Clemastine, ß-hydroxybutyrate, and Omegaven. We provide the results of the first systematic preclinical screening of potential neuroprotective treatments and present alternative single therapies that may be promising treatment options for HT in LMIC.
... The developing brain is uniquely susceptible to oxidative stress and subsequent injury [1][2][3]. Hydrogen peroxide, in particular, accumulates in the neonatal, but not the adult, brain after hypoxia-ischemia (HI) [4]. Endogenous antioxidant mechanisms, such as catalase and glutathione peroxidase (GPX1), are not sufficient to prevent injury from a hypoxic-ischemic challenge. ...
Article
Full-text available
The developing brain is uniquely susceptible to oxidative stress and endogenous antioxidant mechanisms are not sufficient to prevent injury from a hypoxic-ischemic challenge. Glutathione peroxidase (GPX1) activity reduces hypoxic-ischemic injury. Therapeutic hypothermia also reduces hypoxic-ischemic injury, in the rodent and the human brain, but the benefit is limited. Here, we combined GPX1 overexpression with hypothermia in a P9 mouse model of hypoxia-ischemia (HI) to test the effectiveness of both treatments together. Histological analysis showed that WT mice with hypothermia were less injured than WT with normothermia. In the GPX1-tg mice, however, despite a lower median score in the hypothermia treated mice, there was no significant difference between hypothermia and normothermia. GPX1 protein expression was higher in the cortex of all transgenic groups at 30 min and 24 h, as well as in WT 30 min after HI, with and without hypothermia. GPX1 was higher in the hippocampus of all transgenic groups and WT with HI and normothermia, at 24 h, but not at 30 min. Spectrin 150 was higher in all groups with HI, while spectrin 120 was higher in HI groups only at 24 h. There was reduced ERK1/2 activation in both WT and GPX1-tg HI at 30 min. Thus, with a relatively moderate insult we see a benefit with cooling in the WT, but not the GPX1-tg mouse brain. The fact that we see no benefit with increased GPx1 here in the P9 model (unlike in the P7 model), may indicate that oxidative stress in these older mice is elevated to an extent that increased GPx1 is insufficient for reducing injury. The lack of benefit of overexpressing GPX1 in conjunction with hypothermia after HI indicates that pathways triggered by GPX1 overexpression may interfere with the neuroprotective mechanisms provided by HT.
... In addition, this treatment has variable efficacy in asphyxiated children and is more effective in treating larger babies than smaller babies (Wyatt et al., 2007). Thus, the complicated pathophysiology of HIE makes treatment challenging and necessitates the development of multiple approaches (Juul and Ferriero, 2014). ...
Article
Full-text available
Neonatal hypoxic-ischaemic events, which can result in long-term neurological impairments or even cell death, are among the most significant causes of brain injury during neurodevelopment. The complexity of neonatal hypoxic-ischaemic pathophysiology and cellular pathways make it difficult to treat brain damage; hence, the development of new neuroprotective medicines is of great interest. Recently, numerous neuroprotective medicines have been developed to treat brain injuries and improve long-term outcomes based on comprehensive knowledge of the mechanisms that underlie neuronal plasticity following hypoxic-ischaemic brain injury. In this context, understanding of the medicinal potential of cannabinoids and the endocannabinoid system has recently increased. The endocannabinoid system plays a vital neuromodulatory role in numerous brain regions, ensuring appropriate control of neuronal activity. Its natural neuroprotection against adult brain injury or acute brain injury also clearly demonstrate the role of endocannabinoid signalling in modulating neuronal activity in the adult brain. The goal of this review is to examine how cannabinoid-derived compounds can be used to treat neonatal hypoxic-ischaemic brain injury and to assess the critical function of the endocannabinoid system and its potential for use as a new neuroprotective treatment for neonatal hypoxic-ischaemic brain injury.
Article
Over the last 2 decades, therapeutic hypothermia has become the standard of care to reduce morbidity and mortality in neonates affected by moderate-to-severe hypoxic-ischemic encephalopathy (HIE). There is a significant interest in improving the neurologic outcomes of neonatal HIE, ranging from adjunctive therapy to therapeutic hypothermia. Importantly, the pathophysiologic mechanisms underlying HIE also affect multiple other organs, contributing to high morbidity and mortality in this patient population. This review focuses on the adjunct therapies currently under investigation to mitigate the impact of hypoxic-ischemic injury on the brain, kidneys, liver, heart, and gastrointestinal system.
