Associate editor: M. Mouradian
Therapeutics for neonatal brain injury

https://doi.org/10.1016/j.pharmthera.2008.07.003Get rights and content

Abstract

Neonatal brain injury is an important cause of death and neurodevelopmental delay. Multiple pathways of oxidant stress, inflammation, and excitotoxicity lead to both early and late phases of cell damage and death. Therapies targeting these different pathways have shown potential in protecting the brain from ongoing injury. More recent therapies, such as growth factors, have demonstrated an ability to increase cell proliferation and repair over longer periods of time. Even though hypothermia, which decreases cerebral metabolism and possibly affects other mechanisms, may show some benefit in particular cases, no widely effective therapeutic interventions for human neonates exist. In this review, we summarize recent findings in neuroprotection and neurogenesis for the immature brain, including combination therapy to optimize repair.

Introduction

Injury to the brain during early development is a significant contributor to mortality and long-term morbidity. Perinatal brain injury is that which occurs immediately before or after delivery, while neonatal injury occurs during the perinatal period up through 4 weeks of age. Causes of early brain injury include stroke, birth trauma, metabolic or genetic disorders, status epilepticus, and a variety of asphyxial events resulting in hypoxia and ischemia (HI). HI often leads to periventricular white matter injury in premature infants, while term infants develop cortical/subcortical lesions (Gressens & Luton, 2004). Studies of perinatal stroke conservatively estimate an incidence of 1 in 4000 live births (Nelson & Lynch, 2004), whereas perinatal asphyxia resulting in encephalopathy, or hypoxic–ischemic encephalopathy, occurs in 3 to 5 in 1000 live births (Wu et al., 2004), with an increasing incidence in Western countries (Hagberg et al., 1996). While many die during the neonatal period, the majority of survivors exhibit neurological deficits that persist throughout life, such as epilepsy, cerebral palsy or mental retardation (Dilenge et al., 2001). No established therapeutic regimens exist, and treatment and care for the sequelae of early brain injury requires significant resources, often with little improvement in an individual patient's overall abilities and with long-term effects on their family and society (Ferriero, 2004).

There has long been a search for therapies that can either prevent injury progression or enhance repair of the immature brain, hopefully improving long-term motor and behavioral outcomes. Making this challenging is the fact that the neonatal and adult brain do not respond similarly to injury, with altered susceptibility to oxidative stress and excitotoxicity and differences in gene regulation during hypoxia (McQuillen & Ferriero, 2004). Damage occurs via multiple pathways, and repair occurs over a period of days to weeks, if not months (Geddes et al., 2001). Neuronal death during the newborn period appears to occur in two phases: primary neuronal death from cellular hypoxia and energy depletion, followed by reperfusion and increases in excitotoxicity, free radical formation, and nitric oxide production with secondary energy failure and delayed death (Perlman, 2006). While some therapies that manipulate these pathways show promise, not all neonates will benefit from treatment. Damage may be so severe or prolonged that repair may not be possible, or survivors may be particularly devastated (Gluckman et al., 2005). Genetic factors and gender effects may also affect susceptibility to damage (Harding et al., 2004, Kerk et al., 2006).

Optimizing therapy for neonatal brain injury will require capitalizing on multiple pathways which prevent cell death and enhance cell growth, differentiation, and long-term integration into neural networks. Classically, the term neuroprotection has been used, but should we only think about protecting neurons? Injury to other cell types, such as oligendrocytes and astrocytes, impedes development and results in long-term damage. By targeting the injury response, the goal is to utilize targeted pharmacotherapies to salvage cells that would otherwise die, protect cells from becoming injured or at risk for death by increasing tolerance, and also repairing injured cells and enhancing neurogenesis. These post-insult therapies include glutamate receptor antagonists, free radical blockers or scavengers, microglial affectors or cytokine inhibitors, altering the cell death cascade effects on apoptosis, or modifying cerebral metabolism with hypothermia. More recent evidence suggests that therapies may be combined to enhance these protective and reparative processes.

