Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 1.
Published in final edited form as: Nat Rev Neurol. 2021 Jan 27;17(4):199–214. doi: 10.1038/s41582-020-00447-8

White matter injury in infants with intraventricular haemorrhage: mechanisms and therapies

Praveen Ballabh 1,2,, Linda S de Vries 3,4
PMCID: PMC8880688  NIHMSID: NIHMS1679935  PMID: 33504979

Abstract

Intraventricular haemorrhage (IVH) continues to be a major complication of prematurity that can result in cerebral palsy and cognitive impairments in survivors. No approved therapy exists to prevent IVH or to treat its consequences. IVH varies in severity and can present as a bleed confined to the germinal matrix, small-to-large IVH or periventricular haemorrhagic infarction. Moderate-to-severe haemorrhage dilates the ventricle and also damages the periventricular white matter. This white matter injury (WMI) results from a constellation of blood-induced pathological reactions, including oxidative stress, glutamate excitotoxicity, inflammation, perturbed signaling pathways and remodeling of the extracellular matrix. Potential therapies for IVH are currently undergoing investigation in preclinical models, and evidence from clinical trials suggests that stem cell treatment and/or endoscopic removal of clots from the cerebral ventricles could transform the outcome of infants with IVH. This Review presents an integrated view of new insights into the mechanisms underlying WMI in premature infants with IVH and highlights the importance of early detection of disability and immediate intervention in optimizing the outcomes of IVH survivors.

Introduction

Germinal matrix haemorrhage (GMH) and intraventricular hemorrhage (IVH) are the most common neurological disorders of preterm infants, occurring in about 12,000 infants every year in the USA.1,2 The major neurodevelopmental sequelae of IVH include cerebral palsy, cognitive deficits and hydrocephalus. With technological and scientific advances in perinatal and neonatal care, the survival of extremely premature infants (<1,000 g birth weight) has improved remarkably and neonatal morbidity profiles in IVH survivors have shifted towards milder neurocognitive impairments.3,4 The rate of severe cerebral palsy has declined, whereas the rate of mild cerebral palsy has increased.5

IVH typically originates in the germinal matrix (also known as the ganglionic eminence) of premature newborn babies at 23–32 weeks gestational age (Fig. 1). As a germinal matrix bleed enlarges, the underlying ependyma breaks, filling up the ventricle with blood. The diagnosis of IVH is made on screening cranial ultrasonography, which is performed in extremely premature infants (≤32 weeks gestation) in neonatal intensive care units (NICU) during the first week of life. Most of these infants are asymptomatic during their stay in the NICU, although some might manifest subtle abnormalities in consciousness, movement, tone, respiration or eye movements.

Figure 1. Germinal matrix and intraventricular haemorrhage in human preterm infants.

Figure 1.

a | Grade I germinal matrix haemorrhage. Microscopic image of hematoxylin-eosin stained section showing haemorrhage (arrowheads) confined to the germinal matrix and not extending to the ventricle in a 25-week preterm infant at postnatal day 3. Arrows indicate the ependymal layer. b | Grade II intraventricular haemorrhage (IVH). Coronal section of the forebrain of a 25-week infant showing blood (block arrows) in the germinal matrix and lateral ventricle. c | Grade II IVH. Coronal section of the forebrain of a 24 week infant with IVH and ventricular dilatation (red arrows). d | Grade IV periventricular haemorrhagic infarction (PHVI). Coronal section of the forebrain of a 28 week preterm infant revealing IVH with ventricular dilatation of ventricle (red arrow) as well as an infarct dorsal to the ventricle (PVHI, block arrow).

The severity of IVH is variable and is classified according to the ultrasound scan findings as haemorrhage confined to the germinal matrix (grade I), IVH without ventricular dilatation (grade II) or IVH with acute ventricular dilatation (grade III).6,7 The presence of an intraparenchymal echodense region is often referred to as grade IV IVH; however, such lesions usually represent periventricular haemorrhagic infarction (PVHI), which is attributed to compression of the medullary veins draining the white matter rather than the consequence of IVH extension.6,8,9 Accumulation of blood in the cerebral ventricles of infants with IVH also damages the adjacent white matter. Autopsy studies of brain samples have revealed that 50–78% of premature infants with IVH show evidence of white matter injury (WMI).1012 About 45–50% of infants with moderate-to-severe IVH survive, whereas more than 80% of infants with grade I–II IVH survive.13,14

In the USA, after discharge from the NICU, infants with IVH are followed in the neonatal follow-up clinic and in publicly funded Early Intervention Programmes for disabled children. Survivors of IVH often develop neurodevelopmental impairments, including motor, cognitive, speech, hearing and visual deficits. About 15% of all survivors of IVH develop cerebral palsy and 27% have moderate-to-severe neurosensory impairments at 18–24 months of age.13 Morbidity is increased/high in infants with moderate-to-severe IVH and about one-half to two-thirds of them develop features of hydrocephalus, cerebral palsy, cognitive deficits and/or intellectual disability.1517 About one-quarter of non-disabled survivors of IVH develop behavioural disorders and problems with executive function in infancy and childhood.1517 As these infants grow into childhood and adolescence, they often show symptoms of neuropsychiatric disorders such as attention deficit hyperactivity disorder (ADHD), major depressive disorder episodes, obsessive-compulsive disorders and epilepsy.1618 Studies of the psychiatric sequelae of IVH in adolescents who developed IVH following preterm birth show that these children have an increased risk of ADHD and tic disorders at 6 years of age, as well as an increased risk of major depressive disorder and obsessive-compulsive disorder at 16 years of age.18,19 Parenchymal lesions and ventricular enlargement exacerbates the risk of these neurobehavioural deficits.18 Thus, IVH adversely affects brain maturation and the trajectory of growth and development. Despite the high incidence of this disorder and its high societal and financial burden, the treatment of disabled survivors of IVH is currently limited to physical, occupational and speech therapy aimed at optimizing their motor, cognitive and communication skills.

In this Review, we discuss novel mechanisms of WMI in IVH as well as the role of neuroimaging in diagnosis of IVH and prediction of IVH outcome. Up-to-date neurodevelopmental data are included to illustrate the need for early diagnosis of IVH and timely intervention. Mechanism-based strategies successfully employed in preclinical models and ongoing clinical trials to improve the neurological outcome of these infants are also discussed.

Patterns of IVH-induced injury

The major neuropathological findings associated with IVH include germinal matrix damage, acute and post-haemorrhagic dilatation of the lateral ventricles and PVHI.6,20,21 On gross examination of the brain at autopsy, blood clot(s) can be seen in the ventricle, germinal matrix or brain parenchyma of infants who lived only for a short period after the onset of IVH. In infants who survive, resorption of the clot occurs over several weeks. A small clot in the germinal matrix is often replaced by a germinolytic cyst, whereas PVHI evolves into either a single cavity communicating with the lateral ventricle or multiple cysts that remain separate from the lateral ventricle. Dilatation of the lateral ventricles indicates post-haemorrhagic ventricular dilatation (PHVD).

Typical microscopic findings during the early stage of IVH evolution are apoptosis, necrosis and cerebral oedema along with infiltration of leukocytes and microglia. Subsequently, scarring and astrogliosis develop, and reactive astrocytes, haemosiderin-laden macrophages and calcification are seen in the periventricular white matter and germinal matrix.11,20 Immunostaining reveals an increased number of non-myelinating pre-oligodendrocytes, a diminished number of mature oligodendrocytes, and hypomyelination of the white matter at near-term age (35–42 weeks’ gestation).22 Haemosiderin and iron deposition can be observed using specialized stains.

PVHI is characteristically observed in infants with large IVHs and has been reported in 15% of all infants with IVH (Fig. 2).20 This haemorrhagic necrosis of periventricular white matter is typically unilateral and asymmetrical. PVHI can involve the ventricular and/or subventricular zone and adjacent white matter of mostly the frontoparietal lobe.23 These infarcts subsequently evolve into a single porencephalic cyst or multiple periventricular cysts. The pathogenesis of PVHI is attributed to compression of the terminal vein and its tributaries, which drains the cerebral white matter and germinal matrix.6,20 In one postmortem case series of infants who died with IVH, 75% had associated WMI.10 WMI is often haemorrhagic in infants with IVH.10,24 Ischaemic WMI and pontine neuronal necrosis frequently accompany IVH, but are not necessarily caused by it.25

Figure 2. Sequential neuroimaging studies of a preterm infant with bilateral intraventricular haemorrhage and periventricular haemorrhagic infarction.

Figure 2.

a | Cranial ultrasound (coronal view) shows a bilateral germinal matrix haemorrhage (arrowheads) and periventricular haemorrhagic infarction (PVHI), seen as a large echogenic lesion in the white matter (arrows) in a preterm infant of 29 weeks gestational age. b | A T2-weighted MRI sequence performed on day 10 confirmed the PVHI (arrows) and also shows some abnormalities (arrowhead) in the white matter on the contralesional side. c | A subsequent T1-weighted MRI scan performed at term-equivalent age shows an area of cavitation (arrows) and asymmetry of myelination of the posterior limb of the internal capsule (PLIC). d | A direction-encoded collar map confirms asymmetry of the PLIC (seen as blue, rostral-caudal direction), with lower fractional anisotropy suggestive of reduced myelination on the affected side.

The presence of IVH causes axonal degeneration and damages oligodendroglial progenitor cells (OPCs) resulting in reduced myelination of the white matter26. Accordingly, MRI studies have shown evidence of reduced myelination in premature infants with IVH compared to infants without IVH.27,28 Diffuse axonal injuries have been reported in the white matter adjacent to large necrotic lesions in human infants.29 Consistent with these human studies, studies in rabbit and rat models of IVH have demonstrated reduced myelination and the presence of axonal degeneration in the white matter of preterm animals with IVH relative to controls without IVH (Fig. 3).3032

Figure 3. Degenerating axons and reduced numbers of myelinated axons in an animal model of IVH.

Figure 3.

a | Representative brain coronal section immunolabeled with amyloid-β precursor protein (APP)-specific antibody showing swelling along contiguous axons (varicosity, arrowhead) or terminal bulbs suggesting axonal damage in periventricular corona radiata of premature rabbit kits (embryonic day (ED) 29; term 32 days) with intraventricular haemorrhage (IVH) at postnatal day 1 and day 3, but not in day 3 controls without IVH. b | Fluoro-Jade-C-labelled coronal section showing neurofilaments and varicosities (arrowheads) in the periventricular corona radiata of prematurely born rabbit kits with IVH at postnatal day 7 and day 14, but not in day 14 controls without IVH. c | Electron micrograph showing fewer myelinated axons (yellow arrows) in the corona radiata of a kit with IVH compared to a control kit without IVH, both at postnatal day 14. Features of axonal degeneration can also be seen in the corona radiata of the kit with IVH, including an intra-axonal vacuole (orange arrowhead), granular disintegration to total loss of microtubules and rupture of the axolemma (black arrows). Scale bar 1μm.

Pathogenesis

Inducers of white matter injury

Collection of blood into the lateral ventricle destroys the periventricular germinal matrix, damages the corpus callosum and corona radiata of the white matter and causes mass effects owing to compression of periventricular structures. IVH increases intracranial pressure, reduces cerebral blood flow and cerebral oxygen metabolism, and also seems to disrupt the blood–brain barrier and permeability of the blood–CSF barrier.33,34 The haematoma formed in the cerebral ventricles and brain parenchyma releases thrombin, complement, haem and iron, which are toxic to the brain (Fig. 4). Early brain injury is produced by plasma components of the blood, including thrombin, complement, immunoglobulins and other bioactive molecules. Subsequently, the cerebral injury is exacerbated by lysis of red blood cells, which releases cytotoxic haemoglobin and iron.35 The toxicity of these blood components has been demonstrated in both cell culture experiments and in adult animal models of intracranial haemorrhage.35,36

Figure 4: Mechanisms and treatment options for white matter injury.

Figure 4:

Intraventricular haemorrhage (IVH) results in the collection of blood in cerebral ventricles, clot lysis and the subsequent release of haemoglobin, iron, thrombin and complement. These blood products induce oxidative stress, glutamate excitotoxicity and inflammation, which are the main pathological reactions that result in damage to oligodendrocyte progenitor cells (OPCs), reduced myelination. In addition, IVH causes mass effects that compress the brain region around the ventricle and damage the blood–brain barrier. Therapeutic strategies to improve myelination have been classified into three categories: removal of blood products; inhibition of inflammation, glutamate excitotoxicity and oxidative stress; and promotion of proliferation and maturation of OPCs. BMP, bone morphogenetic protein; COX2, cyclo-oxygenase 2; TNF, tumour necrosis factor.

