White matter injury in the premature infant



Chapter 4: White matter injury in the premature infant


Dalit Cayam-Rand, Steven Paul Miller



Case history


JD is a 30-week appropriate for gestational-age male infant born to a 26-year-old primigravida woman whose pregnancy was complicated by intermittent vaginal bleeding from 20 weeks’ gestation. Partial placental abruption was diagnosed and mom was placed on bedrest, given a course of steroids and treated with magnesium sulfate. At 30 weeks, she had spontaneous rupture of membranes. Within several hours, labor progressed, and the infant was delivered vaginally with delayed cord clamping. He initially cried vigorously, followed by a series of apneas in the delivery room necessitating continuous positive airway pressure (CPAP). Apgar scores were 7 and 7 at 1 and 5 minutes, respectively and he was brought to the neonatal intensive care unit on CPAP with 40% FiO2. He was treated empirically with antibiotics until cultures came back sterile. He was weaned off respiratory support at 12 days of life. The clinical course was complicated by recurrent apneas, for which he was treated with caffeine. Cranial ultrasound (US) on the third day of life showed bilateral periventricular echogenicities (Fig. 4.1). Follow-up US 1 week later showed white matter cysts (Fig. 4.2) and magnetic resonance imaging (MRI) performed at 33 weeks postmenstrual age showed cystic periventricular leukomalacia (PVL) (Fig. 4.3). Repeat MRI at term-equivalent age (TEA) demonstrated white matter volume loss and ventriculomegaly. Developmentally, at 9 months of age, he was a communicative and curious infant with significant motor delays. He was diagnosed with diplegic spastic cerebral palsy (CP) for which he received occupational and physical therapies. Follow-up at 2 years old revealed language delays requiring speech therapy and at 6 years old he has an individualized education plan including physical therapy and special education services at his school.





This case illustrates the classic evolution of severe white matter injury (WMI) in the preterm infant characterized by hyperechogenicity or “flaring,” the subsequent appearance of cystic changes, and eventually, enlarged lateral ventricles indicating progressive volume loss. While MRI is a more sensitive modality for visualizing WMI, cranial US is the most frequent bedside imaging modality used in the NICU. Recognition of the patterns of injury prior to discharge enables providers to refer to developmental follow-up and support for those at highest risk for neurodevelopmental impairments.


The scope and spectrum of white matter injury


WMI, the most common form of brain injury in preterm infants, occurs most often among infants born between 23 and 32 weeks’ gestation with a peak incidence at 28 weeks’ gestational age. The prevalence of WMI varies according to the gestational age of the cohort, as well as the timing and modality of imaging. A recent systematic review has described that up to 40% of preterm infants are born before 28 weeks’ gestational age.1 The spectrum of preterm WMI ranges from discrete lesions to diffuse volume loss and includes three pathologic forms: cystic necrosis, microscopic necrosis, and nonnecrotic lesions. The most severe injury consists of destructive lesions of all cellular elements with areas of cystic necrosis greater than 2 mm in diameter, known as cystic PVL. The incidence of these lesions has decreased substantially in the last few decades and occurs in current cohorts of very low birth weight infants at a rate of less than 5%.2,3 More commonly, microscopic foci less than 1 mm in diameter occur and evolve into areas of gliosis, commonly referred to as noncystic PVL. Interestingly, the incidence of noncystic WMI is decreasing over time in some cohorts of preterm neonates, without a clear explanation.4 In contemporary cohorts of preterm newborns, the most common form of injury is diffuse WMI. Unlike necrotic injury, diffuse WMI is caused by selective degeneration of premyelinating oligodendrocytes (pre-OLs) that fail to mature into oligodendrocytes, leading to myelination failure and secondary axonal disturbances.5


Notably, the patterns of WMI associated with very preterm infants, born before 32 weeks, are increasingly being recognized in other populations. Recent studies have demonstrated WMI in a substantial number of moderate to late preterm infants, though severe lesions are less common than at lower gestational ages.68 In a prospective study of neonates born between 32 and 36 weeks, WMI was the most frequent form of brain injury encountered, with MRI at TEA revealing diffuse injury in 23% and punctate lesions in 16%.8 Term-born infants with severe congenital heart disease demonstrate similar lesions,9,10 postulated to be a result of destructive and dysmaturational disturbances that overlap with the mechanisms in preterm infants.11 Punctate lesions have also been reported among near-term and term-born infants associated with perinatal asphyxia and genetic diagnoses.12 Nevertheless, as the hallmark brain lesion among very preterm infants, the imaging characteristics, pathogenesis, and outcomes of WMI are currently best understood in the context of prematurity as illustrated below.