Article
Cerebral palsy is a neurodevelopmental disease characterized by postural, motor, and cognitive disorders, being one of the main causes of physical and intellectual disability in childhood. To minimize functional impairments, the use of resveratrol as a therapeutic strategy is highlighted due to its neuroprotective and antioxidant effects in different regions of the brain. Thus, this study aimed to investigate the effects of neonatal treatment with resveratrol on postural development, motor function, oxidative balance, and mitochondrial biogenesis in the brain of rats submitted to a cerebral palsy model. Neonatal treatment with resveratrol attenuated deficits in somatic growth, postural development, and muscle strength in rats submitted to cerebral palsy. Related to oxidative balance, resveratrol in cerebral palsy decreased the levels of MDA and carbonyls. Related to mitochondrial biogenesis, was observed in animals with cerebral palsy treated with resveratrol, an increase in mRNA levels of TFAM, in association with the increase of citrate synthase activity. The data demonstrated a promising effect of neonatal resveratrol treatment, improving postural and muscle deficits induced by cerebral palsy. These findings were associated with improvements in oxidative balance and mitochondrial biogenesis in the brain of rats submitted to cerebral palsy.
Article
Full-text available
Therapeutic hypothermia is well established as a standard treatment for infants with hypoxic-ischemic (HI) encephalopathy but it is only partially effective. The potential for combination treatments to augment hypothermic neuroprotection has major relevance. Our aim was to assess the effects of treating newborn rats following HI injury with cannabidiol (CBD) at 0.1 or 1 mg/kg, i.p., in normothermic (37.5°C) and hypothermic (32.0°C) conditions, from 7 d of age (neonatal phase) to 37 d of age (juvenile phase). Placebo or CBD was administered at 0.5, 24, and 48 h after HI injury. Two sensorimotor (rotarod and cylinder rearing) and two cognitive (novel object recognition and T-maze) tests were conducted 30 d after HI. The extent of brain damage was determined by magnetic resonance imaging, histologic evaluation, magnetic resonance spectroscopy, amplitude-integrated electroencephalography, and Western blotting. At 37 d, the HI insult produced impairments in all neurobehavioral scores (cognitive and sensorimotor tests), brain activity (electroencephalography), neuropathological score (temporoparietal cortexes and CA1 layer of hippocampus), lesion volume, magnetic resonance biomarkers of brain injury (metabolic dysfunction, excitotoxicity, neural damage, and mitochondrial impairment), oxidative stress, and inflammation (TNFα). We observed that CBD or hypothermia (to a lesser extent than CBD) alone improved cognitive and motor functions, as well as brain activity. When used together, CBD and hypothermia ameliorated brain excitotoxicity, oxidative stress, and inflammation, reduced brain infarct volume, lessened the extent of histologic damage, and demonstrated additivity in some parameters. Thus, coadministration of CBD and hypothermia could complement each other in their specific mechanisms to provide neuroprotection.
Article
Full-text available
The consequences of perinatal brain injury include immeasurable anguish for families and substantial ongoing costs for care and support of effected children. Factors associated with perinatal brain injury in the preterm infant include inflammation and infection, and with increasing gestational age, a higher proportion is related to hypoxic-ischemic events, such as stroke and placental abruption. Over the past decade, we have acquired new insights in the mechanisms underpinning injury and many new tools to monitor outcome in perinatal brain injury in our experimental models. By embracing these new technologies, we can expedite the screening of novel therapies. This is critical as despite enormous efforts of the research community, hypothermia is the only viable neurotherapeutic, and this procedure is limited to term birth and postcardiac arrest hypoxic-ischemic events. Importantly, experimental and preliminary data in humans also indicate a considerable therapeutic potential for melatonin against perinatal brain injury. However, even if this suggested potential is proven, the complexity of the human condition means we are likely to need additional neuroprotective and regenerative strategies. Thus, within this review, we will outline what we consider the key stages of preclinical testing and development for a neuroprotectant or regenerative neurotherapy for perinatal brain injury. We will also highlight examples of novel small animal physiological and behavioral testing that gives small animal preclinical models greater clinical relevance. We hope these new tools and an integrated bench to cribside strategic plan will facilitate the fulfillment of our overarching goal, improving the long-term brain health and quality of life for infants suffering perinatal brain injury.
Article
Full-text available
The choroid plexus is the site of the blood-cerebrospinal fluid (CSF) barrier (BCSFB) and has also been considered as a possible route for peripheral immune signals and cells to transfer to the central nervous system. Infection/inflammation stimulates innate and subsequent adaptive immune responses via Toll-like receptors (TLRs). In this study, we have investigated the mRNA expression of TLRs, cytokines, and tight junction proteins in the choroid plexus in the immature brain after systemic inflammation, as well as accumulation of immune cells into the CSF. Specific ligands for TLR-1/2, TLR-3, and TLR-4 were administered to postnatal day 8 mice and mRNA expression for the targeted genes was examined in the choroid plexus. We found that mRNA for all four TLRs was detected in the choroid plexus under control conditions. Following immune stimulation, expression of all the TLRs was upregulated by their respective ligands, except for TLR-4 mRNA, which was downregulated by Pam3CSK4 (PAM; a TLR-1/2 ligand). In addition, we investigated BCSFB regulation after TLR stimulation and found that TLR-1/2 and TLR-4 activation was associated with changes in mRNA expression of the tight junction protein occludin in the choroid plexus. PAM induced choroid plexus transcription of TNF-α and resulted in the most dramatic increase in numbers of white blood cells in the CSF. The data suggest a possible mechanism whereby systemic inflammation stimulates TLRs in the choroid plexus, which may lead to disturbances in choroid plexus barrier function, as well as infiltration of immune cells through the plexus.