Among the products of clot lysis, thrombin and iron have been widely studied and seem to be the key inducers of brain injury in neonatal and adult animal models of IVH.3740 Thrombin is an enzyme principally known for its role in blood coagulation. However, thrombin also has chemotactic properties that induce inflammation and mitogenic effects that stimulate cell growth. For example, thrombin stimulates both inflammation and brain injury by activating proteinase-activated receptors (PARs) 1, 2, and 4. PAR1 is expressed on OPCs and inhibition of PAR1 both reduces inflammation and promotes myelination in the spinal cord of a neonatal mouse model of IVH.41 Thrombin injection into the cerebral ventricle of neonatal rats on postnatal day 2 (P2) induces apoptotic neural cell death in the periventricular regions and dilatation of the cerebral ventricles.36,42 The induction of ventricular enlargement by thrombin has been attributed to a loss of cadherin in the choroid plexus and increased CSF production owing to activation of PAR1 receptors.42

Lysis of red blood cells releases haemoglobin into the extracellular space, which is partly scavenged by haptoglobin and endocytosed by macrophages. Haemoglobin is toxic to the brain, and treatment with haemoglobin scavengers reduces tissue injury and periventricular inflammation in preterm rabbit kits with IVH.43,44 Haemoglobin induces cerebral oedema and injury in adult rats, via activation of haeme oxygenase 1 and by release of its degradation products.45 Non-scavenged haemoglobin is metabolized to haeme and then to iron. Both haeme and iron trigger the Fenton reaction, which is generation of highly reactive hydroxyl radicals from hydrogen peroxide. This leads to oxidative damage of neurons and glia.46,47 Indeed, iron chelation therapy can reduce the toxic effects of reactive oxygen species (ROS) on cultured oligodendrocytes and has offered neuroprotection in both neonatal and adult rat models of IVH.47,48,49

Complement activation leads to formation of the membrane attack complex (C5b–9), which can damage neurons and glia. Complement components C3a and C5a are potent chemoattractants that trigger cerebral inflammation in adult models of IVH.50 Although the effects of C3a have not been studied in neonatal models of IVH, experiments in a hypoxic-ischaemic brain injury model suggest that the effect of C3a is complex and dependent on context, timing and developmental stage.51 The presence of C3a induces neutrophil infiltration, inflammatory brain injury and expansion of infarct size.52 Conversely, C3a signalling promotes neuroplasticity by enhancing neurogenesis, axonal regeneration and synaptogenesis in animal models of hypoxic-ischaemic brain injury.5355 Lipid lysophosphatidic acid (LPA) is another bioactive molecule found in the blood that works though the G-protein-coupled receptors LPAR1–LPAR6.56 Studies in a mouse model of IVH have shown that the presence of LPA in blood contributes to the development of post-IVH hydrocephalus via activation of LPAR1.57,58 Moreover, LPA inhibits the survival and maturation of OPCs and might exacerbate WMI.59

Together, a number of reports have highlighted the roles of thrombin and iron in inducing brain injury in both adult and neonatal animals with IVH. However, no studies have addressed the effects of neonatal IVH on complement activation, blood–brain barrier damage and brain–CSF barrier disruption.

Effects on OPCs

At 23–28 weeks gestational age, the human cerebral cortex is populated mostly by early and late OPCs, which are pre-myelinating cells.47 At around 30–32 weeks, a wave of OPC differentiation is observed, which is characterized by a three-fold increase in myelinating OPCs and a marked increase in myelinated axons.60,61 Infants born prematurely during the developmental window of OPC maturation (23–28 weeks) are at the highest risk of IVH and are vulnerable to IVH-triggered OPC injury. As both IVH and OPCs originate in the lateral and medial ganglionic eminence,62 IVH exposes early and late oligodendrocyte progenitors (pre-oligodendrocytes) to blood-induced injury. Studies in preterm rabbit kits have shown that IVH induces apoptosis of pre-oligodendrocytes (Fig. 5) and reduces their proliferation26,63 In addition, preterm animals with IVH have an abundance of pre-oligodendrocytes (pre-myelinating) and paucity of immature and mature oligodendrocytes (myelinating), which suggests that maturation of OPC is arrested by IVH at the pre-oligodendrocyte stage.26 A similar arrest of OPC maturation resulting in hypomyelination is observed in neonatal models of hypoxic-ischaemic brain injury and in extremely preterm human infants.22,64 By contrast, enhanced OPC proliferation is seen in rodents with hypoxic-ischaemic brain injury.22 In conclusion, reduced myelination in IVH is attributed to increased apoptosis, reduced proliferation and arrested maturation of OPCs, which causes a dearth of myelinating oligodendrocytes and leads to myelination failure of the white matter (Fig. 5).

Figure 5: IVH induces apoptosis of oligodendrocyte progenitor cells and reduces myelination of the white matter.

Figure 5:

a | Coronal sections of the periventricular white matter of the frontal lobe of the forebrain of a preterm (born at ED29; term 32 days) rabbit kit with intraventricular haemorrhage (IVH) on postnatal day 2 and a premature human infant (born at gestational age 27 weeks) on postnatal day 3 with IVH. Both sections were immunolabelled with O4 antibody (red) and TUNEL (terminal deoxynucleotidyl transferase dUTP nick-end labeling, which detects DNA breaks) staining (green). Apoptotic O4-positive and TUNEL-positive cells are shown (arrowheads). Scale bar 20 μm. b | Representative coronal section of the forebrain of an ED29 rabbit kit with IVH on postnatal day 14 immmunolabelled with an antibody targeting myelin basic protein versus a control kit without IVH, also on day 14. Reduced myelination is evident in the corona radiata of the kit with IVH. ED, embryonic day; V, ventricle. Scale bar 100 μm.

Oligodendrogenesis is regulated by several different families of transcription factors.65 Basic helix-loop-helix (bHLH) transcription factors can be inhibitory (ID2 and ID4) or excitatory (OLIG1, OLIG2, and MASH1). The most important homeodomain transcription factors are Nkx2.2 and Nkx6.1, and the high mobility group transcription factors include SOX9, SOX10, and SOX17. The presence of IVH downregulates the excitatory transcription factors OLIG1, OLIG2 and SOX10 and upregulates inhibitory transcription factors, including ID2 and ID4, in preterm rabbits,26 thereby inhibiting OPC differentiation and contributing to myelination failure.

Underlying mechanisms

The development of IVH stimulates a constellation of signaling cascades that trigger inflammation, generate ROS and cause glutamate excitotoxicity in the periventricular germinal matrix and adjacent white matter.63,66,67 Oxidative stress, excitotoxicity and inflammation are the interlinked consequences of glial cell reactions to the presence of haemorrhage.68 Activated microglial cells and invading macrophages release various pro-inflammatory cytokines, chemokines, ROS and reactive nitrogen species, which collectively induce inflammatory and oxidative injury.63,67 For example, IVH leads to a release of glutamate into the extrasynaptic space. Owing to the reduced capacity of glial transporters to clear this glutamate, OPCs experience glutamate excitotoxicity and calcium-mediated injury.69 Similar mechanisms of perinatal brain injury have been described in models of hypoxic-ischaemic brain injury and neuroinflammation in neonatal animals.70,71

Studies in preterm rabbit kits and mouse pups have shown that IVH results in apoptotic cell death, infiltration of neutrophils and microglia, increased protein expression of cyclooxygenase 2 (COX2) and prostaglandin E2, and increased levels of pro-inflammatory cytokines in the periventricular germinal matrix and white matter.30,72,38,41 Consistent with these findings, autopsy samples from human preterm infants with IVH have demonstrated a similar escalation of apoptosis and immune cell infiltration in the periventricular white matter.30 Importantly, inflammation-suppressing treatment with COX2 or TNF inhibitors has restored myelination in rabbit kits with IVH, which suggests that IVH-induced inflammation inhibits myelination.

The presence of haemorrhage in the ventricle induces glutamate excitotoxicity by activation of AMPA receptors and an increased influx of calcium into OPCs, causing their degeneration and death.66 Moreover, treatment with AMPA receptor inhibitors reduced inflammation, apoptosis of OPCs, myelination failure and neurological dysfunction in preterm rabbits with IVH, which suggests that glutamate excitotoxicity is involved in OPC injury.66 Glutamate excitotoxicity causing OPC injury and restoration of myelination with AMPA receptor inhibition have also been demonstrated in neonatal models of hypoxia-ischaemia injury.73,74

Low antioxidant levels in premature infants render them more vulnerable than term infants to the deleterious effects of free radicals. The reduced levels of several antioxidant molecules, including superoxide dismutase, glutathione peroxidase, glutathione, melatonin, ceruloplasmin and vitamin E, are attributed to low endogenous production as well as interrupted maternal transfer of these products owing to premature delivery.70,75,76,77 Indeed, excessive production of superoxide, hydrogen peroxide and other free radicals damages white matter by inducing apoptotic cell death and inflammation in an animal model of IVH.67 Free-radical generation could be mediated by a number of oxidative pathways involving NAD(P)H oxidase, cyclooxygenases, xanthine oxidase or nitric oxide synthase and mitochondria.67 NAPH is expressed primarily in microglia and neutrophils in response to injury, and contributes to inflammatory brain injury.78 In preterm animals with IVH, NADPH oxidase is the major source of free radicals and treatment with NAPDH inhibitors offers neuroprotection.67 These findings are consistent with observations made in adult models of hypoxic-ischaemic brain injury and neurodegeneration.78,79 In a postnatal day 9 (P9) mouse model of hypoxic-ischaemic injury, gene expression of Cybb (which encodes cytochrome b-245 heavy chain, also known as NADPH oxidase 2, is elevated at 24 h and 72 h after the insult, but genetic or pharmacological inhibition of NADPH oxidase 2 did not reduce brain injury in these animals.80 The lack of any neuroprotective effect of NADPH oxidase 2 inhibition in these full-term mice can be attributed to either the timing of the experiment or to specific changes in NAPDH and glutamatergic signaling in the maturing brain of the neonatal mouse. Since activation of NADPH oxidase initiates redox signaling, inflammation, and injury to periventricular oligodendrocytes in a rabbit model of IVH,67 we speculate that increased NADPH oxidase 2 activity contributes to OPC injury in premature infants with IVH.81 Together, inflammation, oxidative stress, and glutamate toxicity contribute to both WMI and neurological deficits in survivors of IVH.

Dysregulated OPC maturation

Key signaling pathways.

Bone morphogenetic protein (BMP), Notch, Wnt and sonic hedgehog signaling pathways are the key determinants of oligodendrogenesis during the perinatal period (Fig. 6).26,82,83 In addition, epidermal growth factor (EGF), insulin-like growth factor 1 (IGF-I, also known as somatomedin-C) and thyroid hormone signaling in the OPC, as well as hyaluronan levels in the extracellular matrix, have important roles in OPC generation and maturation.8486

Figure 6: Regulation of oligodendrogenesis.

Figure 6:

Key signalling pathways that regulate the production and maturation of oligodendrocyte progenitor cells (OPCs) are likely to be affected by intraventricular haemorrhage (IVH)-induced injury to OPCs. (1) Bone morphogenetic proteins (BMPs) activate BMP receptors 1 and 2 and phosphorylate downstream SMAD proteins (SMAD1, SMAD5, SMAD8) to induce transcription of the DNA-binding protein inhibitors ID2 and ID4, which suppresses both the specification and maturation of OPCs. (2) Sonic hedgehog protein (SHH) binds to protein patched homolog 1 (PTC1) resulting in activation of smoothened homolog (SMO). Stimulation of SMO increases the levels of zinc finger proteins GLI1 and GLI2. This upregulation of GLI1 and GLI2 and degradation of transcriptional activator GLI3 contribute to the upregulation of oligodendrocyte transcription factor 2 (OLIGO2), which promotes oligodendrogenesis. (3) Notch activation releases the notch protein intracellular domain (NICD), which then translocates to the nucleus and induces the transcription of Notch-targeted genes. Notch activation inhibits the differentiation of OPCs. (4) Wnt activation results in dissociation of the β-catenin destruction complex. The resultant rise in cytoplasmic levels of β-catenin, and its translocation to the nucleus, induces transcription of Wnt target genes. IVH downregulates this Wnt signaling. APC, adenomatous polyposis coli protein; CK1, casein kinase 1 isoform-α; CSL, a family of transcription factors; GSK3B, glycogen synthase kinase-3β; LRP, lipoprotein receptor-related protein; PKA, protein kinase A; STK36, serine/threonine-protein kinase 36, also known as fused homolog; SUFUH, supressor of fused homolog; TCF7L2, transcription factor 7-like 2.