Diagnosis and imaging


The evolution of preterm WMI from the first few days of life until TEA has been extensively characterized by serial cranial US imaging.13 This method has been used to diagnose cystic WMI since the 1980s due to its availability, safety, and reliability. Typically, the initial presentation of WMI on US is enhanced periventricular echogenicity. This may persist or resolve within several days; after resolution, it may then evolve into gliotic or cystic changes or, alternatively, leave no abnormality.13 Cysts may only appear after 2 to 6 weeks and may subsequently disappear due to resorption of fluid within gliotic brain tissue and can therefore be missed without sequential imaging. At TEA, US may only demonstrate ex vacuo ventriculomegaly and enlarged extra-axial spaces. Thus the number and timing of US studies are critical to consider when evaluating the diagnostic performance of this imaging modality.


When cystic WMI persists, it is associated with significant developmental abnormalities that are primarily motor impairments, classically, spastic diplegic CP. A widely utilized grading classification for the severity of cystic PVL was developed by de Vries et al.14 and increasing severity of PVL according to this classification is associated with increased incidence and severity of CP.15 Prolonged duration of periventricular hyperechogenicities, even in the absence of cystic evolution, has also been associated with adverse motor outcomes.16,17


MRI is the preferred modality to diagnose diffuse WMI. At TEA and onward, diffuse WMI may appear on MRI as ventriculomegaly, irregularly shaped ventricles, white matter loss (enlarged cerebrospinal fluid spaces surrounding the sulci), white matter tract thinning (e.g., thin corpus callosum), diffuse signal intensity changes, and myelination disturbances.18 Scoring systems for these white matter abnormalities have been developed that are inversely associated with neurodevelopmental outcomes at preschool age.1921


Many centers also perform early MRIs, at approximately 32 weeks postmenstrual age, or as soon as infants are sufficiently stable to undergo the exam. Increased use of early MRI has demonstrated more subtle focal lesions that may not be appreciated on US imaging or follow-up TEA imaging.22 Visualized soon after the insult, WMI lesions may appear as clusters of punctate hyperintense T1 signal abnormalities often accompanied by diffusion restriction.18,23,24 Linear punctate lesions with a hemorrhagic component, indicated by signal loss on susceptibility-weighted imaging, may also be seen.23,25 Clinical factors associated with punctate lesions include higher gestational age and birth weight among very preterm neonates and the presence of intraventricular hemorrhage (IVH).26,27 These multifocal WMI lesions are most readily visualized on early-life MRI. In a significant number of infants, these early lesions fade or become challenging to detect on MRI at TEA, though they are still associated with neurodevelopmental outcomes, even if harder to detect on these later age MRI scans. The change in imaging characteristics of these lesions over time supports the destructive/necrotic nature of these early lesions and not that they are transient imaging phenomena.22,28 Importantly for clinicians, both the volume and location of lesions on early-life MRI are predictive of outcomes, with anterior lesions being most concerning for motor and cognitive deficits.29,30 A summary of US and MRI findings can be found in Table 4.1.



More sophisticated imaging modalities reveal microstructural disturbances and aberrant connectivity patterns among preterm infants with WMI. Diffusion tensor imaging (DTI) shows reduced fractional anisotropy in white matter tracts, indicating delayed maturation among preterm neonates with focal lesions,31,32 including distal to the original site of injury33 and even in the absence of overt injury on conventional MRI.34 Diffuse white matter abnormalities are also associated with abnormal radial and axial diffusivities on DTI.35,36 Functional resting state MRI has also demonstrated altered connectivity among neonates with WMI.3739