In addition to developing new approaches for neuroprotection, we need to quickly identify neonates that will benefit from treatment. A variety of clinical predictors have been used to identify those at risk for hypoxic brain injury. These include an Apgar score < 5 at 5 min, elevated cord blood or early arterial acidosis, and seizures or the presence of encephalopathy on examination (Miller et al., 2004). More recently, cerebral function monitoring using bedside amplitude integrated EEG has provided an efficient and fairly accurate means for identifying encephalopathy or prolonged seizure (Hellstrom-Westas & Rosen, 2006), but it does not replace full EEG (Shellhaas et al., 2007). Brain imaging, specifically magnetic resonance imaging (MRI) with newer techniques such as spectroscopy (MRS), diffusion weighted (DWI) and diffusion tensor imaging (DTI), and volumetric analyses provides the most accurate assessment of injury. These allow identification of severity and evolution of brain injury, with specific injury patterns being associated with poor outcomes, such as loss of gray/white differentiation, watershed injury, and thalamic or basal ganglia injury (Miller et al., 2005). However, early and sequential imaging in neonates is often not possible because of scanner availability or difficulty in transporting these critically ill patients. Biomarkers for inflammation and oxidative stress, or indicating injury to other organ systems, are currently being studied but are of equivocal value in identifying early neonatal brain injury. For example, some serum neuronal and glial proteins such as myelin basic protein, S100B, and neuron-specific enolase show promise in traumatic brain injury (Berger et al., 2007). Given all the available evidence, a combination of encephalopathic physical exam and seizures provide an indication of infants that may be at risk for brain injury (Miller et al., 2004). Early identification is vital to improve outcomes, as early therapy appears to be superior to late (Thoresen, 2000) and identification of those who may benefit from those who will not is increasingly important. This review will focus on newer developments in treating neonatal brain injury, as well as combination therapy that will potentially optimize long-term outcomes.

Section snippets

Inhibition of excitotoxicity

Glutamate plays a key role in brain development, affecting progenitor cell differentiation, proliferation, migration and survival. Excitotoxicity refers to excessive glutamatergic activation that leads to cell injury and death (Olney, 2003). Glutamate accumulates in the brain after HI (Gucuyener et al., 1999) from a variety of causes, including vesicular release from axons (Kukley et al., 2007) and reversal of glutamate transporters (Fern and Moller, 2000, Rossi et al., 2000). Excitotoxicity

Antioxidants

Oxidative stress is an important component of early injury to the neonatal brain (Ferriero, 2001). Oxygen toxicity results from excess formation of free radicals (FR) (reactive oxygen species (ROS) and reactive nitrogen species (RNS)) from oxidative metabolism that occurs under pathological conditions (Fig. 1). FRs include superoxide anion (O2radical dot), hydroxyl radical (OHradical dot), singlet oxygen (1O2radical dot) and hydrogen peroxide (H2O2) (Fridovich, 1997, Halliwell, 1999). FRs target lipids, protein and DNA,

Anti-inflammatory agents

Maternal infection is a known risk factor for white matter disease and poor outcomes, such as cerebral palsy (Dammann et al., 2002, Wu and Colford, 2000, Wu et al., 2003). The inflammatory response that accompanies infection plays a vital role in cell damage and loss, with cytokines possibly being the final mediators of both initial damage and later injury to penumbral tissue (Stirling et al., 2005). Local microglia are activated early and produce pro-inflammatory cytokines such as TNF-α, IL-1β

Cell death inhibitors

Apoptosis is critical for normal brain development, but it is also an important component of injury following neonatal HI and stroke (Northington et al., 2005) (Fig. 2). Activation of intrinsic or extrinsic apoptotic pathways leads to cleavage and activation of caspase-3, which is maximally produced in the neonatal period (Hu et al., 2000). Proapoptotic Bax is present in high concentrations during the first two postnatal weeks (Lok & Martin, 2002). While necrosis plays a major role in early

Preconditioning

Lessons about protection can be gleaned from the response to milder forms of injury. Preconditioned animals that are treated with sublethal stress are protected from subsequent insults that would otherwise be lethal (Bergeron et al., 2000, Sheldon et al., 2007). For example, P6 rats exposed to 8% hypoxia have reduced brain injury following HI that occurs 24 h after a preconditioning stimulus, with protection that persists 1–3 weeks later (Gidday et al., 1994, Vannucci et al., 1998). It is

Growth factors

EPO is a 34-kDa glycoprotein that was originally identified for its role in erythropoiesis, but has since been found to have a variety of other roles. The pleiotropic functions of this cytokine include modulation of the inflammatory and immune responses (Villa et al., 2003), vasogenic and proangiogenic effects through its interaction with VEGF (Chong et al., 2002, Wang et al., 2004a, Wang et al., 2004b), as well as effects on CNS development and repair. EPO and EPO receptor are expressed by a

Stem cell therapy

Neural stem cells (NSCs) are multi-potent precursors that self-renew and retain the ability to differentiate into a variety of neuronal and non-neuronal cell types in the CNS. They reside in neurogenic zones throughout life, such as the subventricular zone and subgranular zone of the dentate gyrus in rodent models, and help maintain cell turnover at baseline and replace injured cells by migrating to penumbral tissue after injury. NSC transplantation has shown potential as a therapeutic strategy