BMPs act as negative regulators of OPC differentiation87 and inhibit oligodendrocyte lineage commitment by diverting oligodendroglial precursors towards an astrocytic lineage.88 BMP4 suppresses both specification and maturation of OPCs by inducing ID2 and ID4 transcription factors.89 In both preterm rabbit kits with IVH and premature human infants with IVH, levels of BMP2 and BMP4 and levels of their downstream mediators phosphorylated SMAD1, SMAD5 and SMAD8 are elevated. Furthermore, BMP inhibition by human recombinant noggin can restore OPC differentiation and myelination in preterm rabbits with IVH.26 Consistent with these studies, noggin overexpression in a transgenic mouse model of hypoxic-ischaemic brain injury reverses the maturational arrest of OPCs.90 BMP and transforming growth factor-β (TGFβ) belong to the same superfamily. However, TGFβ signaling drives OPC differentiation and promotes myelination by modulating the expression of MYC and p21 genes.91 TGFβ1 and TGFβ2 levels are elevated in a rat model of IVH,92 but TGFβ inhibition by decorin and colchicine does not affect motor impairment or hydrocephalus in these animals with IVH.92,93 Both BMP and TGFβ levels are raised in animal models of IVH, and BMP elevation inhibits myelination in preterm animals with IVH. Wnt signaling has an important role in oligodendrocyte production and maturation and affects oligodendrogenesis in a context-dependent manner. Treatment with Wnt antagonists94 and inactivation of Wnt signaling in mice increases OPC production,95,96 although later studies showed that Wnt signaling promoted oligodendrogenesis in both in vivo and in vitro experiments.97,98 This suggests that Wnt signaling has distinct roles in OPC proliferation, differentiation, and myelination in a context dependent manner. In preterm rabbit kits with IVH, activation of β-catenin and expression of TCF4 and AXIN2 transcription factors are all reduced versus their counterparts in controls without IVH, suggesting that downregulation of Wnt signaling occurs in the context of IVH.82 Importantly, activation of Wnt signaling by glycogen synthase kinase-3β (GSK3β) inhibition, using the small molecule AR-A014418, accelerates maturation of OPCs, myelination and neurological recovery in preterm rabbits with IVH.82 In these experiments, GSK3β inhibition suppressed both inflammation and Notch signaling, the absence of which would contribute to myelination. During development, Notch signaling inhibits OPC differentiation and myelination both in vitro and in vivo.99,100 In agreement with these studies, treatment with a peptide inhibitor of GSK3β reduces pro-inflammatory microglial activation, WMI and neurobehavioural deficits in a postnatal day 1 (P1) mouse model of neuroinflammation.101 The sonic hedgehog signaling pathway also promotes the formation of ventrally derived OPCs in the forebrain (and thus fosters myelination) by inducing production of the OLIG2 transcription factor.102,103

Hormones and growth factors.

During the development of OPCs, thyroid hormone signaling promotes oligodendrocyte specification and maturation.104,105 Indeed, hypothyroidism causes myelination failure, whereas thyroxine treatment promotes myelination in adult animal models of demyelination.106,107 Intracellular availability of triiodothyronine is regulated by activating type 2 deiodinase (D2), and inactivating type 3 deiodinase (D3).108,109 D2 converts (inactive) thyroxine to (active) triiodothyronine, whereas D3 inactivates both triiodothyronine and thyroxine. In brain samples from humans and rabbits with IVH, D2 levels are reduced and D3 levels are increased versus their levels in controls without IVH.85 This situation is likely to result in reduced thyroid hormone signaling owing to diminished levels of T3 in neural cells. Accordingly, thyroxine treatment accelerates the proliferation and maturation of oligodendrocytes, increases transcription of OLIG2 and SOX10 genes, and consequently augments myelination in rabbit kits with IVH.85 Therefore, thyroxine treatment restores the maturation of OPCs as well as myelination.

EGF and its receptor EGFR regulate the survival, proliferation, and migration of neural precursor cells and stimulate their differentiation towards an oligodendrocyte lineage.110 Studies by our group show that expression of EGF and EGFR is more abundant in the ganglionic eminence relative to the cortical plate and white matter of human premature infants and that the development of IVH reduces EGF levels, but not EGFR expression, in both rabbits and humans.64 Importantly, treatment with recombinant human (rh)EGF promotes proliferation and maturation of OPCs, myelination of white matter, and neurological recovery in rabbits with IVH.64 Hence, EGF has an important role in oligodendrogenesis and enhances myelination in newborn infants with IVH.

IGF-I is a neurotropic hormone that plays a crucial part in brain development and maturation. Binding of IGF-I to its receptor IGF1R recruits canonical signaling pathways, including PI3K–Akt and Ras–Raf–MAP kinase signaling pathways, which promote neuroplasticity.86 IGF-I affects both neurons and glia. This growth factor enhances the proliferation as well as maturation of OPCs and thus promotes myelination in the neonatal rat model of hypoxic-ischaemic injury and demyelination.33,111 To our knowledge, IGF-I has not been tested in an animal model of IVH. However, in a phase 2 randomized controlled trial, rhIGF-I complexed with its binding protein rhIGFBP3) was administered to extremely preterm infants to minimize the complications of prematurity.112 This treatment reduced the occurrence of severe bronchopulmonary dysplasia and was associated with a trend towards a decline in the incidence of grade III IVH and PVHI. Long-term follow-up data on these infants are not yet available. As treatment with rhIGF-I can rescue OPCs in the immature white matter and promotes myelination and neurobehavioural function following hypoxic-ischaemic injury,113,114 we speculate that treatment with rhIGF-I–rhIGFBP3 complex might reduce motor and cognitive deficits in premature infants, and might be particularly effective in infants with IVH.

Neurodevelopmental outcomes

The long-term developmental consequences of IVH in children aged 5–10 years include an increased incidence of cerebral palsy and cognitive impairment after either mild or severe IVH in infancy compared to children without IVH.115117 Premature birth or the occurrence of IVH increase the risk of psychiatric disorders in adolescence, including ADHD, poor social skills, anxiety disorder and depression. Among adolescent survivors (72.9%) from the Neonatal Brain Haemorrhage Study (NBHS) cohort of 1,105 infants, the incidence of major depressive disorder (13% versus 5.2%) and obsessive-compulsive neurosis (11.6% vesus 1.4%) was higher in those with preterm birth who developed IVH than in those preterm infants who did not develop IVH.17,18,19 The presence of ventricular dilatation and parenchymal lesions were associated with an increased risk of developing ADHD.18 However, isolated GMH-IVH did not increase the risk of autism spectrum disorder.118 The mechanisms leading to this increased risk of psychiatric disorders in IVH survivors are not well understood, but might be related to both WMI and cortical injury in infancy and possibly to an associated cerebellar injury.

Neuroimaging findings

Cranial ultrasonography is routinely performed during the first week of life to detect IVH and early, severe WMI in premature infants of ≤30 weeks of gestational age.119 Cranial ultrasonography is repeated or MRI is performed at near-term age (35–42 weeks of postnatal age), just before the infants are discharged home. Neuro-imaging findings at near-term age predict the risk of neurodevelopmental impairment.120,121 The advantage of cranial ultrasonography is that it is a cost-effective, bedside technique that can show the evolution of the brain lesion. Following the diagnosis of severe IVH, sequential cranial ultrasonography is performed to identify PHVD and allow timely intervention122

The merits of MRI (especially diffusion-weighted and susceptibility-weighted MRI) include detection of subtle WMI lesions that would be missed by cranial ultrasonography, such as punctate lesions, diffuse white matter abnormalities and small cerebellar haemorrhages.123,124 In addition, advanced MRI-based imaging techniques, including diffusion tensor imaging (DTI), functional MRI (fMRI) and magnetic resonance spectroscopy (MRS) can provide information on white matter development, brain growth, connectivity, myelination, metabolism and brain function.125 DTI has been used to assess maps of white matter tracts and myelination in preterm infants with IVH. DTI reveals extensive white matter involvement with considerably lower factional anisotropy values in infants with both mild and severe IVH compared to matched controls without IVH.23,126,127 In preterm infants with PVHI, asymmetry of the corticospinal tracts can be assessed using DTI within weeks after its onset (Fig. 2).128 In addition, DTI tractography has also demonstrated disruptions in cerebellar white matter in infants with low-grade IVH.126 Standard and advanced MRI techniques are important tools that can be employed for diagnosis of IVH, to guide the management of affected patients and in research studies to evaluate the efficacy of interventions.

Diagnosis and Developmental Care

Increased severity of haemorrhage and earlier gestational ages of premature infants are associated with higher rates of death and neurodevelopmental impairment. In a 2014 study of a large population of preterm babies (n = 1,762 survivors), infants with grade III IVH-PVHI have higher rates of cerebral palsy (30%) and developmental delay (17.5%) than infants with low-grade IVH or no IVH at 2 years of age13. In addition, infants with low-grade IVH have substantially worse neurological outcomes than those of infants without IVH in terms of both rates of cerebral palsy (10.4% versus 6.5%) and developmental delay (7.8 versus 3.4%).13 These data are consistent with those of other major studies and a meta-analysis evaluating the developmental outcome of infants with IVH.129132 However, a few other studies with smaller sample sizes have reported similar outcomes in infants with low-grade IVH and no IVH.133

Very preterm infants with IVH are at an increased risk of motor, cognitive, behavioural and speech impairments in early childhood. Prompt diagnosis of these neurobehavioural impairments is crucial. The initial diagnosis of neurodevelopmental impairment is made by clinical history, physical examination and neuroimaging. A combination of tests, including the Prechtl General Movement Assessment, Hammersmith Infant Neurologic Examination (HINE) and MRI, offers the best predictive validity for detecting cerebral palsy before 5 months of age.123,134 However, after 5 months of age, the best predictive tools to detect cerebral palsy are MRI, HINE and the developmental assessment of young children (C index).123,125 In infants with IVH, MRI and cranial ultrasonography findings of cystic WMI, porencephaly, as well as cortical and deep gray matter lesions (representing thalamic and basal ganglia injury) are especially strongly associated with cerebral palsy.124,134 After the diagnosis of cerebral palsy has been made, the infant should be promptly referred to Early Intervention Program for deficit-specific management and regular medical and neurological monitoring. In addition, these infants should be screened and appropriately treated for other deficits or co-morbidities, including impairment in speech, vision, and hearing, to optimize their outcomes.

Developmental care to improve cognitive and behavioral outcome initiates in the NICUs. This includes promoting skin-to-skin contact with parents (Kangaroo care), enhancing positive auditory stimulation (parents talking to their infants, music therapy), reducing noise and lighting in NICU, initiating oral stimulation (non-nutritive sucking) and supporting breast feeding. These developmental interventions in NICU have positive effects on brain structure as well as on the cognitive and behavioral outcomes.135 Following discharge form the NICU, Early Intervention Program in the USA and European Agency for Special Needs and Inclusive Education in the Europe provide developmental care to these infants. These organizations offer multidisciplinary service to children 0–5 years of age and aims to promote child health and wellbeing, accelerate emerging competencies, reduce developmental delays, remediate existing or emerging disabilities, minimize functional deterioration and foster adoptive parenting and family function.