WMI does not typically have a clinical correlate in the immediate neonatal period and the diagnosis is usually made by routine imaging. Guidelines from the American Academy of Pediatrics and Canadian Paediatric Society recommend performing serial cranial US in very preterm infants in the first week of life, at 4 to 6 weeks, and again prior to discharge, with minor differences between the guidelines regarding the gestational age cutoff and timing of the first US.40,41 The first US is primarily meant for the detection of IVH and the latter for WMI, although, as discussed earlier, US may miss the more prevalent diffuse WMI, better demonstrated on MRI. Some controversy exists surrounding the routine use of MRI as a screening tool at TEA given its limited overall predictive value for long-term outcomes,4244 and in current guidelines, MRI is suggested for infants with abnormal imaging findings on US. Other experts offer evidence-based indications for using routine MRI in high-risk infants in conversation with the family concerning the performance of this imaging modality to predict long-term neurodevelopment.45 It is important to note that the absence of WMI carries a particularly high negative predictive value for significant adverse outcomes,46 allowing clinicians to be reassuring when WMI is not demonstrated. And, as noted above, multifocal WMI lesions are more readily detected on early-life MRI rather than at TEA.


Pathogenesis


The central mechanisms of WMI in preterm neonates are hypoxia-ischemia and infection-inflammation, leading to oxidative stress, glutamate-mediated excitotoxicity, and the production of proinflammatory cytokines and reactive oxygen species (ROS) that are toxic to the developing white matter. The primary cellular target of injury is oligodendrocyte precursors, called premyelinating oligodendrocytes (pre-OLs). Extensive human and experimental studies have demonstrated the vulnerability of pre-OLs, the predominant white matter cells in the preterm brain during the peak window of WMI.47 Oligodendrocytes, primarily derived from radial glial cells, mature in four principal stages that have distinct morphology, immune markers, and functional characteristics.48 The earliest form of oligodendrocyte progenitors can migrate and proliferate, whereas pre-OLs are proliferative but nonmigratory, and the more mature oligodendrocytes have myelinating properties. Pre-OLs are uniquely vulnerable to oxidative stress due to maturation-dependent features, including an immature antioxidant system that renders them vulnerable to ROS.49,50 This intrinsic susceptibility is not seen in earlier and later oligodendrocyte lineage stages, or in cortical neurons,51 and it is potentiated by both hypoxia-ischemia and inflammation, which in turn, potentiate each other.52 The role of hypoxia-ischemia is postulated to be a result of disturbances in cerebral autoregulation, heart-rate-dependent cardiac output, and the immature vasculature precipitated by episodes of hypoxia, bradycardia, and hypotension.53 Infection-inflammation is mediated by microglial activation via Toll-like receptors (TLRs), as a result of common inflammatory disturbances in the preterm neonate, including sepsis and necrotizing enterocolitis (NEC).54 These mechanisms trigger a cascade of events, leading to the degeneration of pre-OLs and a subsequent increase in dysmature oligodendrocyte progenitors that are unable to fully differentiate into myelin-producing cells, causing diffuse injury characterized by impaired myelination and volume loss.55


Hypoxia-ischemia


The specific factors in the preterm neonate that predispose to hypoxia-ischemia are low basal cerebral blood flow (CBF), impaired autoregulatory mechanisms, and underdeveloped vasculature.53 These clinical issues are of particular concern given the role of repeated hypoxia-ischemia in experimental models leading to severe WMI.56 Furthermore, the regenerated pool of pre-OLs, the vulnerable cell type, leads to an ongoing vulnerability of the white matter to repeated hypoxic-ischemic events. Repeated hypoxic-ischemic events also generate an inflammatory response culminating in WMI, as well as related comorbidities, such as bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP), that predispose to WMI, further exacerbating the damage.57,58


Cerebral blood flow and autoregulation

Basal CBF in preterm infants is low, even in the absence of disturbances.59,60 CBF is impacted by changes in blood oxygen and carbon dioxide, acidosis, and hypoglycemia, common occurrences in sick preterm neonates. Under physiologic conditions, CBF is maintained by cerebral autoregulation, a mechanism that allows for relatively stable CBF across a range of systemic blood pressures. Above and below the limits of this range, CBF increases or decreases passively along with changes in systemic blood pressure, known as pressure-passive circulation. Preterm infants, especially those who are critically ill or mechanically ventilated, often display pressure-passive circulation, as well as impaired responses to metabolic derangements.6166 Cerebral autoregulation is a complex protective phenomenon involving neurogenic, myogenic, hormonal, and metabolic processes and the immature vasculature in the second and third trimesters has not developed the full capacity for autoregulation.67 Studies in preterm sheep demonstrate development of the muscular layers of arteries, critical for vasoreactive responses, at 13 weeks’ gestation, and functional autoregulation at 19 weeks’ gestation, corresponding to the human equivalent of 25 and 36 weeks’ gestation, respectively.68 This is consistent with human studies showing progression of autoregulation in the third trimester.69 Vasoreactivity is further impaired in the presence of brain injury.70,71