Article
Full-text available
Importance Perinatal infections are commonly present in preterm and very low-birth-weight (VLWB) infants and might contribute to adverse neurodevelopmental outcome. Objective To summarize studies evaluating the effect of perinatal infections on neurodevelopmental outcome in very preterm/VLBW infants. Evidence Review On December 12, 2011, we searched Medline, PsycINFO, Embase, and Web of Knowledge for studies on infections and neurodevelopmental outcome. All titles and abstracts were assessed for eligibility by 2 independent reviewers. We also screened the reference lists of identified articles to search for additional eligible studies. Preselected criteria justified inclusion in this meta-analysis: (1) the study included infants born very preterm (≤32 weeks) and/or with VLBW (≤1500 g); (2) the study compared infants with and without perinatal infection; (3) there was follow-up using the Bayley Scales of Infant Development 2nd edition; and (4) results were published in an English-language peer-reviewed journal. The quality of each included study was assessed using the Newcastle-Ottawa Scale. Findings This meta-analysis includes 18 studies encompassing data on 13.755 very preterm/VLBW infants. Very preterm/VLBW infants with perinatal infections had poorer mental (d = −0.25; P < .001) and motor (d = −0.37; P < .001) development compared with very preterm/VLBW infants without infections. Mental development was most impaired by necrotizing enterocolitis (d = −0.40; P < .001) and meningitis (d = −0.37; P < .001). Motor development was most impaired by necrotizing enterocolitis (d = −0.66; P < .001). Chorioamnionitis did not affect mental (d = −0.05; P = .37) or motor (d = 0.19; P = .08) development. Conclusions and Relevance Postnatal infections have detrimental effects on mental and motor development in very preterm/VLBW infants.
Article
Full-text available
Toll-like receptors (TLRs) are members of the pattern recognition receptor family that detect components of foreign pathogens or endogenous molecules released in response to injury. Recent studies demonstrate that TLRs also have a functional role in regulating neuronal proliferation in the developing brain. This study investigated cellular expression of TLR3 using immunohistochemistry on human brain tissue. The tissue sections analysed contained anterior and lateral periventricular white matter from the frontal and parietal lobes in post-mortem neonatal cases with a postmenstrual age range of 23.6-31.4 weeks. In addition to preterm brains without overt pathology (control), preterm pathology cases with evidence of white matter injuries (WMI) were also examined. In order to identify TLR-positive cells, we utilized standard double-labelling immunofluorescence co-labelling techniques and confocal microscopy to compare co-expression of TLR3 with a neuronal marker (NeuN) or with glial markers (GFAP for astrocytes, Iba-1 for microglia and Olig2 for oligodendrocytes). We observed an increase in the neuronal (28 vs. 17%) and astroglial (38 vs. 21%) populations in the WMI group compared to controls in the anterior regions of the periventricular white matter in the frontal lobe. The increase in neurons and astrocytes in the WMI cases was associated with an increase in TLR3 immunoreactivity. This expression was significantly increased in the astroglia. The morphology of the TLR3 signal in the control cases was globular and restricted to the perinuclear region of the neurons and astrocytes, whilst in the cases of WMI, both neuronal, axonal and astroglial TLR3 expression was more diffuse (i.e., a different intracellular distribution) and could be detected along the extensions of the processes. This study demonstrates for the first time that neurons and glial cells in human neonatal periventricular white matter express TLR3 during development. The patterns of TLR3 expression were altered in the presence of WMI, which might influence normal developmental processes within the immature brain. Identifying changes in TLR3 expression during fetal development may be key to understanding the reduced volumes of grey matter and impaired cortical development seen in preterm infants.