Early intervention could be hospital-based, home-based, or community-based. The focus of developmental intervention should be family-centered, aimed at enhancing parent–infant relationships. Interactions between infants and their parents can benefit from enabling parents to appreciate their infant’s distinctive characteristics, sensitizing parents to the infant’s cues and readiness for interaction and allaying parental anxiety and guilt related to the preterm birth. A number of studies underscore the superiority of early intervention over usual care in this setting.136,137 Moreover, early intervention services have been effective not only improving the cognitive outcome, but also in family functioning.138

Post-haemorrhagic ventricular dilatation

As this Review is primarily focused on WMI, we only briefly describe important advances in the management of post-haemorrhagic hydrocephalus. Increasing evidence supports the importance of early intervention for PHVD. Rather than waiting to initiate treatment after the onset of overt signs and symptoms of hydrocephalus (including bulging fontanelles, splayed sutures, sun-setting ophthalmological sign, apnoeas and a rapid increase in head circumference), intervention should be commenced in response to a rapid increase in measurements of the size of the lateral ventricles on cranial ultrasonography. Several observational retrospective studies have reinforced the notion that infants with PHVD who are managed on the basis of changes in cranial ultrasonography parameters exhibit better outcomes than do those who received treatment after the onset of clinical symptoms.139 Intervention is started at specific cut-off values of cranial ultrasonography parameters such as the ventricular index, anterior horn width and thalamic-occipital distance; for example, one such cut-off is 4 mm above the 97th centile in a cross-sectional chart of the ventricular index.140 The ELVIS (Early Versus Late Ventricular Intervention Study) was conducted to compare the clinical outcomes of infants with PHVD assigned to early intervention (that is, at a ventricular index >97th centile) versus late intervention (at a ventricular index >97th centile + 4 mm). Treatment of these infants included CSF tapping by lumbar puncture followed by reservoir placement.141

Few randomized controlled trials of interventions for PHVD have been conducted. The ELVIS results showed that rates of a composite outcome (consisting of death and cerebral palsy with Bayley Scales of Infant and Toddler Development cognitive and/or motor scores <70) were significantly better in the early intervention than in the late intervention group at 2 years of age, after adjustment for gestational age and severity of IVH.124 Moreover, outcome was superior for those infants who did not develop PVHI and did not require CSF diversion surgery. Of note, patient enrollment for ELVIS took ten years to complete. Our group faced a number of difficulties in obtaining parental consent, as the randomized nature of the trial meant that the parents, who did not want to delay intervention, were reluctant to expose their child to a 50% chance of being assigned to the delayed intervention group. Also, infants with a ventricular index exceeding the cut-off for late intervention (97th centile + 4 mm) when the haemorrhage was first diagnosed could not be included in the study. The study procedure was also cumbersome because the enrolled infants required daily cranial ultrasonography to guide treatment decision-making.

The families of infants who develop PHVD experience a substantially increased burden compared to the families of their preterm peers.142 Associated neurodevelopmental morbidities and low socioeconomic status further adversely affect these families.143,144 Hence, a family-centred intervention that aims to improve family support systems is necessary. Early intervention delivered according to cranial ultrasonographic parameters in conjunction with family-centred support could offer improved neurocognitive outcome to the survivors of IVH compared to usual care.

Neuroprotective strategies

Myelination is essential for both efficient axonal conduction of electrical impulses and for preserving axonal integrity.145,146 Myelin sheath formation requires the organization of voltage-dependent channels at nodes and paranodes, which are required for salutatory conduction. The myelin sheath reduces the need for ATP-dependent Na/K exchange in maintaining the resting potential of the axolemma. Delayed or arrested myelination in preterm infants results in continuous conduction through widely distributed sodium channels in the axolemma, thereby increasing ATP consumption. Thus, myelination failure increases energy consumption and axonal damage. Strategies to enhance myelination can be grouped into three categories: removal of blood products; inhibition of pathological processes in the brain, including inflammation, glutamate toxicity, and oxidative stress; and promotion of the proliferation and maturation of OPCs (Fig. 7).

Removal of blood products

The DRIFT (Drainage, Irrigation and Fibrinolytic Therapy) randomized clinical trial was based on the premise that the removal of haematoma and products of blood clot lysis would reduce the incidence of PHVD and neurological impairment in infants with IVH.147 This UK-based trial included 77 preterm infants with IVH and ventricular dilatation who received fibrinolytic therapy (intracerebroventricular infusions of recombinant tissue plasminogen activator) to lyse the clot, followed by irrigation of the ventricles with artificial CSF to wash out the blood clots. The results showed that DRIFT reduces the incidence of severe cognitive disability (Bayley-II Mental Development Index (MDI) score <55) in survivors at 2 years.147 The difficulties in performing the DRIFT clinical trial must have been enormous, as this intervention was quite invasive for premature infants.

A 10 year follow-up study of 52 children (median age at enrollment 20 days, range 7–28 days) randomly assigned to DRIFT (n=28) or standard care (n=24) showed that the cognitive quotient score was better for DRIFT-treated children than for controls after adjustment for sex, birth weight and IVH grades.148 The main limitation of the study is the small sample size, as DRIFT was stopped before enrollment was complete as 1/3 of those treated with DRIFT developed a rebleed.. In subsequent studies, a sophisticated neuroendoscopic ventricular irrigation procedure in infants with IVH led to a statistically significant reduction in rates of permanent shunt and post-IVH hydrocephalus.149151 In a retrospective study, 56 patients (26 weeks median gestation) underwent neuroendoscopic lavage (NEL) at median postmenstrual age of 31 weeks and followed for 34 months. Of these, 31 patients required permanent ventriculo-peritoneal shunt, suggesting a reduction of shunt placement in 43% cases.151 In a meta-analysis of 700 infants with PHVD performed in 2020, DRIFT was compared to conservative treatment, lumbar puncture, diuretics, as well as fibrinolytic therapy and was found to be superior to other modalities of treatment.152 Together, there is a need of double blinded randomized controlled trial using DRIFT or NEL, in which neuroendoscopic ventricle irrigation is performed at a correct postnatal age and long term outcome of the infants are assessed. Furthermore, those infants who are undergoing DRIFT can be followed for PHVD using head ultrasound and should be managed according to the recommendations made by de Vries and her co-investigators of the ELVIS trial.124,153

A number of preclinical studies have been performed in animal models to investigate chemical removal of haemoglobin and iron, or inactivation of downstream mediators of the thrombin signaling pathway. Intracerebroventricular treatment with either haptoglobin (for scavenging haemoglobin) or α1 microglobulin (for eliminating haeme) reduces inflammation and cell injury around the cerebral ventricles of preterm rabbits with IVH.43,44 However, the effect of these invasive therapies on WMI and neurological outcomes was not assessed, which reduces the translational potential of these studies. Treatment with iron chelators in a neonatal rat model of IVH reduced brain oedema and hydrocephalus.48,49 Additionally, iron-chelation therapy can alleviate free-radical-induced injury to oligodendrocytes in cell culture experiments.47 To counteract thrombin-mediated toxicity to OPCs, dabigatran (a thrombin inhibitor) or SCH79797 (a PAR1 receptor antagonist) have been tested in a neonatal rodent model of IVH.154 Both treatments resulted in reduced ventricular dilatation, increased myelination and improved neurological outcomes.41,155,156 Dabigatran is a FDA approved anticoagulant, but has a high risk of inducing hemorrhage in the brain and other organs upon treatment154. Thus, the safety of using this anticoagulant in premature infants after IVH might limit its use for minimizing WMI in future clinical trials.

Inhibition of inflammatory signaling

Inhibition of inflammation, oxidative stress or AMPA-receptor-induced glutamate excitotoxicity reduces WMI in animal models of IVH.63,66,67 The COX2 inhibitor celecoxib reduces levels of pro-inflammatory cytokines (TNF and IL1β), free-radical generation and microglial infiltration as well as enhancing myelination and neurological recovery in both rat and rabbit models of IVH.63,157 Neuroprotective benefits of celecoxib in several other animal models of brain injury reinforce the observations made in models of IVH. Indeed, postnatal celecoxib is a promising treatment for preterm infants with IVH that might enhance their neurological outcomes. Adverse effects of celecoxib include myocardial infarction, stroke, intestinal perforations and gastrointestinal bleeding. However, the concerns regarding myocardial infarction and stroke are relevant only to the geriatric population. Minocycline is a microglia inhibitor that has been tested in a rat model of germinal matrix haemorrhage. Minocycline treatment resulted in reductions in ferritin levels, brain oedema, brain cell death and hydrocephalus in rat pups with IVH relative to their levels in controls without IVH.48 Minocycline reduces microgliosis and levels of nitric oxide synthase, COX2 and prostaglandin, thereby exerting both anti-oxidative and anti-inflammatory effects.158 Moreover, minocycline treatment was effective in improving functional independence and was safe in a clinical trial of adult patients with ischaemic and haemorrhagic stroke159. However, this clinical trial was not adequately powered to assess its safety and efficacy Although minocycline has adverse effects including pigmentation of skin, teeth and bone, this agent has potential to improve the outcomes of premature infants.

AMPA (or kainate) receptor inhibition has shown neuroprotective potential in a rabbit model of IVH and other models of brain injury.66 Two competitive AMPA receptor antagonists, CNQX (6-cyano-7-nitroquinoxaline-2, 3-dione) and NBQX (2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline), have been found to be protective in several models of brain injury. The major limitation of these agents is their renal toxicity and, therefore, they are not in clinical use. However, perampanel, an FDA-approved antiepileptic drug, had similar beneficial effects to NBQX treatment (namely, enhanced myelination and neurological recovery) in a rabbit model of IVH.66 Perampanel is a potent and selective AMPA receptor antagonist and a broad-spectrum anticonvulsant.160 This agent has a favourable adverse-effect profile and lacks psychomimetic properties as it does not interact with NMDA receptors.161 Importantly, perampanel has shown efficacy and safety in phase III clinical trials as a treatment for partial seizures at doses of 8 mg and 12 mg.162,163 Hence, perampanel is another candidate neuroprotective agent that could be investigated in clinical trials aiming to improve the outcomes of IVH survivors.

Together, downregulation of these three interlinked cascades—inflammation, oxidative stress and glutamate excitotoxicity—offers a unique opportunity to reduce WMI in infants with IVH.

Supporting OPCs and myelination

In animal models, treatment with thyroxine, epidermal growth factor and stem cell treatment promote myelination and neurological recovery. Opportunities exist for translating these discoveries into clinical trials.

Thyroid hormone treatment.

Intramuscular thyroxine treatment increases the proliferation and maturation of OPCs, enhances myelination and promotes neurological recovery in premature rabbits with IVH.85 In addition, thyroxine treatment is associated with improved myelination and neurological function in several neonatal and adult models of brain injury. Thyroxine treatment is approved by the FDA for this indication and has been shown to be safe in premature infants in multiple clinical trials.164166 A randomized clinical trial of thyroxine treatment in premature infants (delivered at <27 gestational weeks) showed MDI values were 18 points higher at 24 months of age in thyroxine-treated infants than in those who had received placebo.165 The potential role of thyroxine treatment in minimizing WMI as well as enhancing the neurological outcomes of premature infants with IVH warrants investigation in future clinical trials.

Epidermal growth factor.

EGF treatment can restore myelination and promote clinical recovery in a rabbit model of IVH.167,168 Intranasal EGF has also been successfully used to enhance myelination in a rodent model of hypoxic–ischaemic demyelination 168 However, EGF treatment can also induce apoptosis, oxidative cell death, reduced cell maturation and malignant transformation in various cell lines.169 As EGF treatment has never been tried in humans, evidence of the safety of EGF treatment in infants with IVH is lacking.

Cell-based therapies.

Stem cell therapy has shown promise in several neonatal and adult models of intracranial haemorrhage. Mesenchymal stem cells (MSCs) are the most commonly used stem cells for clinical trials because of their excellent safety profile, ease of isolation and propagation and pleiotropic properties in relieving organ injury.148 Transplanted MSCs also enhance recovery owing to paracrine effects, including secretion of growth factors, such as brain-derived growth factor and stromal cell-derived factor-1, which induce neural cell proliferation and maturation.148 MSCs also reduce inflammation and generation of ROS and reactive nitrogen species by affecting microglia, macrophages and lymphocytes. 170 Moreover, MSCs promote angiogenesis and transfer mitochondria to damaged cells.170,171

In a rat model of IVH, transplantation of umbilical cord blood-derived MSCs into the cerebral ventricles reduced apoptotic cell death, periventricular inflammation and post-haemorrhagic hydrocephalus. Treated animals also showed enhanced myelination and neurologic recovery.172,173 MSC therapy has also shown success in neonatal animal models of hypoxia-ischaemia injury.174 A phase I clinical trial of MSC transplantation in nine premature infants (born at 26.1 ± 0.7 weeks gestation) with grade 4 IVH (PVHI) revealed that this treatment was well-tolerated and not associated with any serious adverse effects.175 With the success of the phase I trial, a phase II trial has been initiated to determine the therapeutic efficacy of MSC treatment in survivors of IVH (NCT02890953).175 The main barriers to the success of stem cell therapy in this setting are selection of the right stem cell type, identification of the appropriate dose and postnatal age of the infant, and identification of a suitable mode of cell delivery.