Developing vasculature

The vascular supply of the white matter arises from the external surface of the brain via perforating branches of leptomeningeal arteries in a centripetal fashion. These penetrating vessels include shorter subcortical branches and longer medullary branches supplying the deep white matter72 and extend in length and complexity with increasing gestational age.73 During the second trimester of gestation, these vessels may remain short with limited anastomoses, potentially leaving the periventricular deep white matter vulnerable to ischemic injury.74 The existence of an additional centrifugal vascular supply to the deep white matter via perforating branches of choroidal arteries, initially described by Van den Bergh,75 remains controversial.76 Past studies hypothesized that brain tissue at the boundary of the centripetal and centrifugal territories served as a watershed border zone that is susceptible to ischemic injury, possibly explaining the distribution and location of PVL.77,78 More recent studies dispute this on several fronts; first, anatomic studies using histological and stereoscopic methods in the past three decades have revealed these centrifugal vessels to be early subependymal veins draining the periventricular area and do not support the existence of centrifugal arteries.7983 Moreover, as demonstrated by Nelson et al.,82 in addition to the long medullary arteries originating at the gyral crests supplying the periventricular white matter, shorter medullary arteries extending from the depths of the sulci also supply these areas, giving it a rich vascular supply. Abundant arterioles from peripheral branches of medullary arteries were also found on autopsy in neonates born 20 to 41 weeks of gestation by Nakamura,79 further challenging a hypovascular border zone hypothesis.


Physiologic evidence further refuting the end artery and border zone theory can be found in animal studies on ischemia in the immature brain. McClure et al.84 measured CBF in various brain regions after ischemia-reperfusion caused by carotid occlusion and found no significant difference in CBF between the cortex and periventricular white matter during ischemia or reperfusion. Moreover, no differences were found in CBF between inferior, middle, and superior white matter regions, despite the predilection of the inferior areas to develop WMI. Areas of WMI, which localized to deeper white matter, were not associated with higher levels of ischemia, suggesting that the susceptibility of these areas is unrelated to blood flow.


Infection-inflammation


Mounting evidence in animal and human studies supports inflammatory mechanisms triggered by hypoxia-ischemia that injure the developing white matter due to its intrinsic susceptibility. Episodes of intermittent hypoxemia have been linked to local microglial activation,85 proinflammatory cytokines,86 and elevated levels of the products of lipid peroxidation.87 A small study recently showed a relationship between intermittent episodes of hypoxia and plasma CRP level, a marker of systemic inflammation.88 These inflammatory changes are associated with DTI measures and histological evidence of diffuse WMI, impaired myelination, and functional outcomes.86,8991 Thus it is important not to consider hypoxia-ischemia and inflammation as completely independent phenomena.


Evidence for a primary role for infectious and inflammatory processes in the pathogenesis of WMI initially came from autopsy studies showing increased WMI among neonates with bacteremia.92 This observation led to experimentally induced infection in kittens implicating the bacterial lipopolysaccharide endotoxin,93 later found to activate Toll-like receptor 4 (TLR4).9496 This receptor is highly expressed on microglia, but not on pre-OLs, the primary site of damage.97 The final link to WMI was elucidated with studies demonstrating the unique susceptibility of pre-OL and the developing white matter to the products of activated microglia and oxidative stress, as discussed below. These experimental findings are supported by the clinical observation of higher rates of WMI in preterm neonates with multiple infections.98