Hypothermia

A recent therapy gaining momentum is the use of therapeutic hypothermia for brain injury. It is postulated to work by modifying apoptosis and interrupting early necrosis (Edwards et al., 1995), reducing cerebral metabolic rate and the release of excitotoxins, NO, and FRs (Globus et al., 1995). Multiple animal models of perinatal brain injury demonstrate histological and functional benefit of early initiation of hypothermia (Gunn et al., 1998a, Gunn et al., 1997, Gunn et al., 1998b, Laptook et

Manipulating mechanisms: Combination therapy

Single therapy with many of the aforementioned interventions often results in only mild improvement. Anti-apoptotic therapies may prevent delayed cell death, but would not suppress early necrotic and excitotoxic injury. Since hypothermia has shown benefit in moderately encephalopathic newborns, it is rapidly becoming standard of care in many institutions. However, it does not completely protect or repair a brain that has been injured, so the search for adjuvant therapies continues. In addition,

Concluding remarks

The initiation and development of injury to the neonatal brain is complex, with multiple contributing mechanisms and pathways resulting in both early and delayed injury. Studies have focused on these different processes, including oxidative stress, inflammation, and excitotoxicity, which differ between the mature and immature brain. More recent evidence has focused on synthesizing therapies that attack these pathways from multiple vantage points, helping us to understand how brain injury occurs

References (226)

  • EdwardsA.D. et al.

    Specific inhibition of apoptosis after cerebral hypoxia–ischaemia by moderate post-insult hypothermia

    Biochem Biophys Res Commun

    (1995)
  • EicherD.J. et al.

    Moderate hypothermia in neonatal encephalopathy: efficacy outcomes

    Pediatr Neurol

    (2005)
  • FengY. et al.

    Inhibiting caspase-8 after injury reduces hypoxic–ischemic brain injury in the newborn rat

    Eur J Pharmacol

    (2003)
  • FerrieroD.M. et al.

    Neonatal mice lacking neuronal nitric oxide synthase are less vulnerable to hypoxic–ischemic injury

    Neurobiol Dis

    (1996)
  • ForstreuterF. et al.

    Vascular endothelial growth factor induces chemotaxis and proliferation of microglial cells

    J Neuroimmunol

    (2002)
  • FridovichI.

    Superoxide anion radical (O2−.), superoxide dismutases, and related matters

    J Biol Chem

    (1997)
  • GiddayJ.M. et al.

    Neuroprotection from ischemic brain injury by hypoxic preconditioning in the neonatal rat

    Neurosci Lett

    (1994)
  • GluckmanP.D. et al.

    Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: Multicentre randomised trial

    Lancet

    (2005)
  • GucuyenerK. et al.

    Excitatory amino acids and taurine levels in cerebrospinal fluid of hypoxic ischemic encephalopathy in newborn

    Clin Neurol Neurosurg

    (1999)
  • HamrickS.E. et al.

    A role for hypoxia-inducible factor-1alpha in desferoxamine neuroprotection

    Neurosci Lett

    (2005)
  • HanB.H. et al.

    Selective, reversible caspase-3 inhibitor is neuroprotective and reveals distinct pathways of cell death after neonatal hypoxic–ischemic brain injury

    J Biol Chem

    (2002)
  • Hellstrom-WestasL. et al.

    Continuous brain-function monitoring: state of the art in clinical practice

    Semin Fetal Neonatal Med

    (2006)
  • HicksA.U. et al.

    Enriched environment enhances transplanted subventricular zone stem cell migration and functional recovery after stroke

    Neuroscience

    (2007)
  • HiguchiY. et al.

    Increase in nitric oxide in the hypoxic–ischemic neonatal rat brain and suppression by 7-nitroindazole and aminoguanidine

    Eur J Pharmacol

    (1998)
  • AherS. et al.

    Late erythropoietin for preventing red blood cell transfusion in preterm and/or low birth weight infants

    Cochrane Database Syst Rev

    (2006)
  • AlkanT. et al.

    Neuroprotective effects of MK 801 and hypothermia used alone and in combination in hypoxic–ischemic brain injury in neonatal rats

    Arch Physiol Biochem

    (2001)
  • ArvinK.L. et al.

    Minocycline markedly protects the neonatal brain against hypoxic–ischemic injury

    Ann Neurol

    (2002)
  • AzzopardiD. et al.

    Pilot study of treatment with whole body hypothermia for neonatal encephalopathy

    Pediatrics

    (2000)
  • BagnardD. et al.

    Semaphorin 3a-vascular endothelial growth factor-165 balance mediates migration and apoptosis of neural progenitor cells by the recruitment of shared receptor

    J Neurosci

    (2001)
  • BallardR.A. et al.

    Inhaled nitric oxide in preterm infants undergoing mechanical ventilation

    N Engl J Med

    (2006)
  • BaudO. et al.