Intranasal delivery can provide a practical and non-invasive method of intracerebral treatment. Therapeutic agents are transported from the nasal cavity to the brain by olfactory and trigeminal nerve routes, bypassing the blood–brain barrier. Indeed, nasal application of neurotrophins and MSCs has offered neuroprotection in experimental models of brain injury.58,168 In one study, preterm infants with moderate-to-severe IVH received intranasal breast milk (which contains neurotrophins and stem cells) on postnatal days 5–28176. The incidence of PHVD, shunt surgery and severe porencephalic cyst all showed trends toward a reduction in the 16 infants receiving intranasal breast milk compared to 15 controls.176 The benefit of intranasal breast milk observed in this small retrospective analysis needs to be confirmed in a randomized controlled trial.

Stem cell therapy might become a promising treatment for preterm infants with moderate or severe IVH. However, in view of the potential adverse effects of this therapy, including tumour formation, graft rejection and inflammation, the risk and benefits of stem cell therapy can be assessed only after completion of randomized clinical trials.

Conclusions and future directions

GMH and IVH continue to be major complications of prematurity that result in WMI and neurological impairments in surviving infants. The past two decades have witnessed a spate of preclinical and clinical work in the field of IVH that has improved our understanding of the mechanisms underlying IVH-induced WMI, which include adverse effects on OPCs and axons triggered by blood components, particularly iron and thrombin

Current therapies for this disorder are limited to rehabilitation measures. As such, an unmet need remains to accelerate the development of new therapies that improve the outcomes of these infants. The most promising strategies investigated to date in infants with IVH include blood clot removal (DRIFT), early intervention (ELVIS) and stem cell transplantation.124,147 However, a number of barriers have to be overcome before the results of these studies can be translated into clinical practice. The mode of delivery of a therapeutic agent is important; intraventricular injection and surgical intervention are associated with inherent risks and adverse effects. Selection of the optimal window of treatment is also critical for success; in infants with IVH, treatment must be instituted before scarring and irreversible damage sets in. The DRIFT clinical trial was conducted in infants with IVH at a median age of 20 days, which resulted in some benefits, but not as many as anticipated.147 The median age at intervention in ELVIS was 9 days, which resulted in the lowest rate of ventriculoperitoneal shunts reported so far in infants with IVH and improved outcomes at 2 years of age.111 Nevertheless, caution is warranted because very early surgical intervention might worsen IVH and could result in exacerbation of injury.

Several other mechanism-targeted strategies have been investigated in preclinical models. Among the animal studies, oral or intramuscular thyroxine or celecoxib seem the most promising treatments as these agents are already FDA-approved in adults, convenient to deliver, and have a favourable adverse-effect profile. By contrast, the use of iron chelators or anti-thrombin treatment might be poorly tolerated and associated with major adverse effects.

Owing to considerable heterogeneity in the population of patients with IVH, an individualized approach to the selection of treatment is likely to be required. For example, blood clot removal is suitable for infants with grade III IVH, but not for those with isolated PVHI. Likewise, regenerative treatments and agents that counteract inflammation and oxidative stress in the brain (including celecoxib, antioxidants, thyroxine and stem cells) are likely to be most appropriate for infants with moderate-to-severe IVH, whereas the risk of adverse effects associated with these treatments might limit their benefits in infants with grade I IVH. Moreover, tiny premature infants frequently develop other complications, including severe respiratory distress syndrome, low blood pressure, infection, patent ductus arteriosus and low platelet levels that might hinder the timely institution of treatment. Despite these challenges, the establishment of viable treatment options for infants with IVH does not seem too far in the future.

Key points.

  • Intraventricular haemorrhage (IVH) results in periventricular white matter injury (WMI) in premature infants of 23–32 weeks gestation; survivors can develop neurodevelopmental sequelae including cerebral palsy, cognitive deficits and hydrocephalus.

  • IVH triggers robust inflammation around the cerebral ventricles, damages axons and induces apoptosis and maturational arrest of oligodendrocyte progenitors, leading to reduced myelination of white matter.

  • A constellation of blood-induced reactions, including oxidative stress, glutamate excitotoxicity, inflammation, deranged signaling pathways and alteration of the extracellular matrix, contribute to WMI in infants with IVH.

  • Early diagnosis of neurobehavioural impairments, timely referral to deficit-specific early intervention and family-centred care is crucial to optimize the neurodevelopmental outcomes of these infants.

  • Neuroimaging studies are important for early diagnosis of neurodevelopmental impairments, predicting outcomes and evaluating the efficacy of therapeutic interventions in both individual patients and clinical trials.

  • New therapies being tested in preclinical models might reduce cerebral inflammation and promote myelination; ongoing clinical trials are investigating stem cell treatment and endoscopic removal of clots.

Acknowledgements

The authors sincerely thank Robert Hevner, University of California San Diego, CA, USA for the images of brain slices from a human premature infant with IVH shown in Figure 1a and 1b. The authors also sincerely thank George Kleinman, New York Medical College, Valhalla, NY, USA and Peter Nikkels, University Medical Center, Utrecht, Netherlands, for the images of brain slices from a human premature infant with IVH in Figure 1c and 1d, respectively. The authors are grateful to Joseph Volpe, Harvard University, Boston, MA, USA for a critical review of the manuscript. The authors’ research work is supported by NIH grants RO1 NS110760 and R21NS102897 (both to P.B.).

Footnotes

Competing interests

The authors declare no competing interests.