Glutamate-mediated excitotoxicity

Glutamate, the primary excitatory neurotransmitter in the central nervous system, is vital to physiologic processes in the developing brain, regulating survival, proliferation, migration, and differentiation of neural progenitor cells.99 However, when excessively accumulated in pathologic conditions, glutamate also functions as a neurotoxin.100 Following ischemia, glutamate is released from dying cells and the buildup of extracellular glutamate is toxic to adjacent pre-OLs, as first shown by Oka et al.101 This injury is mediated by both glutamate transporters and glutamate receptors.102 Glutamate shares a transporter with cystine; excess extracellular glutamate inhibits the uptake of cystine which is critical for the synthesis of glutathione, a potent endogenous antioxidant. The ensuing cellular glutathione depletion renders the cells vulnerable to free radical toxicity.101 Parallel to the transporter mechanism of injury, glutamate activates AMPA, kainate, and NMDA receptors that are expressed by developing oligodendrocytes.103105 The activation of AMPA and kainate receptors causes an influx of calcium that accumulates in the mitochondria, leading to increased production of free radicals and triggering caspases, causing apoptosis.106 Cell death then leads to additional glutamate being released, creating a feedback loop that enhances cell injury.47 The exact mechanism of injury mediated by NMDA receptors is still unclear. Notably, the discovery of receptor-mediated damage has revealed a potential therapeutic target for WMI; rodent models of preterm WMI treated with memantine, an NMDA-receptor antagonist, and topiramate, an AMPA-receptor antagonist, have shown attenuation of oligodendrocyte degeneration.107,108


The excitotoxicity of glutamate may also be mediated by astrocytes via the excitatory amino acid transporter, EAAT2, a glutamine transporter encoded by the gene SLC1A2. The transient expression of this transporter in immature oligodendrocytes has been discovered at the peak time of WMI, as well as in reactive astrocytes after hypoxic-ischemic injury109111 and may play a role in chronic white matter dysfunction.100


Activated microglia and proinflammatory cytokines

Microglia, the immune cells of the brain, play a key role in normal processes of brain development including apoptosis of excessive neurons, synaptic pruning, phagocytosis of cellular debris, and maintaining brain homeostasis.112 As such, microglia are abundant in the developing white matter during the peak window of WMI,113 including a recently discovered unique subset with myelinogenic properties.114 Although initial activation of microglia is beneficial to the brain, prolonged activation triggers neurodegeneration.115 Microglia are activated by TLRs, a family of receptors that recognize pathogen-associated molecular patterns, expressed by bacterial pathogens, as well as endogenous danger-associated molecular patterns, released after cellular stress or tissue injury.116 Microglia migrate to the pathogen or site of injury and release proinflammatory cytokines, such as tumor necrosis factor-alpha, interferon-gamma, interleukin-2, and interleukin-6 that exert a toxic effect on oligodendrocytes.117120 Elevated levels of these cytokines and their receptors have been found in the brains of preterm infants with cystic injury119,121,122 and with diffuse noncystic WMI.123


Reactive oxygen and nitrogen species

Animal and human studies implicate ROS, and reactive nitrogen species, the by-products of lipid peroxidation and protein oxidation, and biomarkers of oxidative stress, in preterm WMI.51,124126 An increase in ascorbyl radicals was found in the brains of fetal sheep exposed to intrauterine asphyxia and reperfusion125 and several studies have shown elevated isoprostanes, a marker of lipid peroxidation, among preterm neonates with WMI.51 ROS are kept in check by endogenous antioxidants, such as superoxide dismutase, catalase, and glutathione peroxidase. The predilection of preterm periventricular white matter for damage due to ROS may be related to delayed expression of some of these enzymes. The expression of superoxide dismutases one and two are significantly lower in preterm oligodendrocytes at 20 to 29 weeks, rendering them susceptible to free radical damage and ill-equipped for the oxygen-rich postnatal environment.127 This vulnerability, unique to periventricular white matter, was shown by Back et al., who found elevated levels of isoprostanes in white matter lesions, but not in the cortex.51 This biomarker for oxidative injury has been shown to be predictive of WMI, but not predictive of 2-year outcomes, highlighting the complexity of factors relating to outcomes.128


Chronic diffuse WMI


The final common pathway for these mechanisms of injury is the degeneration of pre-OLs and subsequent myelination failure. In severe focal lesions, myelination failure results from pan-cellular necrosis causing degeneration of glial cells and axons within the foci of injury, as initially described by Banker and Larroche,129 as well as axonal injury in surrounding gliotic areas beyond the regions of necrosis.130 The immature unmyelinated axons demonstrate a vulnerability to ischemia that is similar to pre-OLs131 and the severity of axonopathy seems to be related to the severity of WMI necrosis.132