    Nitric oxide-induced cell death in developing oligodendrocytes is associated with mitochondrial dysfunction and apoptosis-inducing factor translocation

    Eur J Neurosci

    (2004)
  • BeckmanJ.S. et al.

    Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and ugly

    Am J Physiol

    (1996)
  • BendersM.J. et al.

    Early postnatal allopurinol does not improve short term outcome after severe birth asphyxia

    Arch Dis Child Fetal Neonatal Ed

    (2006)
  • BenitoC. et al.

    Cannabinoid CB1 and CB2 receptors and fatty acid amide hydrolase are specific markers of plaque cell subtypes in human multiple sclerosis

    J Neurosci

    (2007)
  • BergerR.P. et al.

    Serum biomarker concentrations and outcome after pediatric traumatic brain injury

    J Neurotrauma

    (2007)
  • BergeronM. et al.

    Role of hypoxia-inducible factor-1 in hypoxia-induced ischemic tolerance in neonatal rat brain

    Ann Neurol

    (2000)
  • BernaudinM. et al.

    A potential role for erythropoietin in focal permanent cerebral ischemia in mice

    J Cereb Blood Flow Metab

    (1999)
  • BonaE. et al.

    Protective effects of moderate hypothermia after neonatal hypoxia–ischemia: short- and long-term outcome

    Pediatr Res

    (1998)
  • BoucherJ.L. et al.

    Nitric oxide biosynthesis, nitric oxide synthase inhibitors and arginase competition for l-arginine utilization

    Cell Mol Life Sci

    (1999)
  • BrinesM.L. et al.

    Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury

    Proc Natl Acad Sci U S A

    (2000)
  • BrionL.P. et al.

    Vitamin e supplementation for prevention of morbidity and mortality in preterm infants

    Cochrane Database Syst Rev

    (2003)
  • BuonocoreG. et al.

    Free radicals and brain damage in the newborn

    Biol Neonate

    (2001)
  • BustoR. et al.

    Effect of mild hypothermia on ischemia-induced release of neurotransmitters and free fatty acids in rat brain

    Stroke

    (1989)
  • CaponeC. et al.

    Neurosphere-derived cells exert a neuroprotective action by changing the ischemic microenvironment

    PLoS ONE

    (2007)
  • CarloniS. et al.

    Melatonin protects from the long-term consequences of a neonatal hypoxic–ischemic brain injury in rats

    J Pineal Res

    (2008)
  • ChangY.S. et al.

    Erythropoietin improves functional and histological outcome in neonatal stroke

    Pediatr Res

    (2005)
  • ChenH.S. et al.

    The chemical biology of clinically tolerated NMDA receptor antagonists

    J Neurochem

    (2006)
  • ChenM. et al.

    Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington disease

    Nat Med

    (2000)
  • ChengE.T. et al.

    Functional recovery of transected nerves treated with systemic BDNF and CNTF

    Microsurgery

    (1998)
  • ChengY. et al.

    Marked age-dependent neuroprotection by brain-derived neurotrophic factor against neonatal hypoxic–ischemic brain injury

    Ann Neurol

    (1997)
  • Cited by (85)

    • Chronic Oxytocin treatment has long lasting therapeutic potential in a rat model of neonatal hypercapnic-hypoxia injury, through enhanced GABAergic signaling and by reducing hippocampal gliosis with its anti-inflammatory feature

      2021, Peptides
      Citation Excerpt :

      Hypoxia is an inadequate supply of oxygen to the cell that disrupts aerobic metabolism and can lead to mitochondrial dysfunction, apoptosis and neurodegeneration, causing serious morbidity and mortality depending on degree and localization of the tissue hypoxia. As a result of hypoxia, convulsions, cognitive dysfunction and sensorimotor dysfunction happens [1]. In addition to hypoxia, hypercapnia may contribute to this neurodegeneration via increase in perivascular pH, activation of potassium channels, influx of potassium, hyperpolarization of the endothelial cell and subsequent decrease in intracellular calcium levels in vascular smooth muscles leading to detoriation of the perfusion [2].

    • Effects of erythropoietin on neonatal hypoxia–ischemia brain injury in rat model

      2017, Physiology and Behavior
      Citation Excerpt :

      The resulting cognitive and behavioral problems became a huge burden for both the family and society [3,4]. Although there is no established intervention that fully treat HI induced perinatal brain injury, many potential therapies that may prevent injury progression and enhance repair are under investigation [5]. Erythropoietin (EPO) is a 34-kDa glycoprotein that was originally identified for its essential role in erythropoiesis.

    • Neonatology for Anesthesiologists

      2016, Smith's Anesthesia for Infants and Children, Ninth Edition
    View all citing articles on Scopus
    View full text