References

  • 1.Courtney SE et al. High-frequency oscillatory ventilation versus conventional mechanical ventilation for very-low-birth-weight infants. N Engl J Med 347, 643–652, doi: 10.1056/NEJMoa012750347/9/643 [pii] (2002). [DOI] [PubMed] [Google Scholar]
  • 2.Horbar JD et al. Trends in mortality and morbidity for very low birth weight infants, 1991–1999. Pediatrics 110, 143–151 (2002). [DOI] [PubMed] [Google Scholar]
  • 3.McGowan EC & Vohr BR Neurodevelopmental Follow-up of Preterm Infants: What Is New? Pediatric clinics of North America 66, 509–523, doi: 10.1016/j.pcl.2018.12.015 (2019). [DOI] [PubMed] [Google Scholar]
  • 4.Vohr BR Neurodevelopmental outcomes of extremely preterm infants. Clinics in perinatology 41, 241–255, doi: 10.1016/j.clp.2013.09.003 (2014). [DOI] [PubMed] [Google Scholar]
  • 5.Stoll BJ et al. Neonatal outcomes of extremely preterm infants from the NICHD Neonatal Research Network. Pediatrics 126, 443–456, doi: 10.1542/peds.2009-2959 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Volpe JJ Intraventricular hemorrhage in the premature infant--current concepts. Part II. Annals of neurology 25, 109–116, doi: 10.1002/ana.410250202 (1989). [DOI] [PubMed] [Google Scholar]
  • 7.Papile LA, Burstein J, Burstein R & Koffler H Incidence and evolution of subependymal and intraventricular hemorrhage: a study of infants with birth weights less than 1,500 gm. The Journal of pediatrics 92, 529–534, doi: 10.1016/s0022-3476(78)80282-0 (1978). [DOI] [PubMed] [Google Scholar]
  • 8.Perlman JM & Volpe JJ Cerebral blood flow velocity in relation to intraventricular hemorrhage in the premature newborn infant. The Journal of pediatrics 100, 956–959, doi: 10.1016/s0022-3476(82)80527-1 (1982). [DOI] [PubMed] [Google Scholar]
  • 9.Valdez Sandoval P, Hernandez Rosales P, Quinones Hernandez DG, Chavana Naranjo EA & Garcia Navarro V Intraventricular hemorrhage and posthemorrhagic hydrocephalus in preterm infants: diagnosis, classification, and treatment options. Child’s nervous system : ChNS : official journal of the International Society for Pediatric Neurosurgery 35, 917–927, doi: 10.1007/s00381-019-04127-x (2019). [DOI] [PubMed] [Google Scholar]
  • 10.Armstrong DL, Sauls CD & Goddard-Finegold J Neuropathologic findings in short-term survivors of intraventricular hemorrhage. Am J Dis Child 141, 617–621 (1987). [DOI] [PubMed] [Google Scholar]
  • 11.Rushton DI, Preston PR & Durbin GM Structure and evolution of echo dense lesions in the neonatal brain. A combined ultrasound and necropsy study. Arch Dis Child 60, 798–808 (1985). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Skullerud K & Westre B Frequency and prognostic significance of germinal matrix hemorrhage, periventricular leukomalacia, and pontosubicular necrosis in preterm neonates. Acta Neuropathol 70, 257–261 (1986). [DOI] [PubMed] [Google Scholar]
  • 13.Bolisetty S et al. Intraventricular hemorrhage and neurodevelopmental outcomes in extreme preterm infants. Pediatrics 133, 55–62, doi: 10.1542/peds.2013-0372 (2014). [DOI] [PubMed] [Google Scholar]
  • 14.Davis AS et al. Outcomes of extremely preterm infants following severe intracranial hemorrhage. J Perinatol 34, 203–208, doi: 10.1038/jp.2013.162 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Vohr BR et al. School-age outcomes of very low birth weight infants in the indomethacin intraventricular hemorrhage prevention trial. Pediatrics 111, e340–346 (2003). [DOI] [PubMed] [Google Scholar]
  • 16.Nosarti C et al. Impaired executive functioning in young adults born very preterm. J Int Neuropsychol Soc 13, 571–581, doi: 10.1017/S1355617707070725 (2007). [DOI] [PubMed] [Google Scholar]
  • 17.Indredavik MS et al. Low-birth-weight adolescents: psychiatric symptoms and cerebral MRI abnormalities. Pediatr Neurol 33, 259–266, doi:S0887–8994(05)00253–5 [pii] 10.1016/j.pediatrneurol.2005.05.002 (2005). [DOI] [PubMed] [Google Scholar]
  • 18.Whitaker AH et al. Neonatal head ultrasound abnormalities in preterm infants and adolescent psychiatric disorders. Arch Gen Psychiatry 68, 742–752, doi: 10.1001/archgenpsychiatry.2011.62 (2011). [DOI] [PubMed] [Google Scholar]
  • 19.Taylor HG, Minich N, Bangert B, Filipek PA & Hack M Long-term neuropsychological outcomes of very low birth weight: associations with early risks for periventricular brain insults. Journal of the International Neuropsychological Society : JINS 10, 987–1004, doi: 10.1017/s1355617704107078 (2004). [DOI] [PubMed] [Google Scholar]
  • 20.Volpe JJ Edward B Neuhauser lecture. Current concepts of brain injury in the premature infant. AJR Am J Roentgenol 153, 243–251, doi: 10.2214/ajr.153.2.243 (1989). [DOI] [PubMed] [Google Scholar]
  • 21.Larroche JC Post-haemorrhagic hydrocephalus in infancy. Anatomical study. Biol Neonate 20, 287–299, doi: 10.1159/000240472 (1972). [DOI] [PubMed] [Google Scholar]
  • 22.Buser JR et al. Arrested preoligodendrocyte maturation contributes to myelination failure in premature infants. Annals of neurology 71, 93–109, doi: 10.1002/ana.22627 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bassan H et al. Ultrasonographic features and severity scoring of periventricular hemorrhagic infarction in relation to risk factors and outcome. Pediatrics 117, 2111–2118, doi: 10.1542/peds.2005-1570 (2006). [DOI] [PubMed] [Google Scholar]
  • 24.Takashima S, Mito T & Ando Y Pathogenesis of periventricular white matter hemorrhages in preterm infants. Brain Dev 8, 25–30 (1986). [DOI] [PubMed] [Google Scholar]
  • 25.Volpe JJ Intraventricular hemorrhage in the premature infant--current concepts. Part I. Annals of neurology 25, 3–11, doi: 10.1002/ana.410250103 (1989). [DOI] [PubMed] [Google Scholar]
  • 26.Dummula K et al. Bone morphogenetic protein inhibition promotes neurological recovery after intraventricular hemorrhage. J Neurosci 31, 12068–12082, doi: 10.1523/JNEUROSCI.0013-11.2011 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ou X et al. Impaired white matter development in extremely low-birth-weight infants with previous brain hemorrhage. AJNR Am J Neuroradiol 35, 1983–1989, doi: 10.3174/ajnr.A3988 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.van de Bor M, Guit GL, Schreuder AM, Wondergem J & Vielvoye GJ Early detection of delayed myelination in preterm infants. Pediatrics 84, 407–411 (1989). [PubMed] [Google Scholar]
  • 29.Haynes RL, Billiards SS, Borenstein NS, Volpe JJ & Kinney HC Diffuse axonal injury in periventricular leukomalacia as determined by apoptotic marker fractin. Pediatr Res 63, 656–661, doi: 10.1203/PDR.0b013e31816c825c (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Georgiadis P et al. Characterization of acute brain injuries and neurobehavioral profiles in a rabbit model of germinal matrix hemorrhage. Stroke 39, 3378–3388, doi: 10.1161/STROKEAHA.107.510883 (2008). [DOI] [PubMed] [Google Scholar]
  • 31.Chua CO et al. Consequences of intraventricular hemorrhage in a rabbit pup model. Stroke 40, 3369–3377, doi: 10.1161/STROKEAHA.109.549212 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Goulding DS et al. Acute brain inflammation, white matter oxidative stress, and myelin deficiency in a model of neonatal intraventricular hemorrhage. Journal of neurosurgery. Pediatrics, 1–11, doi: 10.3171/2020.5.PEDS20124 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lin S et al. IGF-1 protects oligodendrocyte progenitor cells and improves neurological functions following cerebral hypoxia-ischemia in the neonatal rat. Brain research 1063, 15–26, doi: 10.1016/j.brainres.2005.09.042 (2005). [DOI] [PubMed] [Google Scholar]
  • 34.Keep RF et al. Brain endothelial cell junctions after cerebral hemorrhage: Changes, mechanisms and therapeutic targets. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 38, 1255–1275, doi: 10.1177/0271678X18774666 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Aronowski J & Zhao X Molecular pathophysiology of cerebral hemorrhage: secondary brain injury. Stroke 42, 1781–1786, doi: 10.1161/STROKEAHA.110.596718 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Xue M et al. Does thrombin play a role in the pathogenesis of brain damage after periventricular hemorrhage? Brain pathology 15, 241–249, doi: 10.1111/j.1750-3639.2005.tb00527.x (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Xi G, Keep RF & Hoff JT Mechanisms of brain injury after intracerebral haemorrhage. Lancet Neurol 5, 53–63, doi: 10.1016/S1474-4422(05)70283-0 (2006). [DOI] [PubMed] [Google Scholar]
  • 38.Coughlin SR Thrombin signalling and protease-activated receptors. Nature 407, 258–264, doi: 10.1038/35025229 (2000). [DOI] [PubMed] [Google Scholar]
  • 39.Gao F et al. Hydrocephalus after intraventricular hemorrhage: the role of thrombin. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 34, 489–494, doi: 10.1038/jcbfm.2013.225 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lekic T et al. PAR-1, −4, and the mTOR Pathway Following Germinal Matrix Hemorrhage. Acta Neurochir Suppl 121, 213–216, doi: 10.1007/978-3-319-18497-5_38 (2016). [DOI] [PubMed] [Google Scholar]
  • 41.Yoon H, Radulovic M, Drucker KL, Wu J & Scarisbrick IA The thrombin receptor is a critical extracellular switch controlling myelination. Glia 63, 846–859, doi: 10.1002/glia.22788 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hao XD et al. Thrombin disrupts vascular endothelial-cadherin and leads to hydrocephalus via protease-activated receptors-1 pathway. CNS neuroscience & therapeutics 25, 1142–1150, doi: 10.1111/cns.13129 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ley D et al. High Presence of Extracellular Hemoglobin in the Periventricular White Matter Following Preterm Intraventricular Hemorrhage. Front Physiol 7, 330, doi: 10.3389/fphys.2016.00330 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Romantsik O et al. The heme and radical scavenger alpha1-microglobulin (A1M) confers early protection of the immature brain following preterm intraventricular hemorrhage. J Neuroinflammation 16, 122, doi: 10.1186/s12974-019-1486-4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Huang FP et al. Brain edema after experimental intracerebral hemorrhage: role of hemoglobin degradation products. Journal of neurosurgery 96, 287–293, doi: 10.3171/jns.2002.96.2.0287 (2002). [DOI] [PubMed] [Google Scholar]
  • 46.Wu H, Wu T, Xu X, Wang J & Wang J Iron toxicity in mice with collagenase-induced intracerebral hemorrhage. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 31, 1243–1250, doi: 10.1038/jcbfm.2010.209 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Masuda T et al. Oral administration of metal chelator ameliorates motor dysfunction after a small hemorrhage near the internal capsule in rat. J Neurosci Res 85, 213–222, doi: 10.1002/jnr.21089 (2007). [DOI] [PubMed] [Google Scholar]
  • 48.Guo J et al. Minocycline-induced attenuation of iron overload and brain injury after experimental germinal matrix hemorrhage. Brain Res 1594, 115–124, doi: 10.1016/j.brainres.2014.10.046 (2015). [DOI] [PubMed] [Google Scholar]
  • 49.Meng H et al. Deferoxamine alleviates chronic hydrocephalus after intraventricular hemorrhage through iron chelation and Wnt1/Wnt3a inhibition. Brain Res 1602, 44–52, doi: 10.1016/j.brainres.2014.08.039 (2015). [DOI] [PubMed] [Google Scholar]
  • 50.Wang M et al. Complement Inhibition Attenuates Early Erythrolysis in the Hematoma and Brain Injury in Aged Rats. Stroke; a journal of cerebral circulation 50, 1859–1868, doi: 10.1161/STROKEAHA.119.025170 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ahmad S, Bhatia K, Kindelin A & Ducruet AF The Role of Complement C3a Receptor in Stroke. Neuromolecular medicine 21, 467–473, doi: 10.1007/s12017-019-08545-7 (2019). [DOI] [PubMed] [Google Scholar]
  • 52.Ducruet AF et al. C3a receptor modulation of granulocyte infiltration after murine focal cerebral ischemia is reperfusion dependent. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 28, 1048–1058, doi: 10.1038/sj.jcbfm.9600608 (2008). [DOI] [PubMed] [Google Scholar]
  • 53.Jarlestedt K et al. Receptor for complement peptide C3a: a therapeutic target for neonatal hypoxic-ischemic brain injury. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 27, 3797–3804, doi: 10.1096/fj.13-230011 (2013). [DOI] [PubMed] [Google Scholar]
  • 54.Moran J et al. Intranasal C3a treatment ameliorates cognitive impairment in a mouse model of neonatal hypoxic-ischemic brain injury. Experimental neurology 290, 74–84, doi: 10.1016/j.expneurol.2017.01.001 (2017). [DOI] [PubMed] [Google Scholar]
  • 55.Stokowska A et al. Complement peptide C3a stimulates neural plasticity after experimental brain ischaemia. Brain : a journal of neurology 140, 353–369, doi: 10.1093/brain/aww314 (2017). [DOI] [PubMed] [Google Scholar]
  • 56.Yung YC, Stoddard NC, Mirendil H & Chun J Lysophosphatidic Acid signaling in the nervous system. Neuron 85, 669–682, doi: 10.1016/j.neuron.2015.01.009 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yung YC et al. Lysophosphatidic acid signaling may initiate fetal hydrocephalus. Science translational medicine 3, 99ra87, doi: 10.1126/scitranslmed.3002095 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Yu-Taeger L et al. Intranasal Administration of Mesenchymal Stem Cells Ameliorates the Abnormal Dopamine Transmission System and Inflammatory Reaction in the R6/2 Mouse Model of Huntington Disease. Cells 8, doi: 10.3390/cells8060595 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lopez-Serrano C et al. Lysophosphatidic acid receptor type 2 activation contributes to secondary damage after spinal cord injury in mice. Brain, behavior, and immunity 76, 258–267, doi: 10.1016/j.bbi.2018.12.007 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Back SA et al. Late oligodendrocyte progenitors coincide with the developmental window of vulnerability for human perinatal white matter injury. J Neurosci 21, 1302–1312 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Back SA, Riddle A & McClure MM Maturation-dependent vulnerability of perinatal white matter in premature birth. Stroke 38, 724–730, doi: 10.1161/01.STR.0000254729.27386.05 (2007). [DOI] [PubMed] [Google Scholar]