In the more prevalent chronic form of diffuse nonnecrotic WMI, myelination failure is not mediated by severe axonal injury, but rather by dysmaturation. This milder form of injury is characterized by the acute degeneration of pre-OLs that is accompanied by inflammatory astrocytosis and microgliosis. Interestingly, the loss of pre-OLs also stimulates a reactive proliferation of oligodendrocyte progenitor cells, resulting in a paradoxical increase in the pre-OL population.133 Human and animal studies have shown a marked increase in progenitor cells primarily within WMI lesions, with a small increase also in the subventricular zone, the transient fetal structure where they are naturally generated.133135 These progenitors give rise to a large pool of regenerated pre-OLs in the injured areas; however, these pre-OLs are unable to differentiate into myelinating cells.5 The mechanisms behind their disrupted maturation are still being elucidated but seem to be related to the surrounding astrogliosis and microgliosis in the damaged extracellular matrix. Reactive astrocytes in the extracellular matrix secrete hyaluronic acid and the products of hyaluronic acid degradation by specific hyaluronidases inhibit the maturation of pre-OLs.136138 Increased levels of hyaluronic acid have been found in preterm sheep models of WMI and reactive astrocytes with the hyaluronic acid receptor CD44 have been found in human preterm WMI.55,56 The accumulation of selective bioactive hyaluronic acid fragments resulting from this degradation induce signal transduction via TLRs to attenuate the transcription of myelin basic protein, thereby impeding myelination. Other proposed mechanisms for myelination failure include interactions between CD44 and hyaluronic acid that involve recognizing and producing these fragments, independent of signaling.139


Data from studies on adult demyelinating diseases have revealed additional potential mechanisms for dysmaturation involving signaling pathways such as Notch and Wnt-beta catenin that disrupt oligodendrocyte maturation,140 as well as epidermal growth factor receptor (EGFR) signaling that promotes maturation.141 The Wnt signaling pathway was subsequently found to be involved in neonatal WMI mediated by hypoxia-inducible factor alpha (HIFα), critical for angiogenesis and myelination.142,143 More recently, HIFα was also found to regulate myelination, independent of this signaling pathway.144 Elucidating the exact mechanisms is crucial for developing targeted treatments. In contrast to irreversible necrotic injury with resultant death of all cellular components, dysmaturation seems to be a form of regeneration and plasticity that may be potentially modifiable and reversible. Novel treatments that have been investigated include hyaluronidase blockers that have been shown to promote pre-OL maturation145 and a selective EGFR ligand administered intranasally to mice with diffuse WMI that promotes recovery and maturation of oligodendrocytes.146


Clinical risk factors for WMI


Numerous neonatal complications, including comorbid conditions and NICU procedures and therapies, are associated with neonatal WMI. Hemorrhagic infarcts of deep white matter that accompany IVH and evolve into porencephalic cysts are discussed in the chapter on IVH. As noted above, the prevalence of WMI is higher in neonates with IVH for a given gestational age at birth. White matter dysmaturation can also occur following IVH, via the release of blood products such as iron and thrombin that induce oxidative stress, inflammation, and glutamate excitotoxicity.147,148


Neonatal infections are highly associated with WMI in multiple studies.98,149152 This association has been observed in culture-positive infections from the cerebrospinal fluid,152 as well as from sites remote from the brain (blood and tracheal cultures),151 and in culture-negative clinical sepsis.153 Regarding the type of WMI, cystic PVL has been linked with neonatal sepsis,154 while noncystic progressive WMI has been linked to recurrent infections in the neonatal period.149 Multiple infections (≥3) have also been associated with indices of dysmaturation in white matter tracts on DTI in the absence of overt injury on conventional MRI.98 The pathogenesis is related to proinflammatory cytokines and activated microglia via expression of TLRs as discussed above.54,155


NEC has also been associated with an increased risk of WMI, particularly when it requires surgical management.156158 The mechanism of NEC-related brain injury, long suspected to be mediated by the innate immune response and TLRs,54,97 has only recently begun to be elucidated in mouse models, shedding new light on the gut-brain signaling axis. Nino et al.159 discovered the release of proteins functioning as TLR ligands in the intestine that promote microglial activation and accumulation of ROS in the brain. This group of researchers subsequently demonstrated a key role for intestinal CD4+ T-lymphocytes in inducing brain injury via release of interferon-gamma and microglial activation.160 Importantly, inhibition of these lymphocytes and neutralization of interferon-gamma with targeted antibodies were able to prevent WMI, revealing a potential therapeutic avenue to avert brain injury in neonates with NEC.