Article
Full-text available
Despite treatment with therapeutic hypothermia, almost 50% of infants with neonatal encephalopathy still have adverse outcomes. Additional treatments are required to maximize neuroprotection. Melatonin is a naturally occurring hormone involved in physiological processes that also has neuroprotective actions against hypoxic-ischaemic brain injury in animal models. The objective of this study was to assess neuroprotective effects of combining melatonin with therapeutic hypothermia after transient hypoxia-ischaemia in a piglet model of perinatal asphyxia using clinically relevant magnetic resonance spectroscopy biomarkers supported by immunohistochemistry. After a quantified global hypoxic-ischaemic insult, 17 newborn piglets were randomized to the following: (i) therapeutic hypothermia (33.5°C from 2 to 26 h after resuscitation, n = 8) and (ii) therapeutic hypothermia plus intravenous melatonin (5 mg/kg/h over 6 h started at 10 min after resuscitation and repeated at 24 h, n = 9). Cortical white matter and deep grey matter voxel proton and whole brain (31)P magnetic resonance spectroscopy were acquired before and during hypoxia-ischaemia, at 24 and 48 h after resuscitation. There was no difference in baseline variables, insult severity or any physiological or biochemical measure, including mean arterial blood pressure and inotrope use during the 48 h after hypoxia-ischaemia. Plasma levels of melatonin were 10 000 times higher in the hypothermia plus melatonin than hypothermia alone group. Melatonin-augmented hypothermia significantly reduced the hypoxic-ischaemic-induced increase in the area under the curve for proton magnetic resonance spectroscopy lactate/N-acetyl aspartate and lactate/total creatine ratios in the deep grey matter. Melatonin-augmented hypothermia increased levels of whole brain (31)P magnetic resonance spectroscopy nucleotide triphosphate/exchangeable phosphate pool. Correlating with improved cerebral energy metabolism, TUNEL-positive nuclei were reduced in the hypothermia plus melatonin group compared with hypothermia alone in the thalamus, internal capsule, putamen and caudate, and there was reduced cleaved caspase 3 in the thalamus. Although total numbers of microglia were not decreased in grey or white matter, expression of the prototypical cytotoxic microglial activation marker CD86 was decreased in the cortex at 48 h after hypoxia-ischaemia. The safety and improved neuroprotection with a combination of melatonin with cooling support phase II clinical trials in infants with moderate and severe neonatal encephalopathy.
Article
Neonatal encephalopathy (NE) is the clinical manifestation of disordered neonatal brain function. Lack of universal agreed definitions of NE and the sub-group with hypoxic-ischaemia (HIE) makes the estimation of incidence and the identification of risk factors problematic. NE incidence is estimated as 3.0 per 1000 live births (95%CI 2.7 to 3.3) and for HIE is 1.5 (95%CI 1.3 to 1.7). The risk factors for NE vary between developed and developing countries with growth restriction the strongest in the former and twin pregnancy in the latter. Potentially modifiable risk factors include maternal thyroid disease, receipt of antenatal care, infection and aspects of the management of labour and delivery, although indications for some interventions were not reported and may represent a response to fetal compromise rather than the cause. It is estimated that 30% of cases of NE in developed populations and 60% in developing populations have some evidence of intrapartum hypoxic-ischaemia.
Article
Besides its established function in erythropoiesis, erythropoietin (EPO) is currently also appreciated for its neuroprotective effects. The detrimental sequelae of prolonged cerebral hypoxia and ischemia have been shown to attenuate by EPO treatment. After binding to the EPO receptor, EPO is capable of initiating a cascade of events which –via different pathways– may lead to neuroprotection. The circumstances that determine which specific signalling route(s) are activated by EPO are largely unknown. We aim to provide the reader with a timely overview on the use of EPO in models of stroke and hypoxia–ischemia and to discuss the molecular events that underlie its neuroprotection.
Article
The number of births in the United States declined by 1% between 2010 and 2011, to a total of 3 953 593. The general fertility rate also declined by 1% to 63.2 births per 1000 women, the lowest rate ever reported. The total fertility rate was down by 2% in 2011 (to 1894.5 births per 1000 women). The teenage birth rate fell to another historic low in 2011, 31.3 births per 1000 women. Birth rates also declined for women aged 20 to 29 years, but the rates increased for women aged 35 to 39 and 40 to 44 years. The percentage of all births to unmarried women declined slightly to 40.7% in 2011, from 40.8% in 2010. In 2011, the cesarean delivery rate was unchanged from 2010 at 32.8%. The preterm birth rate declined for the fifth straight year in 2011 to 11.72%; the low birth weight rate declined slightly to 8.10%. The infant mortality rate was 6.05 infant deaths per 1000 live births in 2011, which was not significantly lower than the rate of 6.15 deaths in 2010. Life expectancy at birth was 78.7 years in 2011, which was unchanged from 2010. Crude death rates for children aged 1 to 19 years did not change significantly between 2010 and 2011. Unintentional injuries and homicide were the first and second leading causes of death, respectively, in this age group. These 2 causes of death jointly accounted for 47.0% of all deaths of children and adolescents in 2011.