  • 62.Rakic S & Zecevic N Early oligodendrocyte progenitor cells in the human fetal telencephalon. Glia 41, 117–127, doi: 10.1002/glia.10140 (2003). [DOI] [PubMed] [Google Scholar]
  • 63.Vinukonda G et al. Neuroprotection in a rabbit model of intraventricular haemorrhage by cyclooxygenase-2, prostanoid receptor-1 or tumour necrosis factor-alpha inhibition. Brain 133, 2264–2280, doi: 10.1093/brain/awq107 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Vinukonda G et al. Epidermal growth factor preserves myelin and promotes astrogliosis after intraventricular hemorrhage. Glia 64, 1987–2004, doi: 10.1002/glia.23037 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Nicolay DJ, Doucette JR & Nazarali AJ Transcriptional control of oligodendrogenesis. Glia 55, 1287–1299, doi: 10.1002/glia.20540 (2007). [DOI] [PubMed] [Google Scholar]
  • 66.Dohare P et al. AMPA-Kainate Receptor Inhibition Promotes Neurologic Recovery in Premature Rabbits with Intraventricular Hemorrhage. J Neurosci 36, 3363–3377, doi: 10.1523/JNEUROSCI.4329-15.2016 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zia MT et al. Oxidative- Nitrosative Stress in a Rabbit Pup Model of Germinal Matrix hemorrhage: Role of NAD(P)H Oxidase. Stroke 40 2191–2198 (2009. ). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yang Q, Huang Q, Hu Z & Tang X Potential Neuroprotective Treatment of Stroke: Targeting Excitotoxicity, Oxidative Stress, and Inflammation. Frontiers in neuroscience 13, 1036, doi: 10.3389/fnins.2019.01036 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Tilleux S & Hermans E Neuroinflammation and regulation of glial glutamate uptake in neurological disorders. J Neurosci Res 85, 2059–2070, doi: 10.1002/jnr.21325 (2007). [DOI] [PubMed] [Google Scholar]
  • 70.Panfoli I et al. Oxidative Stress as a Primary Risk Factor for Brain Damage in Preterm Newborns. Frontiers in pediatrics 6, 369, doi: 10.3389/fped.2018.00369 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hagberg H et al. The role of inflammation in perinatal brain injury. Nature reviews. Neurology 11, 192–208, doi: 10.1038/nrneurol.2015.13 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Xue M, Balasubramaniam J, Buist RJ, Peeling J & Del Bigio MR Periventricular/intraventricular hemorrhage in neonatal mouse cerebrum. J Neuropathol Exp Neurol 62, 1154–1165 (2003). [DOI] [PubMed] [Google Scholar]
  • 73.Follett PL et al. Glutamate receptor-mediated oligodendrocyte toxicity in periventricular leukomalacia: a protective role for topiramate. The Journal of neuroscience : the official journal of the Society for Neuroscience 24, 4412–4420, doi: 10.1523/JNEUROSCI.0477-04.2004 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Follett PL, Rosenberg PA, Volpe JJ & Jensen FE NBQX attenuates excitotoxic injury in developing white matter. The Journal of neuroscience : the official journal of the Society for Neuroscience 20, 9235–9241 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Perrone S, Negro S, Tataranno ML & Buonocore G Oxidative stress and antioxidant strategies in newborns. J Matern Fetal Neonatal Med 23 Suppl 3, 63–65, doi: 10.3109/14767058.2010.509940 (2010). [DOI] [PubMed] [Google Scholar]
  • 76.Ozsurekci Y & Aykac K Oxidative Stress Related Diseases in Newborns. Oxid Med Cell Longev 2016, 2768365, doi: 10.1155/2016/2768365 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Perrone S et al. Early identification of the risk for free radical-related diseases in preterm newborns. Early Hum Dev 86, 241–244, doi: 10.1016/j.earlhumdev.2010.03.008 (2010). [DOI] [PubMed] [Google Scholar]
  • 78.Ma MW et al. NADPH oxidase in brain injury and neurodegenerative disorders. Molecular neurodegeneration 12, 7, doi: 10.1186/s13024-017-0150-7 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chen H, Song YS & Chan PH Inhibition of NADPH oxidase is neuroprotective after ischemia-reperfusion. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 29, 1262–1272, doi: 10.1038/jcbfm.2009.47 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Doverhag C et al. Pharmacological and genetic inhibition of NADPH oxidase does not reduce brain damage in different models of perinatal brain injury in newborn mice. Neurobiology of disease 31, 133–144, doi: 10.1016/j.nbd.2008.04.003 (2008). [DOI] [PubMed] [Google Scholar]
  • 81.Choi BY et al. Inhibition of NADPH oxidase activation reduces EAE-induced white matter damage in mice. J Neuroinflammation 12, 104, doi: 10.1186/s12974-015-0325-5 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Dohare P et al. Glycogen synthase kinase-3beta inhibition enhances myelination in preterm newborns with intraventricular hemorrhage, but not recombinant Wnt3A. Neurobiology of disease 118, 22–39, doi: 10.1016/j.nbd.2018.06.015 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bergles DE & Richardson WD Oligodendrocyte Development and Plasticity. Cold Spring Harb Perspect Biol 8, a020453, doi: 10.1101/cshperspect.a020453 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Vinukonda G et al. Hyaluronidase and Hyaluronan Oligosaccharides Promote Neurological Recovery after Intraventricular Hemorrhage. The Journal of neuroscience : the official journal of the Society for Neuroscience 36, 872–889, doi: 10.1523/JNEUROSCI.3297-15.2016 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Vose LR et al. Treatment with thyroxine restores myelination and clinical recovery after intraventricular hemorrhage. J Neurosci 33, 17232–17246, doi: 10.1523/JNEUROSCI.2713-13.2013 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Dyer AH, Vahdatpour C, Sanfeliu A & Tropea D The role of Insulin-Like Growth Factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience 325, 89–99, doi: 10.1016/j.neuroscience.2016.03.056 (2016). [DOI] [PubMed] [Google Scholar]
  • 87.Grinspan JB et al. Stage-specific effects of bone morphogenetic proteins on the oligodendrocyte lineage. J Neurobiol 43, 1–17 (2000). [PubMed] [Google Scholar]
  • 88.Gomes WA, Mehler MF & Kessler JA Transgenic overexpression of BMP4 increases astroglial and decreases oligodendroglial lineage commitment. Dev Biol 255, 164–177 (2003). [DOI] [PubMed] [Google Scholar]
  • 89.Samanta J & Kessler JA Interactions between ID and OLIG proteins mediate the inhibitory effects of BMP4 on oligodendroglial differentiation. Development 131, 4131–4142, doi: 10.1242/dev.01273 (2004). [DOI] [PubMed] [Google Scholar]
  • 90.Chang J, Dettman RW & Dizon MLV Bone morphogenetic protein signaling: a promising target for white matter protection in perinatal brain injury. Neural regeneration research 13, 1183–1184, doi: 10.4103/1673-5374.235025 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Palazuelos J, Klingener M & Aguirre A TGFbeta signaling regulates the timing of CNS myelination by modulating oligodendrocyte progenitor cell cycle exit through SMAD3/4/FoxO1/Sp1. The Journal of neuroscience : the official journal of the Society for Neuroscience 34, 7917–7930, doi: 10.1523/JNEUROSCI.0363-14.2014 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cherian S, Thoresen M, Silver IA, Whitelaw A & Love S Transforming growth factor-betas in a rat model of neonatal posthaemorrhagic hydrocephalus. Neuropathol Appl Neurobiol 30, 585–600, doi: 10.1111/j.1365-2990.2004.00588.x (2004). [DOI] [PubMed] [Google Scholar]
  • 93.Hoque N, Thoresen M, Aquilina K, Hogan S & Whitelaw A Decorin and colchicine as potential treatments for post-haemorrhagic ventricular dilatation in a neonatal rat model. Neonatology 100, 271–276, doi: 10.1159/000327842 (2011). [DOI] [PubMed] [Google Scholar]
  • 94.Shimizu T et al. Wnt signaling controls the timing of oligodendrocyte development in the spinal cord. Dev Biol 282, 397–410, doi: 10.1016/j.ydbio.2005.03.020 (2005). [DOI] [PubMed] [Google Scholar]
  • 95.Langseth AJ et al. Wnts influence the timing and efficiency of oligodendrocyte precursor cell generation in the telencephalon. The Journal of neuroscience : the official journal of the Society for Neuroscience 30, 13367–13372, doi: 10.1523/JNEUROSCI.1934-10.2010 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ye F et al. HDAC1 and HDAC2 regulate oligodendrocyte differentiation by disrupting the beta-catenin-TCF interaction. Nat Neurosci 12, 829–838, doi: 10.1038/nn.2333 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Azim K & Butt AM GSK3beta negatively regulates oligodendrocyte differentiation and myelination in vivo. Glia 59, 540–553, doi: 10.1002/glia.21122 (2011). [DOI] [PubMed] [Google Scholar]
  • 98.Ortega F et al. Oligodendrogliogenic and neurogenic adult subependymal zone neural stem cells constitute distinct lineages and exhibit differential responsiveness to Wnt signalling. Nat Cell Biol 15, 602–613, doi: 10.1038/ncb2736 (2013). [DOI] [PubMed] [Google Scholar]
  • 99.Genoud S et al. Notch1 control of oligodendrocyte differentiation in the spinal cord. J Cell Biol 158, 709–718, doi: 10.1083/jcb.200202002 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wang S et al. Notch receptor activation inhibits oligodendrocyte differentiation. Neuron 21, 63–75 (1998). [DOI] [PubMed] [Google Scholar]
  • 101.Van Steenwinckel J et al. Decreased microglial Wnt/beta-catenin signalling drives microglial pro-inflammatory activation in the developing brain. Brain : a journal of neurology 142, 3806–3833, doi: 10.1093/brain/awz319 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lu QR et al. Common developmental requirement for Olig function indicates a motor neuron/oligodendrocyte connection. Cell 109, 75–86, doi: 10.1016/s0092-8674(02)00678-5 (2002). [DOI] [PubMed] [Google Scholar]
  • 103.Kessaris N et al. Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage. Nat Neurosci 9, 173–179, doi: 10.1038/nn1620 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Carre JL et al. Thyroid hormone receptor isoforms are sequentially expressed in oligodendrocyte lineage cells during rat cerebral development. J Neurosci Res 54, 584–594, doi: [pii] (1998). [DOI] [PubMed] [Google Scholar]
  • 105.Rodriguez-Pena A Oligodendrocyte development and thyroid hormone. J Neurobiol 40, 497–512 (1999). [DOI] [PubMed] [Google Scholar]
  • 106.Fernandez M et al. Thyroid hormone administration enhances remyelination in chronic demyelinating inflammatory disease. Proc Natl Acad Sci U S A 101, 16363–16368, doi: 10.1073/pnas.0407262101 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Harsan LA et al. Recovery from chronic demyelination by thyroid hormone therapy: myelinogenesis induction and assessment by diffusion tensor magnetic resonance imaging. J Neurosci 28, 14189–14201, doi: 10.1523/JNEUROSCI.4453-08.2008 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Courtin F et al. Thyroid hormone deiodinases in the central and peripheral nervous system. Thyroid 15, 931–942, doi: 10.1089/thy.2005.15.931 (2005). [DOI] [PubMed] [Google Scholar]
  • 109.Gereben B et al. Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocr Rev 29, 898–938, doi:er.2008–0019 [pii] 10.1210/er.2008-0019 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Gonzalez-Perez O & Alvarez-Buylla A Oligodendrogenesis in the subventricular zone and the role of epidermal growth factor. Brain Res Rev 67, 147–156, doi: 10.1016/j.brainresrev.2011.01.001 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hlavica M et al. Intrathecal insulin-like growth factor 1 but not insulin enhances myelin repair in young and aged rats. Neuroscience letters 648, 41–46, doi: 10.1016/j.neulet.2017.03.047 (2017). [DOI] [PubMed] [Google Scholar]
  • 112.Ley D et al. rhIGF-1/rhIGFBP-3 in Preterm Infants: A Phase 2 Randomized Controlled Trial. The Journal of pediatrics 206, 56–65 e58, doi: 10.1016/j.jpeds.2018.10.033 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wood TL et al. Delayed IGF-1 administration rescues oligodendrocyte progenitors from glutamate-induced cell death and hypoxic-ischemic brain damage. Developmental neuroscience 29, 302–310, doi: 10.1159/000105471 (2007). [DOI] [PubMed] [Google Scholar]
  • 114.Lin S, Fan LW, Rhodes PG & Cai Z Intranasal administration of IGF-1 attenuates hypoxic-ischemic brain injury in neonatal rats. Experimental neurology 217, 361–370, doi: 10.1016/j.expneurol.2009.03.021 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kiechl-Kohlendorfer U, Ralser E, Pupp Peglow U, Pehboeck-Walser N & Fussenegger B Early risk predictors for impaired numerical skills in 5-year-old children born before 32 weeks of gestation. Acta Paediatr 102, 66–71, doi: 10.1111/apa.12036 (2013). [DOI] [PubMed] [Google Scholar]
  • 116.Klebermass-Schrehof K et al. Impact of low-grade intraventricular hemorrhage on long-term neurodevelopmental outcome in preterm infants. Childs Nerv Syst 28, 2085–2092, doi: 10.1007/s00381-012-1897-3 (2012). [DOI] [PubMed] [Google Scholar]
  • 117.Sherlock RL, Anderson PJ, Doyle LW & Victorian Infant Collaborative Study, G. Neurodevelopmental sequelae of intraventricular haemorrhage at 8 years of age in a regional cohort of ELBW/very preterm infants. Early Hum Dev 81, 909–916, doi: 10.1016/j.earlhumdev.2005.07.007 (2005). [DOI] [PubMed] [Google Scholar]
  • 118.Movsas TZ et al. Autism spectrum disorder is associated with ventricular enlargement in a low birth weight population. The Journal of pediatrics 163, 73–78, doi: 10.1016/j.jpeds.2012.12.084 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Ment LR et al. Practice parameter: neuroimaging of the neonate: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology 58, 1726–1738 (2002). [DOI] [PubMed] [Google Scholar]
  • 120.Spittle AJ et al. Neonatal white matter abnormality predicts childhood motor impairment in very preterm children. Dev Med Child Neurol 53, 1000–1006, doi: 10.1111/j.1469-8749.2011.04095.x (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Woodward LJ, Anderson PJ, Austin NC, Howard K & Inder TE Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N Engl J Med 355, 685–694, doi: 10.1056/NEJMoa053792 (2006). [DOI] [PubMed] [Google Scholar]