Severe ROP is associated with altered microstructural integrity and delayed maturation of white matter tracts, predominantly in the posterior white matter.161163 These changes are predictive of lower motor and cognitive scores at 18 months old.162 The pathogenesis is postulated to be linked to levels of insulin growth factor one,164 an anabolic hormone which exerts effects on the retina and the brain, but the exact mechanisms have yet to be clarified. Other hypotheses include a role of systemic inflammation and hyperoxia, as recently reviewed by Morken et al.165


BPD or prolonged mechanical ventilation is associated with a spectrum of WMI and delayed maturation of white matter at TEA.166170 Several underlying factors seem to be related to adverse white matter development in BPD including duration of mechanical ventilation,171,172 and precursors to BPD, such as cumulative supplemental oxygen and mean airway pressure in the first few weeks of life, which are independently associated with adverse neurodevelopmental outcomes.173 Respiratory disturbances that are often associated with mechanical ventilation such as hypocarbia and hyperoxia are also linked to WMI.174176


There is conflicting evidence to support a role for chorioamnionitis as a risk factor for WMI. Intrauterine inflammation leading to the release of proinflammatory cytokines and fetal immune response syndrome is related to neonatal morbidities, such as postnatal infection, IVH, and adverse outcomes, that are also associated with WMI,177179 but several studies have been unable to show an independent association between chorioamnionitis and WMI.171,180182


Outcome


Given the function of myelinated white matter in the rapid transmittal of information across neural networks, it is not surprising that WMI can have a significant and broad impact on motor, cognitive, and social development. Motor-wise, white matter enables efficient and accurate communication between brain regions that include the motor cortex, basal ganglia, and cerebellum; intact myelination between these areas is vital for motor skills requiring fluency, coordination, and automatization. Accordingly, WMI is associated with a spectrum of motor deficits of varying severity, ranging from developmental coordination disorder (DCD) to CP, with more severe imaging abnormalities predicting more severe impairment.18,46,183,184


Although the incidence of the severe lesion of cystic WMI has decreased, when it occurs, it remains highly predictive of CP. In a recent population-based study over two decades in Canada, approximately 80% of infants with this injury were diagnosed with CP,3 consistent with other studies.185187 In the absence of cystic injury, myelination disturbances in the corticospinal tract are also associated with CP.188,189 Clinically, motor abnormalities predictive of CP may be detected early in infancy; in the first few weeks of life, abnormal general movements, including the absence of normal fidgety movements, are highly sensitive and specific for the subsequent diagnosis of CP.190 In preterm infants, these aberrant patterns of general movements are associated with WMI on TEA-MRI, white matter microstructural abnormalities, lower regional white matter volumes, and adverse motor and cognitive outcomes.191,192 Diffuse white matter microstructural abnormalities, in the absence of gross abnormalities on conventional MRI, are also associated with childhood DCD. Clinically, DCD manifests as difficulty learning and executing coordinated motor movements and reduced fluency in motor skills and the neuroanatomical correlates of these deficits last into childhood.193,194


Several studies have demonstrated increased rates of long-term cognitive impairments at school age among preterm-born children with overt WMI and, as with motor outcomes, the severity of injury is related to the severity of impairment.21,195199 Microstructural disturbances on DTI, even in the absence of lesions on conventional imaging, are also associated with cognitive outcomes.200 Specifically, impairments in complex skills such as executive functions, responsible for cognitive flexibility, self-control, and working memory are associated with white matter microstructural abnormalities among children born preterm who suffered inflammatory neonatal complications, such as BPD, NEC, and sepsis.201 The link between executive dysfunction and inflammatory complications is bolstered by the association between elevated neonatal inflammatory markers in the first month of life and executive function deficits at age 10 years.202 These impairments may explain the increased rates of attention deficit hyperactivity disorder seen in the preterm population.203


The fundamental role of white matter in these higher-order cognitive skills has been gaining recognition204,205 and is likely related to connectivity and network efficiency.206208 The development of the connectome between 30 and 40 gestational weeks is heavily mediated by white matter microstructure209 and microstructural disturbances among preterm-born children result in impaired connectivity and network efficiency.210 These disturbances may also underlie the higher prevalence of autism spectrum disorder (ASD) and anxiety disorders that are seen among children born preterm.211,212 Long-distance connectivity along several white matter tracts, such as the superior longitudinal fasciculus, is integral to the normal development of social cognition and theory of mind.207 Disruptions to the integrity of these tracts are commonly found in children with ASD213,214 and aberrant fractional anisotropy values in white matter tracts at 6 months of age can be found among infants who are subsequently diagnosed with ASD.215 Among preterm-born children, frank WMI on conventional MRI at TEA has been associated with ASD216 and animal models of preterm diffuse WMI provide evidence of inflammatory mechanisms mediating autistic behaviors in mice, mimicking human ASD.217


Cognitive outcomes are also likely mediated by grey matter changes that may accompany WMI. MRI studies have shown reduced cortical and thalamic volumes among children with WMI218220 and reduced caudate neuronal maturation observed experimentally with hypoxia-ischemia.221 Furthermore, the interaction between early neonatal WMI and diminished childhood thalamic volumes has been associated with cognitive outcomes at school age.220 Among neonates with PVL, pathology studies have demonstrated gliosis and neuronal loss in the thalamus222 and DTI studies have demonstrated abnormal microstructure both within the thalamus and in white matter tracts that is associated with diminished thalamic volumes.219,223,224 Animal models of diffuse WMI have attributed grey matter loss to a reduction in the complexity of the dendritic armor and reduced synaptic activity in cortical neurons and corticothalamic pathways.221,225227


Cognitive outcomes are also modified by environmental and genetic influences that are not readily accessible on imaging. The substantial impact of socioeconomic status on outcomes228 suggests potential interventions to improve outcomes, as well as important limitations for predicting outcomes with imaging alone.


Treatment


There is currently no effective treatment to prevent or to cure WMI and therefore mitigate its long-term consequences. In the neonatal period, avoiding associated risk factors for WMI, such as ventilatory complications, neonatal infection, and NEC, is key. When WMI is diagnosed on imaging, surveillance programs for early intervention are put in place to identify any neurodevelopmental impairments and start physical, occupational, and speech and language therapies. These programs have demonstrated a positive influence on outcomes at preschool age.229


The more prevalent chronic diffuse WMI that is seen in contemporary preterm populations opens a window for possible therapeutic strategies to mitigate or prevent the maturation and myelination failure that progressively occurs over many weeks. Potential interventions include treatments targeting inflammation and the immune response. Erythropoietin has been investigated over the last decade as a neuroprotective agent due to its antioxidant and anti-inflammatory effects and as a promotor of neurogenesis. In animal models of neonatal WMI, it prevents pre-OL death and promotes pre-OL development.230,231 Randomized trials subsequently showed improved white matter development and better cognitive outcomes in preterm infants who received erythropoietin.232234 A recent randomized, double-blind trial of prophylactic erythropoietin treatment did not show differences in outcome between the placebo and erythropoietin groups.235 This finding is surprising given the mechanisms of WMI outlined above; as such, erythropoietin as a potential neuroprotection strategy requires further attention (e.g., longer duration of therapy). Other potentially promising interventions in preclinical trials include anti-inflammatory treatments, such as tumor necrosis factor antagonists, associated with reduced gliosis among preterm sheep exposed to inflammation,236 and anti-interleukin-1 monoclonal antibodies, that reduce brain injury in fetal sheep exposed to ischemia.237


Supportive measures such as providing adequate nutrition and nutrient intake have also been shown to improve outcomes and diminish brain injury in preclinical and some clinical studies.238 Specifically, studies on full-term infants suggest that iron may improve myelination; a recent randomized controlled trial showed greater myelin content at 1 year of age among term-born infants who underwent delayed cord clamping associated with higher ferritin levels at 4 months of age.239 The long-term outcomes of adequate nutritional intake are still being evaluated.238 Preventing painful procedures is also an important component of improving brain maturation and neurodevelopmental outcomes.240 Painful procedures are associated with impaired white matter development that persists to school age and is linked with cognitive outcomes.241243 Optimal strategies to measure pain and provide analgesia in neonates require further investigation.


Gaps in knowledge



Stay updated, free articles. Join our Telegram channel

Mar 23, 2024 | Posted by in NEUROLOGY | Comments Off on White matter injury in the premature infant

Full access? Get Clinical Tree

Get Clinical Tree app for offline access