  • 122.Leijser LM et al. Posthemorrhagic ventricular dilatation in preterm infants: When best to intervene? Neurology 90, e698–e706, doi: 10.1212/WNL.0000000000004984 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Cayam-Rand D et al. Predicting developmental outcomes in preterm infants: A simple white matter injury imaging rule. Neurology 93, e1231–e1240, doi: 10.1212/WNL.0000000000008172 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Cizmeci MN et al. Randomized Controlled Early versus Late Ventricular Intervention Study (ELVIS) in Posthemorrhagic Ventricular Dilatation: Outcome at 2 Years. The Journal of pediatrics, doi: 10.1016/j.jpeds.2020.08.014 (2020). [DOI] [PubMed] [Google Scholar]
  • 125.Lean RE et al. Altered neonatal white and gray matter microstructure is associated with neurodevelopmental impairments in very preterm infants with high-grade brain injury. Pediatric research 86, 365–374, doi: 10.1038/s41390-019-0461-1 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Morita T et al. Low-grade intraventricular hemorrhage disrupts cerebellar white matter in preterm infants: evidence from diffusion tensor imaging. Neuroradiology 57, 507–514, doi: 10.1007/s00234-015-1487-7 (2015). [DOI] [PubMed] [Google Scholar]
  • 127.Tortora D et al. The effects of mild germinal matrix-intraventricular haemorrhage on the developmental white matter microstructure of preterm neonates: a DTI study. European radiology 28, 1157–1166, doi: 10.1007/s00330-017-5060-0 (2018). [DOI] [PubMed] [Google Scholar]
  • 128.Roze E et al. Neonatal DTI early after birth predicts motor outcome in preterm infants with periventricular hemorrhagic infarction. Pediatric research 78, 298–303, doi: 10.1038/pr.2015.94 (2015). [DOI] [PubMed] [Google Scholar]
  • 129.Ancel PY et al. Cerebral palsy among very preterm children in relation to gestational age and neonatal ultrasound abnormalities: the EPIPAGE cohort study. Pediatrics 117, 828–835, doi: 10.1542/peds.2005-0091 (2006). [DOI] [PubMed] [Google Scholar]
  • 130.Patra K, Wilson-Costello D, Taylor HG, Mercuri-Minich N & Hack M Grades I-II intraventricular hemorrhage in extremely low birth weight infants: effects on neurodevelopment. The Journal of pediatrics 149, 169–173, doi: 10.1016/j.jpeds.2006.04.002 (2006). [DOI] [PubMed] [Google Scholar]
  • 131.Vavasseur C, Slevin M, Donoghue V & Murphy JF Effect of low grade intraventricular hemorrhage on developmental outcome of preterm infants. The Journal of pediatrics 151, e6; author reply e6–7, doi: 10.1016/j.jpeds.2007.03.051 (2007). [DOI] [PubMed] [Google Scholar]
  • 132.Mukerji A, Shah V & Shah PS Periventricular/Intraventricular Hemorrhage and Neurodevelopmental Outcomes: A Meta-analysis. Pediatrics 136, 1132–1143, doi: 10.1542/peds.2015-0944 (2015). [DOI] [PubMed] [Google Scholar]
  • 133.Reubsaet P et al. The Impact of Low-Grade Germinal Matrix-Intraventricular Hemorrhage on Neurodevelopmental Outcome of Very Preterm Infants. Neonatology 112, 203–210, doi: 10.1159/000472246 (2017). [DOI] [PubMed] [Google Scholar]
  • 134.Morgan C et al. The Pooled Diagnostic Accuracy of Neuroimaging, General Movements, and Neurological Examination for Diagnosing Cerebral Palsy Early in High-Risk Infants: A Case Control Study. Journal of clinical medicine 8, doi: 10.3390/jcm8111879 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Soleimani F et al. Do NICU developmental care improve cognitive and motor outcomes for preterm infants? A systematic review and meta-analysis. BMC pediatrics 20, 67, doi: 10.1186/s12887-020-1953-1 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Spittle A, Orton J, Anderson PJ, Boyd R & Doyle LW Early developmental intervention programmes provided post hospital discharge to prevent motor and cognitive impairment in preterm infants. The Cochrane database of systematic reviews, CD005495, doi: 10.1002/14651858.CD005495.pub4 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Spittle A & Treyvaud K The role of early developmental intervention to influence neurobehavioral outcomes of children born preterm. Seminars in perinatology 40, 542–548, doi: 10.1053/j.semperi.2016.09.006 (2016). [DOI] [PubMed] [Google Scholar]
  • 138.Council on Children With D, Section on Developmental Behavioral P, Bright Futures Steering C & Medical Home Initiatives for Children With Special Needs Project Advisory C Identifying infants and young children with developmental disorders in the medical home: an algorithm for developmental surveillance and screening. Pediatrics 118, 405–420, doi: 10.1542/peds.2006-1231 (2006). [DOI] [PubMed] [Google Scholar]
  • 139.Bassan H et al. Timing of external ventricular drainage and neurodevelopmental outcome in preterm infants with posthemorrhagic hydrocephalus. European journal of paediatric neurology : EJPN : official journal of the European Paediatric Neurology Society 16, 662–670, doi: 10.1016/j.ejpn.2012.04.002 (2012). [DOI] [PubMed] [Google Scholar]
  • 140.Levene MI Measurement of the growth of the lateral ventricles in preterm infants with real-time ultrasound. Archives of disease in childhood 56, 900–904, doi: 10.1136/adc.56.12.900 (1981). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.de Vries LS et al. Treatment thresholds for intervention in posthaemorrhagic ventricular dilation: a randomised controlled trial. Arch Dis Child Fetal Neonatal Ed 104, 70–F75, doi: 10.1136/archdischild-2017-314206 (2019). [DOI] [PubMed] [Google Scholar]
  • 142.Agajany N et al. The impact of neonatal posthemorrhagic hydrocephalus of prematurity on family function at preschool age. Early human development 137, 104827, doi: 10.1016/j.earlhumdev.2019.104827 (2019). [DOI] [PubMed] [Google Scholar]
  • 143.Burnett AC, Cheong JLY & Doyle LW Biological and Social Influences on the Neurodevelopmental Outcomes of Preterm Infants. Clinics in perinatology 45, 485–500, doi: 10.1016/j.clp.2018.05.005 (2018). [DOI] [PubMed] [Google Scholar]
  • 144.Benavente-Fernandez I et al. Association of Socioeconomic Status and Brain Injury With Neurodevelopmental Outcomes of Very Preterm Children. JAMA network open 2, e192914, doi: 10.1001/jamanetworkopen.2019.2914 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Taveggia C, Feltri ML & Wrabetz L Signals to promote myelin formation and repair. Nature reviews. Neurology 6, 276–287, doi: 10.1038/nrneurol.2010.37 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Wilkins A, Chandran S & Compston A A role for oligodendrocyte-derived IGF-1 in trophic support of cortical neurons. Glia 36, 48–57 (2001). [DOI] [PubMed] [Google Scholar]
  • 147.Whitelaw A et al. Randomized trial of drainage, irrigation and fibrinolytic therapy for premature infants with posthemorrhagic ventricular dilatation: developmental outcome at 2 years. Pediatrics 125, e852–858, doi: 10.1542/peds.2009-1960 (2010). [DOI] [PubMed] [Google Scholar]
  • 148.Luyt K et al. Drainage, irrigation and fibrinolytic therapy (DRIFT) for posthaemorrhagic ventricular dilatation: 10-year follow-up of a randomised controlled trial. Archives of disease in childhood. Fetal and neonatal edition, doi: 10.1136/archdischild-2019-318231 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Etus V, Kahilogullari G, Karabagli H & Unlu A Early Endoscopic Ventricular Irrigation for the Treatment of Neonatal Posthemorrhagic Hydrocephalus: A Feasible Treatment Option or Not? A Multicenter Study. Turk Neurosurg 28, 137–141, doi: 10.5137/1019-5149.JTN.18677-16.0 (2018). [DOI] [PubMed] [Google Scholar]
  • 150.Schulz M, Buhrer C, Pohl-Schickinger A, Haberl H & Thomale UW Neuroendoscopic lavage for the treatment of intraventricular hemorrhage and hydrocephalus in neonates. Journal of neurosurgery. Pediatrics 13, 626–635, doi: 10.3171/2014.2.PEDS13397 (2014). [DOI] [PubMed] [Google Scholar]
  • 151.d’Arcangues C et al. Extended Experience with Neuroendoscopic Lavage for Posthemorrhagic Hydrocephalus in Neonates. World neurosurgery 116, e217–e224, doi: 10.1016/j.wneu.2018.04.169 (2018). [DOI] [PubMed] [Google Scholar]
  • 152.Mahoney L, Luyt K, Harding D & Odd D Treatment for Post-hemorrhagic Ventricular Dilatation: A Multiple-Treatment Meta-Analysis. Frontiers in pediatrics 8, 238, doi: 10.3389/fped.2020.00238 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.El-Dib M et al. Management of Post-hemorrhagic Ventricular Dilatation in the Infant Born Preterm. The Journal of pediatrics, doi: 10.1016/j.jpeds.2020.07.079 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Klebe D et al. Dabigatran ameliorates post-haemorrhagic hydrocephalus development after germinal matrix haemorrhage in neonatal rat pups. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 37, 3135–3149, doi: 10.1177/0271678X16684355 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Liu DZ et al. Blood-brain barrier breakdown and repair by Src after thrombin-induced injury. Annals of neurology 67, 526–533, doi: 10.1002/ana.21924 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Ramos-Mandujano G, Vazquez-Juarez E, Hernandez-Benitez R & Pasantes-Morales H Thrombin potently enhances swelling-sensitive glutamate efflux from cultured astrocytes. Glia 55, 917–925, doi: 10.1002/glia.20513 (2007). [DOI] [PubMed] [Google Scholar]
  • 157.Lekic T et al. Cyclooxygenase-2 Inhibition Provides Lasting Protection Following Germinal Matrix Hemorrhage in Premature Infant Rats. Acta Neurochir Suppl 121, 203–207, doi: 10.1007/978-3-319-18497-5_36 (2016). [DOI] [PubMed] [Google Scholar]
  • 158.Garrido-Mesa N, Zarzuelo A & Galvez J Minocycline: far beyond an antibiotic. British journal of pharmacology 169, 337–352, doi: 10.1111/bph.12139 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Malhotra K et al. Minocycline for acute stroke treatment: a systematic review and meta-analysis of randomized clinical trials. J Neurol 265, 1871–1879, doi: 10.1007/s00415-018-8935-3 (2018). [DOI] [PubMed] [Google Scholar]
  • 160.Hanada T et al. Perampanel: a novel, orally active, noncompetitive AMPA-receptor antagonist that reduces seizure activity in rodent models of epilepsy. Epilepsia 52, 1331–1340, doi: 10.1111/j.1528-1167.2011.03109.x (2011). [DOI] [PubMed] [Google Scholar]
  • 161.Rogawski MA Revisiting AMPA receptors as an antiepileptic drug target. Epilepsy Curr 11, 56–63, doi: 10.5698/1535-7511-11.2.56 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Gidal BE et al. Perampanel efficacy and tolerability with enzyme-inducing AEDs in patients with epilepsy. Neurology 84, 1972–1980, doi: 10.1212/WNL.0000000000001558 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Vazquez B, Yang H, Williams B, Zhou S & Laurenza A Perampanel efficacy and safety by gender: Subanalysis of phase III randomized clinical studies in subjects with partial seizures. Epilepsia 56, e90–94, doi: 10.1111/epi.13019 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.La Gamma EF et al. Phase 1 trial of 4 thyroid hormone regimens for transient hypothyroxinemia in neonates of <28 weeks’ gestation. Pediatrics 124, e258–268, doi: 10.1542/peds.2008-2837 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.van Wassenaer AG et al. Effects of thyroxine supplementation on neurologic development in infants born at less than 30 weeks’ gestation. N Engl J Med 336, 21–26, doi: 10.1056/NEJM199701023360104 (1997). [DOI] [PubMed] [Google Scholar]
  • 166.van Wassenaer AG, Westera J, Houtzager BA & Kok JH Ten-year follow-up of children born at <30 weeks’ gestational age supplemented with thyroxine in the neonatal period in a randomized, controlled trial. Pediatrics 116, e613–618, doi: 10.1542/peds.2005-0876 (2005). [DOI] [PubMed] [Google Scholar]
  • 167.Aguirre A, Rizvi TA, Ratner N & Gallo V Overexpression of the epidermal growth factor receptor confers migratory properties to nonmigratory postnatal neural progenitors. The Journal of neuroscience : the official journal of the Society for Neuroscience 25, 11092–11106, doi: 10.1523/JNEUROSCI.2981-05.2005 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Scafidi J et al. Intranasal epidermal growth factor treatment rescues neonatal brain injury. Nature 506, 230–234, doi: 10.1038/nature12880 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Hognason T et al. Epidermal growth factor receptor induced apoptosis: potentiation by inhibition of Ras signaling. FEBS Lett 491, 9–15 (2001). [DOI] [PubMed] [Google Scholar]
  • 170.van Velthoven CT, Gonzalez F, Vexler ZS & Ferriero DM Stem cells for neonatal stroke- the future is here. Front Cell Neurosci 8, 207, doi: 10.3389/fncel.2014.00207 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Chang YS, Ahn SY, Sung S & Park WS Stem Cell Therapy for Neonatal Disorders: Prospects and Challenges. Yonsei Med J 58, 266–271, doi: 10.3349/ymj.2017.58.2.266 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Ahn SY, Chang YS & Park WS Mesenchymal stem cells transplantation for neuroprotection in preterm infants with severe intraventricular hemorrhage. Korean J Pediatr 57, 251–256, doi: 10.3345/kjp.2014.57.6.251 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Ahn SY et al. Mesenchymal stem cells prevent hydrocephalus after severe intraventricular hemorrhage. Stroke 44, 497–504, doi: 10.1161/STROKEAHA.112.679092 (2013). [DOI] [PubMed] [Google Scholar]
  • 174.Kim ES et al. Human umbilical cord blood-derived mesenchymal stem cell transplantation attenuates severe brain injury by permanent middle cerebral artery occlusion in newborn rats. Pediatr Res 72, 277–284, doi: 10.1038/pr.2012.71 (2012). [DOI] [PubMed] [Google Scholar]
  • 175.Ahn SY, Chang YS, Sung SI & Park WS Mesenchymal Stem Cells for Severe Intraventricular Hemorrhage in Preterm Infants: Phase I Dose-Escalation Clinical Trial. Stem Cells Transl Med 7, 847–856, doi: 10.1002/sctm.17-0219 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Keller T et al. Intranasal breast milk for premature infants with severe intraventricular haemorrhage-an observation. European journal of pediatrics 178, 199–206, doi: 10.1007/s00431-018-3279-7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES