Am J Physiol Regul Integr Comp Physiol 306: R773–R786, 2014. First published March 19, 2014; doi:10.1152/ajpregu.00487.2013.

Review

Cerebral vascular regulation and brain injury in preterm infants Nadine Brew,1 David Walker,1,2 and Flora Y. Wong1,3,4 1

The Ritchie Centre, Monash Institute of Medical Research-Prince Henry’s Institute, Melbourne, Clayton, Victoria, Australia; and 2Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia; 3Monash Newborn, Monash Medical Centre, Melbourne, Victoria, Australia; and 4Department of Pediatrics, Monash University, Melbourne, Victoria, Australia Submitted 30 October 2013; accepted in final form 15 March 2014

Brew N, Walker D, Wong FY. Cerebral vascular regulation and brain injury in preterm infants. Am J Physiol Regul Integr Comp Physiol 306: R773–R786, 2014. First published March 19, 2014; doi:10.1152/ajpregu.00487.2013.—Cerebrovascular lesions, mainly germinal matrix hemorrhage and ischemic injury to the periventricular white matter, are major causes of adverse neurodevelopmental outcome in preterm infants. Cerebrovascular lesions and neuromorbidity increase with decreasing gestational age, with the white matter predominantly affected. Developmental immaturity in the cerebral circulation, including ongoing angiogenesis and vasoregulatory immaturity, plays a major role in the severity and pattern of preterm brain injury. Prevention of this injury requires insight into pathogenesis. Cerebral blood flow (CBF) is low in the preterm white matter, which also has blunted vasoreactivity compared with other brain regions. Vasoreactivity in the preterm brain to cerebral perfusion pressure, oxygen, carbon dioxide, and neuronal metabolism is also immature. This could be related to immaturity of both the vasculature and vasoactive signaling. Other pathologies arising from preterm birth and the neonatal intensive care environment itself may contribute to impaired vasoreactivity and ineffective CBF regulation, resulting in the marked variations in cerebral hemodynamics reported both within and between infants depending on their clinical condition. Many gaps exist in our understanding of how neonatal treatment procedures and medications have an impact on cerebral hemodynamics and preterm brain injury. Future research directions for neuroprotective strategies include establishing cotside, real-time clinical reference values for cerebral hemodynamics and vasoregulatory capacity and to demonstrate that these thresholds improve long-term outcomes for the preterm infant. In addition, stimulation of vascular development and repair with growth factor and cell-based therapies also hold promise. cerebral blood flow; cerebral vasoreactivity; infants; newborn; preterm brain injury

Understanding How Cerebral Blood Flow Is Regulated in the Preterm Brain With many infants now born preterm at 24 –32 wk of gestation (117), a full understanding of the physiological regulation of cerebral blood flow (CBF) in the immature brain is becoming increasingly important. Survival of infants born extremely preterm (⬍28 wk gestation) has increased sharply (50 –70%) over the last two decades (38, 42), but this has been accompanied by higher rates of neurodevelopmental disability, which exceed 50% in most studies (42, 127, 218), and at least a quarter of survivors develop substantial neurological morbidity, such as cerebral palsy (170). Most follow-up studies of extremely and very preterm infants (⬍32 wk gestation) show sequelae, such as cognitive deficits, academic underachievement, and the need for increased remedial assistance during mid-childhood and adolescence (169). Extremely and very preterm birth contributes disproportionately to the medical care Address for reprint requests and other correspondence: F. Wong, The Ritchie Centre, Level 5, Monash Medical Centre, 246 Clayton Rd., Clayton, Melbourne, Victoria, Australia, 3168 (e-mail: [email protected]). http://www.ajpregu.org

costs of prematurity, not only because of the intensive medical care required after birth, but also because survivors have disproportionately high rates of disabling conditions that generate high lifetime medical care costs (1, 167). The pathological correlates of neurodevelopmental disability with preterm birth include various cerebral lesions, most notably periventricular leukomalacia (PVL) and germinal matrix-intraventricular hemorrhage (GM-IVH) (132, 223). Such lesions are present from early in the neonatal period, as shown by cranial ultrasound or magnetic resonance imaging (MRI), and they contribute significantly to neonatal mortality, including deaths due to the decision to withdraw or limit life support for poor neurological prognosis (40). Although the etiology of cerebral lesions in the preterm infant is complex, it is generally accepted that hypoxia and poor cerebral perfusion lead to some regions of the preterm brain and, for reasons to be discussed, particularly white matter, falling into a condition best described as hypoxiaischemia (94). Therefore, as neuromorbidity increases in severity with decreasing gestational age (170), it is not unreasonable to think that immaturity in the regulation of the cerebrovascular circulation may be a prime factor in determin-

0363-6119/14 Copyright © 2014 the American Physiological Society

R773

Review R774

CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

ing the severity of preterm brain injury. However, effective brain-oriented neonatal intensive care is still impeded by two major obstacles: 1) the lack of clinically relevant cotside monitoring of cerebral perfusion and oxygenation; and 2) a lack of knowledge about whether maintaining the cerebral hemodynamic parameters within a “normal” range would translate into improved outcomes for preterm infants. Current understanding of how the circulation of the immature brain is regulated is based in large part on data from experimental fetal animal models (13, 143) and studies of preterm infants, in which, necessarily, measurements are made indirectly and noninvasively. Although both approaches have provided important insights, gaps remain in our knowledge of the normal and abnormal regulation of CBF in the preterm brain. The aim of this review is to integrate information on the regulation of cerebral hemodynamics and oxygenation in the preterm brain, and to discuss the relevance of such findings for the management of the preterm infant at risk of hypoxicischemic brain injury. This review will also discuss the challenges and directions for informed “brain-oriented” intensive care of preterm infants and possible new treatments to minimize cerebrovascular lesions. Techniques for Noninvasive Measurement of CBF in the Human Brain Clinically, investigation of CBF in infants is constrained by the necessity of using noninvasive techniques. Considerable insight into the cerebral circulation of preterm infants has been provided by application of the techniques summarized in Supplemental Table S1. Each of these techniques has its own merits and limitations regarding use in newborn infants. Regulation of Cerebral Hemodynamics Cerebral vascular development in the preterm brain. The developmental immaturity of the cerebral vasculature has long been thought to determine the regional vulnerability of the preterm brain to hypoxia-ischemia (94). Accordingly, cerebral vascularization and vasoregulation play a part in determining the differing patterns of cerebral injury that occur in preterm infants born between 24 and 36 wk of gestational age. ANATOMICAL DEVELOPMENT. Development of the cerebral vasculature is closely associated with structural development of the brain and involves a tightly regulated process of vasculogenesis and angiogenesis. In humans arterial development is completed earlier in the brain stem and cerebellum (between 20 and 24 wk of gestation), and then in the basal ganglia and diencephalon by 24 to 28 wk of gestation, and lastly in the cortex and germinal matrix. Initially, a single vascular plexus covers the cerebral surface. Thinner penetrating vessels descend from the cerebral surface, and by 24 wk of gestation, long penetrating vessels extend from the pial surface down into the periventricular regions, forming arterial end zones. Numerous growth factors are implicated in the vascular development in the brain. Among these, vascular endothelial growth factor (VEGF) is a key regulator of both vasculogenesis and angiogenesis, and expression of this hypoxia-responsive gene is critical for endothelial proliferation and migration, and in determining solute permeability in these nascent vascular conduits (55). For example, in mouse embryos, formation of pial collaterals, which provide an alternative route around a vascu-

lar obstruction and determine severity of ischemic conditions, requires VEGF stimulation of liver kinase 1-Notch signaling during a narrow developmental window (113). The formation of collateral arteries early in brain development is a determinant of their presence in the adult brain, such that disturbance of collaterogenesis results in a life-long deficiency (113), and potentially increased risk of stroke and cerebral ischemia in adult life. ANGIOGENESIS AND NEUROGENESIS. Angiogenesis and neurogenesis utilize similar molecular cues to guide migration and development of endothelial cells, neurons and axons (102). For example, the temporal and spatial expression of VEGF in the developing brain is closely correlated with vascularization and endothelial cell growth. Increased VEGF expression in many cell types (e.g., neuroblasts, neuroepithelial cells, radial glia, astrocytes, pericytes, and endothelial cells) coincides with increased neuronal metabolism, which stimulates angiogenesis (12, 174, 205). Disruption of neuronal activity by chronic excessive sensory stimulation and chemically induced seizures decreases angiogenesis in neonatal but not adult mice (213). Endothelial cells provide a microenvironment supporting the maintenance and proper differentiation of neural progenitor cells (194), as shown by in vitro co-culture systems. For example, when neural stem cells are co-cultured with endothelial cells, they develop greater self-renewal activity followed by extensive neurogenesis (179). GERMINAL MATRIX. Angiogenesis is especially active in the germinal matrix in the preterm brain. The germinal matrix is located in the thalamostriate groove and contains a richly vascularized collection of neural progenitor cells. Endothelial cell proliferation, VEGF and angiopoietin-2 expression are substantially increased in the preterm human germinal matrix relative to the white matter and cortex (16). This enhanced angiogenesis is associated with less astrocyte end-feet and pericyte capillary coverage in the germinal matrix vasculature than in the cortical gray or white matter in human fetuses and preterm infants (25, 47). Consequently this is thought to result in a more fragile vasculature with increased propensity for mechanical rupture and bleeding. Indeed, GM-IVH is relatively common in preterm infants, and predisposes them to long-term morbidities, including hydrocephalus, cerebral palsy, and impaired neurodevelopment (45). Cerebral blood flow in the preterm brain. LOW GLOBAL CBF. A dramatic change in CBF occurs at birth. In the newborn lamb CBF decreases to approximately one-third (relative to the term fetus) in the first 24 h after birth (164) and arterial oxygen content increases by more than 50% (61). In the preterm human infant, low CBF (Supplemental Table S2) and high cerebral oxygen extraction (COE) immediately after birth have been reported in several studies (64, 69, 153). Using nearinfrared spectroscopy (NIRS) in extremely preterm infants, CBF values between 5 and 17 ml·100g⫺1·min⫺1 were measured on the first day of life (121, 133, 219), comparable to measurements obtained using the Xenon clearance technique (between 5 and 19 ml·100g⫺1·min⫺1) (64) and positron emission tomography (PET) (between 5 and 17 ml·100g⫺1·min⫺1) (5). The paradox is that these very low CBF values are comparable to (or even less than) the minimal CBF estimated as necessary to maintain neuronal viability and metabolism in the adult brain (15 ml·100g⫺1·min⫺1) (17). Postnatally, CBF then rises over the first few days of life (121), typically to a

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

range of 10 –20 ml·100g⫺1·min⫺1 in growing preterm infants (206). Cerebral perfusion continues to increase throughout childhood before declining at some point in adulthood (65). REGIONAL CBF. Cerebral vascular density increases throughout fetal life and is closely correlated with the absolute value of CBF (Supplemental Table S2). Nevertheless, vascular density in white matter of the preterm infant remains low compared with other brain regions (15), consistent with findings in preterm infants using single photon emission computer tomography (SPECT) (197) and magnetic resonance perfusion images (126, 193), where CBF is higher in the thalami and brain stem (21) compared with white matter. Incomplete development of the deep vascular anatomy is likely to explain this difference between white and gray matter perfusion that is consistently reported in preterm infants and confirmed in studies of fetal sheep (165, 188). These findings lead to the suggestion that the margin of safety for decreased cerebral perfusion in the white matter of the preterm infant brain is lower than in the gray matter and other regions, contributing to the predilection of white matter to hypoxic-ischemic injury. CEREBRAL OXYGEN EXTRACTION. When oxygen delivery to the brain is low, as in cases of cerebral hypoperfusion or anemia, cerebral oxygen demand can then be met by increasing the extraction of oxygen from blood. The indication of increased oxygen extraction is shown by the increase in difference between cerebral arterial and venous oxygen contents, with the reduction in cerebral venous saturation being greater than any change in cerebral arterial saturation. Such increased cerebral oxygen extracton (COE) has been shown to occur in acute brain injury; for example, studies using PET in adult patients and animal models have demonstrated increased COE in areas of the brain surrounding an ischemic lesion, a condition known as “misery perfusion” (154). The neonatal brain also appears to adapt to conditions of low perfusion in a similar way. In the term newborn lamb brain, the marked decrease in CBF immediately after birth is associated with a corresponding increase in COE (83, 164). Similarly, newborn infants have elevated COE on the first day of life, and preterm infants have even higher COE than term infants (153, 181), which then decreases over the next 72 h (95). The ability of the immature brain to increase COE provides an important buffer against increased metabolic demand and hypoxic-ischemic conditions during the perinatal period. However, the high COE under basal conditions in the early postnatal period, especially in preterm infants, suggests that COE might be close to the maximum level possible for the brain and that any decrease of systemic oxygenation or CBF would not elicit any further increase in COE, so increasing the risk of hypoxic-ischemic injury. The maximum level of COE attainable in preterm infants, at which the oxygen extraction mechanism is exhausted and unable to meet demand for oxygen, remains unknown. In fetal lambs, maternal caffeine infusion increases fetal cerebral oxygen consumption, which is met by minimally increased CBF and a 20 –30% increase in COE (199). In fetal lambs and ventilated preterm lambs subjected to reduced cerebral oxygen delivery, COE values of up to 60% have been measured (7, 149), close to values found in adult patients with head injury (123). In support of very high COE representing significant hypoxic-ischemic risk, preterm infants with the highest COE values on day 1 of life are at increased risk of sustaining hemorrhagic parenchymal infarction (95).

R775

Cerebral Vasoreactivity in the Preterm Brain Cerebral vascular resistance is fundamentally regulated by the muscularis layer of the arterioles which, during brain development, is initially confined to the pial vessels and superficial penetrators, before descending into the cerebral parenchyma with further maturation (130). Consequently, cerebral vasoreactivity occurs predominantly in the superficial and peripheral regions of the preterm brain (Fig. 1), remote from the distal vascular beds, with the deeper vessels remaining relatively passive (44). This spatial restriction of vasoreactivity of the immature cerebral circulation is likely to contribute to the pattern of injury—most often, of the white matter— observed in the very preterm brain. It is worth noting that regardless of the stimulus, the cerebral vascular response is dependent on resting tone and diameter, so that vessels already dilated by hypercarbia or hypoxia have limited capacity for further vasodilation in response to additional stimulation, such as systemic hypotension, a relatively common occurrence in preterm infants. Although there are numerous physiological and pathologic stimuli capable of eliciting cerebrovascular responses, the most important ones are the following. Pressure autoregulation. Cerebral autoregulation refers to the maintenance of a relatively constant CBF over a broad range of perfusion pressures (104), the so-called “autoregulatory plateau”. The autoregulatory plateau in the adult human operates over mean blood pressures ranging from ⬃60 to 150 mmHg, and it takes 10 –90 s for CBF to stabilize after a change of systemic blood pressure (27). Autoregulation is not an all-or-none phenomenon, as the plateau usually has a small positive slope. In preterm infants, the plateau range and the position of lower and upper limits have been suggested to be much narrower and lower, but importantly, this varies significantly between and within individuals for reasons that are not yet fully understood (182, 221). MECHANISMS OF PRESSURE AUTOREGULATION. Several hypotheses have been proposed for the mechanisms that underlie autoregulation. One is that autoregulation is the direct result of a myogenic reflex (Fig. 1A), acting via a calcium-dependent pathway in smooth muscle in response to changes of transmural pressure, (and presumably wall tension), caused by changes in perfusion pressure (192). With decreased transmural pressure, endothelial-derived nitric oxide, carbon monoxide, and calcium-activated potassium channels induce vasodilatation (89). Shear-generated endothelial nitric oxide synthase is likely to be the dominant source of nitric oxide to mediate autoregulatory responses (19); for example, rat studies demonstrate that while inhibition of neuronal nitric oxide synthase impairs autoregulation, cerebral vasodilatation during reduced arterial pressure still occurs and vessel wall nitric oxide rises to near-normal levels. The vasoconstrictor effects of nitric oxide synthase inhibition are significantly less in cerebral vessels from young (4 –5 wk old) compared with adult rats, indicating that the contribution of endothelial nitric oxide in pressure autoregulation increases with age (115) and, therefore, may be less important in the preterm brain where other mechanisms may dominate. Apart from the pressure-sensitive myogenic mechanism, pressure- and flow-induced vasoreactivity have been proposed to function together in maintaining effective autoregulation. Increased constriction to flow in isolated hu-

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review R776

CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

A va sc ula riz at ion .

Cerebral Perfusion Pressure

Variable intracranial pressure (CSF drainage, soft skull, unfused fontanelle)

Autonomic center

Cerebral Venous Pressure

Systemic resistance (vasopressors, hypotension)

Cardiac Output (inotropes)

Heart

B Jugular Vein

NeuronsNO ACh VIP CGRP

AstrocytesPGs K+ EETs Adenosine

ECEDHF NO ET CO ROS Prostacyclin

NA DA NPY Serotonin

Intravascular pressure Ca2+ GABA

Fig. 1. A: Major factors contributing to cerebral blood flow (CBF) regulation in newborn infants. Cerebral perfusion pressure, hypercarbia, hypoxia, regional neuronal activation, and immature vascular anatomy (left side of dotted line) are major determinants of CBF in newborns. The vascular tone of cerebral resistance arterioles is modulated by numerous factors, primarily transmural pressure, perivascular pH, neuronal metabolism, as well as numerous vasodilatory molecules, such as potassium (K⫹), hydrogen ions (H⫹), carbon monoxide (CO), nitric oxide (NO), prostaglandins, prostanoids, and adenosine. Intracranial pressure and cerebral venous pressure (right side of dotted line) play a more significant role in CBF regulation in the adult brain. B: neurovascular unit in cerebral resistance arteriole, illustrating relationship between arteriole, interneuron, astrocyte, and neuron-secreted factors on smooth muscle cells, which regulate vascular diameter. A: CO, carbon monoxide; CSF, cerebrospinal fluid; WM, white matter. B: SMC, smooth muscle cell; EC, endothelial cell; Ach, acetylcholine; PGs, prostaglandins; EETs, epoxyeicosatrienoic acids; VIP, vasoactive intestinal polypeptide; CGRP, calcitonin gene related peptide; NA, noradrenaline; DA, dopamine; NPY, neuropeptide Y; EDHF, endothelium derived hyperpolarizing factor; ET, endothelin; GABA, ␥-aminobutyric acid; ROS, reactive oxygen species.

man internal carotid cerebral arteries is mediated by 20-hydroxyeicosatetraenoic acid (20-HETE, an arachadonic acid metabolite) acting via thromboxane A2/prostaglandin H2 receptors (200). Notably, many conflicting observations of flowinduced responses in cerebral vessels have been reported, varying from constriction to dilatation to biphasic responses; these differences in observed responses to flow may be dependent upon species, vessel type, and methods used (99), all of which needs to be resolved. Another proposed mechanism underlying cerebral autoregulation is the metabolic hypothesis, which is that a local reduction in blood flow results in the release of vasoactive factors from neurons or astrocytes, which then elicit dilatation of the adjacent cerebral vessels. The release of adenosine, a known cerebral vasodilator, is increased with reductions in blood pressure and is considered a prime mediator of metabolically regulated autoregulation (216). Other studies suggest

that cerebral vasodilatation, following reduced arterial blood pressure, is dependent on reduced oxygen tension within the tissues, which then either activates a local reflex (210), or causes the secondary release of vasodilator metabolites, such as adenosine (100) or nitric oxide (168). Finally, perivascular nerves may also contribute to the modulation of cerebral autoregulation. Extraparenchymal and intraparenchymal blood vessels are richly innervated by autonomic nerves (111) and have been shown to modify autoregulation. For example, the lower limit of CBF autoregulation is shifted toward higher blood pressure in parasympathetically denervated rats (128), and a similar shift is observed after stimulation of the cerebral sympathetic system (142). In newborn lambs, sympathetic denervation by cervical ganglionectomy results in greater CBF increases associated with blood pressure surges (111). Importantly though, cerebral autoregulation is still functional after cervical ganglionectomy (111,

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

191). It has been proposed that under normotensive conditions, myogenic control of cerebral vasomotor tone is the dominant mechanism determining the reactivity of cerebral autoregulation, with neurogenic and metabolic mechanisms having minimal influence. However, when systemic blood pressure is abnormally increased, sympathetic activity is increased commensurately to attenuate the increase in CBF (28, 30). PRESSURE AUTOREGULATION IN THE PRETERM BRAIN. Coincident changes in both CBF and arterial blood pressure in preterm infants, which appear to indicate little or no autoregulatory capacity (cerebral pressure-passivity), were initially reported more than 25 years ago; on the basis of which it was proposed that autoregulation was either impaired by preterm birth or was not yet fully developed in the immature brain (112). In either case, it might follow that the immature brain of the preterm infant is particularly susceptible during episodes of hypotension, with this perhaps leading to ischemia, as well as to hemorrhage during periods when blood pressure is either high or fluctuating, all of which are common events in the preterm infant undergoing intensive care. Studies in fetal lambs showed that autoregulation is present at 0.9 but not at 0.6 of gestation (74); these fetal ages in sheep correspond with brain maturation approximately equivalent to term and 26 –28 wk gestation in humans, respectively (162). Despite extensive investigation, it is still not clear when cerebral autoregulation really does develop in the human infant, or what the upper and lower limits of the autoregulatory range actually are. Some studies using NIRS, or electroencephalography detection of reduced cerebral activity (indicative of cerebral hypoperfusion), have found the lower blood pressure limit of the autoregulatory plateau to be between ⬃23 and 30 mmHg in very preterm infants (129, 203, 212). It is now apparent that the threshold and range of the autoregulatory plateau are quite variable both between and within infants, and that the general clinical condition of the infant, in addition to blood pressure and brain injury, are contributory factors (220, 221). A pressure-passive blood flow relationship is more likely to be present during episodes of hypotension than hypertension, suggesting that baseline MAP may be close to the lower threshold of the autoregulation plateau in very preterm infants (60). Furthermore, when hypotensive preterm infants are treated with inotropic agents, both CBF and MAP increase together (82, 129, 148, 176), indicating that pressure-passivity of the cerebral circulation persists over a range of arterial pressures in these infants. Other studies suggest that MAP alone is a poor indicator of CBF and autoregulatory capacity in the early postnatal period. While Soul et al. (187) demonstrated an association between systemic hypotension and cerebral pressure-passivity in preterm infants, in fact, the ability to predict the cerebral autoregulatory characteristics from blood pressure values was poor (182). This could be due to variation in the position of the autoregulatory plateau and its upper and lower thresholds within and between infants, and according to changes in their clinical condition (221). Not surprisingly, cross-sectional studies are particularly poor at identifying the characteristics of autoregulation in the human preterm cerebral circulation. When and how autoregulatory thresholds change with gestational or postnatal age in the preterm infant is not clear. In one study of preterm infants (⬍1,500 g birth weight), cerebral pressure-passivity (i.e., absence of autoregulation) was identi-

R777

fied consistently at different times over the first five postnatal days (182). Cerebral pressure-passivity was also more prevalent in younger preterm infants of lower gestational age (182). Differences in autoregulation related to gestational age were also reported in a study comparing hypotensive term and preterm infants treated with low-dose dopamine. Autoregulation was present in hypotensive term infants who received dopamine therapy because CBF velocity did not increase as MAP increased, whereas in hypotensive preterm infants, CBF velocity increased together with the rise in MAP, indicating the absence of autoregulation (176). There is clear evidence from fetal sheep of regional variations in autoregulation in the developing brain (189). For example, the brain stem has a lower autoregulatory threshold than the cerebrum, and as a result, a higher tolerance of hypotension. In contrast, CBF in white matter in these fetal sheep is particularly vulnerable to hypotension because autoregulatory adjustment of CBF is weak or nonexistent (189). These studies are consistent with imaging studies in hypotensive preterm infants that show perfusion of periventricular white matter may decrease when systemic blood pressure falls below 29 mmHg (22). Carbon dioxide vasoreactivity. MECHANISMS OF CARBON-DIOXIDE VASOREACTIVITY. Decreased CBF induced by hyperventilation and hypocapnia was first described 60 years ago in healthy adults (93). The effect of arterial carbon dioxide tension (PaCO2) on vascular smooth muscle is due to the changes in perivascular pH that occur after CO2 crosses the blood-brain barrier (Fig. 1A) and equilibrates with intraparenchymal bicarbonate (2). The pH-mediated changes in contractility of the small cerebral vessels result from activation of the electrogenic sodium pump, and ATP-sensitive, voltage-gated potassium channels leading to hyperpolarization of smooth muscle cells (155), inhibition of voltage-dependent calcium channels (2), and the activation and release of numerous secondary vasoactive substances, such as prostanoids, adenosine, and nitric oxide (105, 119, 152, 215). The larger upstream arteries and arterioles seem to respond to nitric oxide release, whereas both adenosine receptor activation and nitric oxide release mediate pial arteriolar responses to increases in CO2 levels (152). Sympathetic activity may also influence the reactivity of CBF to PaCO2 (31). Nevertheless, with chronic changes in PaCO2, CBF returns to baseline over a period of hours as the pH in cerebrospinal fluid, and hence, extracellular fluid is normalized as a result of the action of carbonic anhydrase, and in accordance with the Henderson-Hasselbach equation (26, 103). CARBON-DIOXIDE VASOREACTIVITY IN THE PRETERM BRAIN.

While vasoreactivity to PaCO2 has been shown to be present in the neonatal cerebral circulation, the underlying mechanisms differ somewhat from that in the adult brain in which the role of nitric oxide appears to predominate (Fig. 1A). In newborn piglets and the extremely preterm human brain, nitric oxide has a minor role in CO2 vasodilation (140, 190), whereas endothelium-derived prostaglandins, including PGE2, PGF1␣ and PGF2␣ seem to mediate most of the CO2 vasoreactivity (75, 141, 145). In fetal sheep, CO2-induced vasodilation is a robust and nitric oxide-independent response and is present from at least 0.65 gestation (⬃26 –28 wk human) to term (71). In healthy term human infants, CBF changes by ⬃25% per kPa of PaCO2 (159), which is similar to the value of 30% per kPa of

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review R778

CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

PaCO2 for adult humans (155). However, in very preterm infants, values between 10% and 30% per kPa of PaCO2 have been reported (155). This important CBF-PaCO2 reactivity is present in the first hours of life in spontaneously breathing, very preterm infants (156, 157), and it increases with postnatal age. Vasoconstriction increases progressively with hypocapnia until PaCO2 reaches ⬃20 mmHg, below which no further vasoconstriction occurs (27). Interestingly, in mechanically ventilated very preterm infants, CBF-PaCO2 reactivity is decreased on the first day of life (158), consistent with studies in newborn animals undergoing ventilation (178). The reason for this decreased CBF-PaCO2 reactivity during mechanical ventilation remains unclear, although CBF-PaCO2 reactivity does have an important role in breathing control. Systemic hypercapnia tends to increase CBF and “wash out” CO2 from brain tissue, thereby affecting tissue pH and the central chemoreceptor stimulus which, in turn, determines the overall ventilatory response to CO2 (3). Reduced CBF-PaCO2 reactivity typically results in an enhanced ventilatory response, as the central chemoreceptors sense a relatively lower tissue pH and provide a greater drive to ventilation. However, when CBF is markedly reduced (say, by 50%), CBF-PaCO2 reactivity is nearly abolished, and the ventilatory response to CO2 is markedly blunted, possibly as a consequence of hypoxic depression of the respiratory neurons (3). Whether these mechanisms apply to preterm infants who require mechanical ventilation, and in whom cerebral hypoxia and hypoperfusion are often present, remains unclear and is an important area of research. Oxygen vasoreactivity. MECHANISMS OF OXYGEN-VASOREACTIVITY. During hypoxia, the normal cerebrovascular response is vasodilatation, which increases CBF and serves to maintain oxygen delivery (Fig. 1A). CBF can increase as much as 2–3-fold in response to hypoxia. With hypoxia, there is local, rapid release of metabolic factors favoring vasodilation, such as perivascular hydrogen ions, potassium ions, adenosine, carbon monoxide, and prostanoids (e.g., prostacyclin, PGE2), along with decreased release of vasoconstricting factors, such as calcium ions (88, 206). Oxygen vasoreactivity depends, in part, on an intact endothelium and nitric oxide production. During development, the endothelial (143) and neuronal (140) contribution of nitric oxide to hypoxic vasodilation is relatively weak, but this increases throughout postnatal life to become more prominent in the adult (19, 43, 76), and is probably due to the increased synthesis of endothelial nitric oxide (143) and maturation of nitric oxide responsiveness (9). OXYGEN-VASOREACTIVITY IN THE PRETERM BRAIN. The ability to increase cerebral perfusion in response to acute hypoxia appears early in fetal development and has been demonstrated at less than 0.7 gestation in fetal sheep (⬃28 –30 wk gestation in humans) (143), although it is not clear whether the hypoxic vasodilatation is sufficient to fully compensate for decreased arterial oxygen content. This is likely due to the emerging and relatively underdeveloped state of the cerebral vasculature, despite the response to mediators of hypoxic dilatation, such as adenosine, already being present (143). Global hypoxia not only produces a global increase in CBF in fetal sheep, but results in a redistribution of blood flow in the brain, so that brain stem perfusion exceeds cerebellar perfusion, which, in turn, is greater than blood flow to the cerebrum (32, 144). Presumably, this robust vasodilatory response in the preterm

brain stem makes it more resistant to hypoxic injury than other brain regions (198). Conversely, hyperoxia decreases CBF, and this response to high levels of oxygen has been reported in fetal, neonatal, and adult animals and in human subjects (32, 84, 92, 214). In preterm infants, hyperoxia decreases CBF by 15–30% per kPa of PaO2 (106, 161), even exceeding the increase in arterial oxygen content, whereas in the mature adult brain, the CBF reduction to hyperoxia is proportional to the increased oxygen content in the blood (180). Importantly, decreased CBF induced by hyperoxia persists in preterm infants beyond the time that blood oxygenation was raised; e.g., CBF was lower in very preterm infants 2 h after brief exposure to oxygen in the delivery room, compared with a similar group of infants randomized to receive room air (114). As with CBF-PaCO2 reactivity, CBF-PaO2 reactivity also shows regional differences in the immature brain, with white matter being the least vasoreactive in response to changes in oxygen availability (32, 188). Cerebral oxygen vasoreactivity differs from CO2 vasoreactivity in that, unlike the response to CO2, cerebral oxygen vasoreactivity occurs outside the physiologic range of PaO2 usually experienced by preterm infants. In addition, unlike chronic hypercapnea, during which CBF returns to baseline after several hours, hypoxic vasodilation persists during sustained hypoxia (143). These observations provide grounds for insisting that hypoxia, as well as hyperoxia, should be avoided, if possible, in preterm infants. Cerebral blood flow-metabolism coupling. CBF has been shown to vary in relation to the cerebral metabolic rate of both glucose and oxygen (35), and tight coupling of cerebral function, metabolism, and blood flow is well established and has been demonstrated in numerous physiological, biochemical, and clinical studies (27, 79). Neurons and astrocytes are present in close physical proximity and are functionally coupled to the smooth muscle cells and endothelial cells of cerebral arterioles. The interactions of these cells have led to the concept that they constitute a functional unit termed the “neurovascular unit” (78). The hyperemic response to neuronal activation occurs within seconds and is highly localized (33, 107). MECHANISMS OF CEREBRAL BLOOD FLOW-METABOLISM COUPLING.

CBF-metabolism coupling (Fig. 1, A and B) is mediated by vasoactive factors that are released locally under the influence of neural activity, and include hydrogen ions, potassium ions, nitric oxide, oxygen, adenosine, calcium ions and arachadonic acid metabolites of the cyclooxygenase (COX) and cytochrome P-450 (CYP) pathways (78). Neuronal metabolic activity leads to local changes in the concentration of these vasoactive factors and, therefore, can lead to local changes in extracellular fluid osmolarity and, hence, cerebral vasodilatation, resulting in increased CBF. For example, nitric oxide, a potent vasodilator, is released in the brain as a consequence of increased synaptic activity (59), and is likely to play an important role in linking neural activity and CBF. COX-2 is present in glutamatergic neurons and COX-2-derived prostanoids participate in the increase of CBF induced by somatosensory activation (79). The importance of perivascular astrocytes, which produce CYP metabolites such as epoxyeicosatrienoic acids, as me-

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

diators of the rapid neurovascular response, has been discussed at length elsewhere (98, 225). CEREBRAL BLOOD FLOW-METABOLISM COUPLING IN THE PRETERM BRAIN. CBF-metabolism coupling is probably developed

in the preterm infants by 32 wk of gestation, if not earlier (67). Excessive neuronal activity during seizures has been associated with increased CBF and CBF velocity in term newborn and preterm infants, respectively (23, 150). Changes in the regional distribution of CBF during infancy and childhood correlate with changes in the local pattern of glucose utilization in the brain (35), supporting the concept of CBF-metabolism coupling in the developing brain. However, hemodynamic responses to neuronal activation appear to be incompletely developed in the neonatal and infant brain, compared with the adult brain. For example, in extremely preterm infants there is no correlation between CBF and spontaneous changes in the cerebral metabolic rate of oxygen (CMRO2) during the first 48 h after birth; instead, changes in COE rather than CBF meet changes in oxygen requirement arising from variations of CMRO2 (95, 219). Furthermore, brain activation studies in neonates and young infants report increases in cerebral deoxyhemoglobin, as measured by functional NIRS (120), and decreases in the blood oxygen level-dependent (BOLD) signal as detected by functional MRI (50), whereas in older infants and adults, the opposite changes in NIRS and BOLD signals occur (50, 73). Another study comparing the BOLD signal response in preterm infants, term infants, and healthy adults identified decreasing response time and increasing signal amplitude with increasing postnatal age (8). These findings suggest that in young infants the increase in cerebral oxygen consumption may be relatively greater than the corresponding increase in CBF during functional activation. Among the mechanisms that determine CBF-metabolism coupling, changes in astrocyte-dependent processes during development may explain these age-related differences in CBF and brain activity. Animal studies have found marked increases in astrocyte number, size, and their connectivity with each other and blood vessels with increased brain development (72, 91). Additionally, the capacity of the local arterioles to increase local CBF through the neurovascular coupling cascade is likely to increase with age, in accordance with histological studies showing enhanced structural development of the arterioles and capillary beds, which continues until close to term age in the human fetal cortex (87, 101, 134). Moreover, cerebral vessel density and volume have been shown to approximately double in the cortex when newborn and adult primates are compared, with the bulk of this increase occurring at the capillary level (166). It is probable that this increase in vascularity translates into faster and increased local CBF responses when cerebral metabolic activity is increased. Preterm Brain Injury and Cerebral Hemodynamics Impact of cerebral hemodynamics on germinal matrix-intraventricular hemorrhage. In the third trimester, the dense, fragile vasculature of the germinal matrix lies within an arterial end zone. As a result, the germinal matrix is liable to be exposed to ischemic conditions resulting from periods of hypoperfusion, and to rupture and hemorrhage due to fluctuations in perfusion pressure, both of which are not unusual in the

R779

preterm infant. Protection against such events is provided by prenatal glucocorticoid administration, which decreases the incidence of GM-IVH in preterm infants (48) by suppressing VEGF and angiogenesis and possibly by stabilization of the existing germinal matrix vasculature (204). In the mature brain, baseline CBF exceeds the threshold for ischemic injury by fivefold (154). However, as discussed above, global and regional CBF are significantly lower in the preterm brain, especially in the white matter. During the first 12–24 h of life, preterm infants are at significant risk of even lower cardiac output, lower systemic blood flow, and CBF (77, 97, 121), resulting in acute brain injury, including GM-IVH. The occurrence of GM-IVH is related to the nadir and duration of decreased CBF (52, 77, 97, 122) and may also occur as a “reperfusion” injury because it is strongly associated with abrupt increases in CBF (125) and fluctuating CBF (96, 135, 151, 201), presumably because there is also a loss of autoregulatory capacity as discussed above. Several studies have reported the association between impairment of pressure autoregulation in preterm infants and adverse clinical indices, including GM-IVH (125, 135, 158, 201, 220). There is also evidence that impaired autoregulation actually precedes, and may, therefore, contribute to the occurrence of the hemorrhage (4, 221). Blood pressure fluctuations are more likely to exceed the autoregulatory capacity in clinically unwell and unstable extremely preterm infants in whom the autoregulatory plateau has become very narrow or even pressure-passive. Indeed, there is increased evidence of brain injury in this group compared with well, physiologically stable infants at the same age (221). Both high and low blood CO2 levels have been implicated in causing GM-IVH (53). Hypercapnic hyperemia may trigger severe GM-IVH through vasodilation and engorgement of microvasculature (85). In addition, hypercapnea may limit the normal cerebrovasodilatory response to stimuli, such as hypotension and hypoxemia, predisposing brain tissue to hypoxicischemic insult. Doppler studies show that permissive hypercapnea [applied to reduce lung injury (124)] is associated with impaired cerebral pressure autoregulation (86). Impact of cerebral hemodynamics on periventricular leukomalacia. Conditions leading to cerebral ischemia are also associated with periventricular leukomalacia (PVL). Severe hypotension is strongly associated with white matter injury and neurodevelopmental disability (39, 54, 62), with low CBF and impaired autoregulation likely to be contributing factors. Hyperventilation and severe hypocapnia are also strongly associated with PVL and cerebral palsy (63, 68, 177), and again, cerebral hypoperfusion will likely be present in these circumstances. While a small increase in intracranial volume in the preterm infant may be accommodated by the open fontanelles and sutures without changing intracranial or cerebral perfusion pressure, hydrocephalus arising as a result of GM-IVH does reduce CBF (183) and more severely so in the white matter (41). In such cases, white matter injury arises from a combination of an acute increase in parenchymal pressure, ischemic damage, inflammation, and free radical-mediated injury (34). Compared with other brain regions, immature white matter has the least vasoreactivity to changes in pressure (22, 66, 189), and infants with evidence of impaired pressure autoregulation have a higher risk for PVL (201). Interestingly, antenatal magnesium sulfate, a tocolytic and vasodilator, is associated

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review R780

CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

with increased CBF-velocity in very preterm infants from day 1 to day 5 of life (81), which may underlie its neuroprotective role in reducing hypoxic-ischemic brain damage in preterm infants. Studies using the preterm lamb have improved our understanding of the role of inflammation and medications on cerebral oxygen metabolism and PVL. Intrauterine inflammation increases cerebral oxygen consumption (7) and decreases PaO2 with a compensatory increase in fetal CBF (171). In fetal lambs exposed to hypoxic or hypotensive stress, greater CBF fluctuations occur, and white matter injury is worse when there is superimposed intrauterine inflammation (171). The increased cerebral metabolic load and hemodynamic fluctuations imposed by intrauterine inflammation may help explain the clinical observation that the risk of white matter injury is increased when there is inflammation and/or infection present, even without a clear role for cytokines (49). On the other hand, while antenatal steroids decreases the risk of GM-IVH, their administration to pregnant ewes decreases fetal CBF without increasing COE, which may be detrimental to brain oxygenation in conjunction with other hypoxic stressors (118). Future directions. PROANGIOGENIC THERAPY FOR PRETERM BRAIN INJURY. Research for adult stroke therapies has shown that enhancement of angiogenesis may improve blood flow and support the regeneration and survival of cells after ischemic brain injury. In this regard, proangiogenic therapies may provide therapeutic opportunity also in preterm PVL, which occurs in a brain region with relatively poor vascularization. Prominent among the angiogenic factors investigated are VEGF and basic fibroblast growth factor (bFGF/FGF2). VEGF significantly augments neurogenesis and angiogenesis and reduces lesion volumes after traumatic brain injury in mice (195). Intracerebroventricular VEGF administration in a rat model of adult stroke reduces infarct size, improves neurological performance, enhances neuron survival, and stimulates angiogenesis (187). VEGF also reduces neurodegeneration and increases total vessel volume in the peri-infarct region in a neonatal rat model of stroke (46). However, VEGF overexpression in mice may worsen cerebral hemodynamics after stroke, as it induces angiogenesis with the “hemodynamic steal” phenomena, resulting in reduced blood flow in ischemic areas (208). FGF2 is a biologically active polypeptide with mitogenic, angiogenic, and neurotrophic properties. Experiments in the chick indicate that FGF signaling is important for the initiation of angioblast specification (163). Increased expression of both FGF2 mRNA and protein occurs in the ischemic cortex of rats, corresponding with robust angiogenesis (110). FGF2 protects against hypoxic-ischemic insults in vitro and in vivo and enhances recovery of rat behavior following traumatic brain injury (211). FGF2 also mediates the positive effects of exercise on the brain, as exercise-induced FGF2 expression promotes functional recovery through enhanced angiogenesis in a mouse model of hypoxic-ischemic brain injury (175). Cell-based therapies also constitute an attractive approach to enhance angiogenesis and neurogenesis after cerebral ischemia. Most notably, endothelial progenitor cells (EPCs) are able to mediate the repair of vascular injury after focal cerebral ischemia. EPCs are mobilized from bone marrow in response to acute hypoxia and are released into the circulation, where they act to maintain vascular integrity and promote tissue

neovascularization. Systemic administration of umbilical cord blood-derived EPCs in an adult mouse model of stroke is neuroprotective, with reduced infarct volume and increased focal blood flow 48 h after ischemia (137). Coadministration of EPCs and smooth muscle progenitor cells to adult mice after stroke enhances angiogenesis and vascular remodeling, enabling the maintenance of neurogenesis and improved neuroblast survival (131). Interestingly, in preterm infants, hyperoxia impairs EPC signaling and proliferation (58). In neonatal mice, hyperoxia decreases vessel density and decreases the number of EPCs in the blood and bone marrow (14), suggesting that overoxygenation in preterm infants could impair the physiological actions of EPCs, whether released endogenously from bone marrow or following exogenous administration. Perspectives and Significance Improved knowledge of cerebral hemodynamics and metabolism during development and blood flow responses to physiological and external stimuli have aided understanding of the pathogenesis of preterm brain injury. Notwithstanding the ongoing debate around the “normal” or recommended ranges for systemic PaO2, PaCO2, and arterial oxygen saturation in preterm infants (124, 185) (Supplemental Table S3), the knowledge gained so far has been translated into the clinical recommendation that these parameters should be maintained steady to stabilize CBF and avoid hypocapnia- or hyperoxiainduced cerebral hypoperfusion (108). Application of a minimal infant-handling regime, to reduce stress and discomfort and minimize fluctuations in blood pressure, is also recommended to reduce potential fluctuations in CBF (109). In the context of preterm infants having low CBF and high baseline COE, treatment of anemia with blood transfusion reduces COE (209), improves cerebral oxygenation (172), and protects against GM/IVH and PVL (20). However, the effect of some common NICU practices, such as the prone body position for ventilated infants, on cerebral oxygen delivery remain unknown, albeit prone positioning compromises cerebral perfusion and oxygenation in older infants (139, 222). Lastly, there is still relatively little understanding of the impact of neuronal activation, seizures, or drug-induced central nervous system depression on preterm cerebral hemodynamics and metabolism. In particular, it is still not known whether abnormal CBF metabolism coupling in the immature brain contributes to preterm neuropathology. Neuronal activation or depression is particularly important as many medications used in neonatal intensive care (e.g., caffeine, dopamine, morphine, midazolam, and various anticonvulsants) may alter brain activity. The effects of these medications on the coupling between CBF and metabolism remains to be explored in the preterm brain. Among the available devices to monitor cerebral perfusion, NIRS-based cerebral oximetry has extended its applicability to different research and clinical settings due to its noninvasiveness, instrument portability, and ease of use (147). However, a major limitation of most NIRS monitors is the lack of detail in the signal output from which the underlying change in cerebral oxygenation is derived. For example, reduced cerebral oxygenation may be due to reduced CBF, increased COE, increased oxygen demand, or a combination of these factors, but it is often difficult to differentiate between these possible mechanisms based on conventional NIRS measurements. Therefore,

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

findings in clinical studies regarding the relationships between NIRS measurements and clinical variables or outcomes are limited to being descriptive or associative, rather than explanatory or causative. This lack of mechanistic insight also makes the rational development of new treatments more difficult. A continuous, real-time, cotside index of cerebral autoregulatory capacity can be derived from NIRS measurements, which may help in identification of autoregulatory impairment and pathophysiology, allowing for improved and timely management (24, 60). Future prospective research demonstrating that use of these tools in guiding treatment, and evidence that maintaining predefined and targeted cerebral hemodynamic measurements improves clinically relevant outcomes for preterm infants is clearly important.

15. 16. 17. 18.

19. ACKNOWLEDGMENTS D. W. Walker is a MIMR-PHI Senior Scientist.

20.

GRANTS This work received support from the Victorian Government’s Operational Infrastructure Support Program. F. Wong is supported by the NHMRC Health Professional Research Fellowship. N. Brew is supported by National Health and Medical Research Council of Australia (NHMRC) Project Grant (APP1025626).

21. 22. 23.

REFERENCES 1. Societal Costs of Preterm Birth. In: Preterm Birth: Causes, Consequences, and Prevention, edited by Behrman R, and Butler A. Washington, DC: National Academies Press (US), 2007. 2. Aalkjaer C, Poston L. Effects of pH on vascular tension: which are the important mechanisms? J Vasc Res 33: 347–359, 1996. 3. Ainslie PN, Duffin J. Integration of cerebrovascular CO2 reactivity and chemoreflex control of breathing: mechanisms of regulation, measurement, and interpretation. Am J Physiol Regul Integr Comp Physiol 296: R1473–R1495, 2009. 4. Alderliesten T, Lemmers PM, Smarius JJ, van de Vosse RE, Baerts W, van Bel F. Cerebral oxygenation, extraction, and autoregulation in very preterm infants who develop peri-intraventricular hemorrhage. J Pediatr 162: 698 –704 e692, 2013. 5. Altman DI, Perlman JM, Volpe JJ, Powers WJ. Cerebral oxygen metabolism in newborns. Pediatrics 92: 99 –104, 1993. 6. Altman DI, Powers WJ, Perlman JM, Herscovitch P, Volpe SL, Volpe JJ. Cerebral blood flow requirement for brain viability in newborn infants is lower than in adults. Ann Neurol 24: 218 –226, 1988. 7. Andersen CC, Pillow JJ, Gill AW, Allison BJ, Moss TJ, Hooper SB, Nitsos I, Kluckow M, Polglase GR. The cerebral critical oxygen threshold of ventilated preterm lambs and the influence of antenatal inflammation. J Appl Physiol 111: 775–781, 2011. 8. Arichi T, Fagiolo G, Varela M, Melendez-Calderon A, Allievi A, Merchant N, Tusor N, Counsell SJ, Burdet E, Beckmann CF, Edwards AD. Development of BOLD signal hemodynamic responses in the human brain. NeuroImage 63: 663–673, 2012. 9. Armstead WM, Zuckerman SL, Shibata M, Parfenova H, Leffler CW. Different pial arteriolar responses to acetylcholine in the newborn and juvenile pig. J Cereb Blood Flow Metab 14: 1088 –1095, 1994. 10. Arora R, Ridha M, Lee DS, Elliott J, Rosenberg HC, Diop M, Lee TY, St. Lawrence, K. Preservation of the metabolic rate of oxygen in preterm infants during indomethacin therapy for closure of the ductus arteriosus. Pediatr Res 73: 713–718, 2013. 11. Ashwal S, Majcher JS, Vain N, Longo LD. Patterns of fetal lamb regional cerebral blood flow during and after prolonged hypoxia. Pediatr Res 14: 1104 –1110, 1980. 12. Baburamani AA, Ek CJ, Walker DW, Castillo-Melendez M. Vulnerability of the developing brain to hypoxic-ischemic damage: contribution of the cerebral vasculature to injury and repair? Front Physiol 3: 424, 2012. 13. Back SA, Riddle A, Dean J, Hohimer AR. The instrumented fetal sheep as a model of cerebral white matter injury in the premature infant. Neurotherapeutics 9: 359 –370, 2012. 14. Balasubramaniam V, Mervis CF, Maxey AM, Markham NE, Abman SH. Hyperoxia reduces bone marrow, circulating, and lung endothelial

24.

25. 26. 27. 28. 29.

30.

31. 32. 33. 34. 35. 36.

R781

progenitor cells in the developing lung: implications for the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 292: L1073–L1084, 2007. Ballabh P, Braun A, Nedergaard M. Anatomic analysis of blood vessels in germinal matrix, cerebral cortex, and white matter in developing infants. Pediatr Res 56: 117–124, 2004. Ballabh P, Xu H, Hu F, Braun A, Smith K, Rivera A, Lou N, Ungvari Z, Goldman SA, Csiszar A, Nedergaard M. Angiogenic inhibition reduces germinal matrix hemorrhage. Nat Med 13: 477–485, 2007. Baron JC. Perfusion thresholds in human cerebral ischemia: historical perspective and therapeutic implications. Cerebrovasc Dis 11 Suppl 1: 2–8, 2001. Batton B, Zhu X, Fanaroff J, Kirchner HL, Berlin S, Wilson-Costello D, Walsh M. Blood pressure, anti-hypotensive therapy, and neurodevelopment in extremely preterm infants. J Pediatr 154: 351–357, 357.e351, 2009. Bauser-Heaton HD, Bohlen HG. Cerebral microvascular dilation during hypotension and decreased oxygen tension: a role for nNOS. Am J Physiol Heart Circ Physiol 293: H2193–H2201, 2007. Bell EF, Strauss RG, Widness JA, Mahoney LT, Mock DM, Seward VJ, Cress GA, Johnson KJ, Kromer IJ, Zimmerman MB. Randomized trial of liberal versus restrictive guidelines for red blood cell transfusion in preterm infants. Pediatrics 115: 1685–1691, 2005. Borch K, Greisen G. Blood flow distribution in the normal human preterm brain. Pediatr Res 43: 28 –33, 1998. Borch K, Lou HC, Greisen G. Cerebral white matter blood flow and arterial blood pressure in preterm infants. Acta Paediatr 99: 1489 –1492, 2010. Borch K, Pryds O, Holm S, Lou H, Greisen G. Regional cerebral blood flow during seizures in neonates. J Pediatr 132: 431–435, 1998. Brady KM, Mytar JO, Lee JK, Cameron DE, Vricella LA, Thompson WR, Hogue CW, Easley RB. Monitoring cerebral blood flow pressure autoregulation in pediatric patients during cardiac surgery. Stroke 41: 1957–1962, 2010. Braun A, Xu H, Hu F, Kocherlakota P, Siegel D, Chander P, Ungvari Z, Csiszar A, Nedergaard M, Ballabh P. Paucity of pericytes in germinal matrix vasculature of premature infants. J Neurosci 27: 12012–12024, 2007. Brubakk AM, Oh W, Stonestreet BS. Prolonged hypercarbia in the awake newborn piglet: effect on brain blood flow and cardiac output. Pediatr Res 21: 29 –33, 1987. Busija DW, Heistad DD. Factors involved in the physiological regulation of the cerebral circulation. Rev Physiol Biochem Pharmacol 101: 161–211, 1984. Busija DW, Heistad DD, Marcus ML. Effects of sympathetic nerves on cerebral vessels during acute, moderate increases in arterial pressure in dogs and cats. Circ Res 46: 696 –702, 1980. Carlo WA, Finer NN, Walsh MC, Rich W, Gantz MG, Laptook AR, Yoder BA, Faix RG, Das A, Poole WK, Schibler K, Newman NS, Ambalavanan N, Frantz ID, 3rd Piazza AJ, Sanchez PJ, Morris BH, Laroia N, Phelps DL, Poindexter BB, Cotten CM, Van Meurs KP, Duara S, Narendran V, Sood BG, O’Shea TM, Bell EF, Ehrenkranz RA, Watterberg KL, Higgins RD. Target ranges of oxygen saturation in extremely preterm infants. N Engl J Med 362: 1959 –1969, 2010. Cassaglia PA, Griffiths RI, Walker AM. Sympathetic nerve activity in the superior cervical ganglia increases in response to imposed increases in arterial pressure. Am J Physiol Regul Integr Comp Physiol 294: R1255–R1261, 2008. Cassaglia PA, Griffiths RI, Walker AM. Sympathetic withdrawal augments cerebral blood flow during acute hypercapnia in sleeping lambs. Sleep 31: 1729 –1734, 2008. Cavazzuti M, Duffy TE. Regulation of local cerebral blood flow in normal and hypoxic newborn dogs. Ann Neurol 11: 247–257, 1982. Chaigneau E, Oheim M, Audinat E, Charpak S. Two-photon imaging of capillary blood flow in olfactory bulb glomeruli. Proc Natl Acad Sci USA 100: 13081–13086, 2003. Cherian S, Whitelaw A, Thoresen M, Love S. The pathogenesis of neonatal post-hemorrhagic hydrocephalus. Brain Pathol 14: 305–311, 2004. Chugani HT, Phelps ME, Mazziotta JC. Positron emission tomography study of human brain functional development. Ann Neurol 22: 487–497, 1987. Collins MP, Lorenz JM, Jetton JR, Paneth N. Hypocapnia and other ventilation-related risk factors for cerebral palsy in low birth weight infants. Pediatr Res 50: 712–719, 2001.

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review R782

CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

37. Cools F, Askie LM, Offringa M, Asselin JM, Calvert SA, Courtney SE, Dani C, Durand DJ, Gerstmann DR, Henderson-Smart DJ, Marlow N, Peacock JL, Pillow JJ, Soll RF, Thome UH, Truffert P, Schreiber MD, Van Reempts P, Vendettuoli V, Vento G. Elective high-frequency oscillatory versus conventional ventilation in preterm infants: a systematic review and meta-analysis of individual patients’ data. Lancet 375: 2082–2091, 2010. 38. Costeloe KL, Hennessy EM, Haider S, Stacey F, Marlow N, Draper ES. Short term outcomes after extreme preterm birth in England: comparison of two birth cohorts in 1995 and 2006 (the EPICure studies). BMJ 345: e7976, 2012. 39. Cunningham S, Symon AG, Elton RA, Zhu C, McIntosh N. Intraarterial blood pressure reference ranges, death and morbidity in very low birthweight infants during the first seven days of life. Early Hum Dev 56: 151–165, 1999. 40. Cuttini M, Kaminski M, Garel M, Lenoir S, Saracci R. End-of-life decisions in neonatal intensive care. Lancet 356: 2190 –2191, 2000. 41. da Silva MC, Michowicz S, Drake JM, Chumas PD, Tuor UI. Reduced local cerebral blood flow in periventricular white matter in experimental neonatal hydrocephalus-restoration with CSF shunting. J Cereb Blood Flow Metab 15: 1057–1065, 1995. 42. Doyle LW, Roberts G, Anderson PJ. Outcomes at age 2 years of infants ⬍28 weeks’ gestational age born in Victoria in 2005. J Pediatr 156: 49 –53, e41, 2010. 43. Dreier JP, Korner K, Gorner A, Lindauer U, Weih M, Villringer A, Dirnagl U. Nitric oxide modulates the CBF response to increased extracellular potassium. J Cereb Blood Flow Metab 15: 914 –919, 1995. 44. du Plessis AJ. Cerebrovascular injury in premature infants: current understanding and challenges for future prevention. Clinics Perinatol 35: 609 –641, v, 2008. 45. du Plessis AJ, Volpe JJ. Perinatal brain injury in the preterm and term newborn. Curr Opin Neurol 15: 151–157, 2002. 46. Dzietko M, Derugin N, Wendland MF, Vexler ZS, Ferriero DM. Delayed VEGF treatment enhances angiogenesis and recovery after neonatal focal rodent stroke. Transl Stroke Res 4: 189 –200, 2013. 47. El-Khoury N, Braun A, Hu F, Pandey M, Nedergaard M, Lagamma EF, Ballabh P. Astrocyte end-feet in germinal matrix, cerebral cortex, and white matter in developing infants. Pediatr Res 59: 673–679, 2006. 48. Elimian A, Garry D, Figueroa R, Spitzer A, Wiencek V, Quirk JG. Antenatal betamethasone compared with dexamethasone (betacode trial): a randomized controlled trial. Obstet Gynecol 110: 26 –30, 2007. 49. Ellison VJ, Mocatta TJ, Winterbourn CC, Darlow BA, Volpe JJ, Inder TE. The relationship of CSF and plasma cytokine levels to cerebral white matter injury in the premature newborn. Pediatr Res 57: 282–286, 2005. 50. Erberich SG, Panigrahy A, Friedlich P, Seri I, Nelson MD, Gilles F. Somatosensory lateralization in the newborn brain. NeuroImage 29: 155–161, 2006. 51. Eucker SA, Hoffman BD, Natesh R, Ralston J, Armstead WM, Margulies SS. Development of a fluorescent microsphere technique for rapid histological determination of cerebral blood flow. Brain Res 1326: 128 –134, 2010. 52. Evans N, Kluckow M. Early ductal shunting and intraventricular haemorrhage in ventilated preterm infants. Arch Dis Child Fetal Neonatal Ed 75: F183–F186, 1996. 53. Fabres J, Carlo WA, Phillips V, Howard G, Ambalavanan N. Both extremes of arterial carbon dioxide pressure and the magnitude of fluctuations in arterial carbon dioxide pressure are associated with severe intraventricular hemorrhage in preterm infants. Pediatrics 119: 299 –305, 2007. 54. Fanaroff JM, Wilson-Costello DE, Newman NS, Montpetite MM, Fanaroff AA. Treated hypotension is associated with neonatal morbidity and hearing loss in extremely low birth weight infants. Pediatrics 117: 1131–1135, 2006. 55. Fong GH. Regulation of angiogenesis by oxygen sensing mechanisms. J Mol Med 87: 549 –560, 2009. 56. Fowlie PW, Davis PG, McGuire W. Prophylactic intravenous indomethacin for preventing mortality and morbidity in preterm infants. Cochrane Database Syst Rev CD000174, 2010. 57. Fujimori K, Honda S, Sanpei M, Sato A. Effects of exogenous big endothelin-1 on regional blood flow in fetal lambs. Obstet Gynecol 106: 818 –823, 2005. 58. Fujinaga H, Baker CD, Ryan SL, Markham NE, Seedorf GJ, Balasubramaniam V, Abman SH. Hyperoxia disrupts vascular endothelial

59.

60.

61.

62.

63. 64. 65.

66. 67.

68. 69.

70.

71.

72.

73.

74.

75.

76.

77.

78. 79. 80.

81.

82.

growth factor-nitric oxide signaling and decreases growth of endothelial colony-forming cells from preterm infants. Am J Physiol Lung Cell Mol Physiol 297: L1160 –L1169, 2009. Gally JA, Montague PR, Reeke GN Jr, Edelman GM. The NO hypothesis: possible effects of a short-lived, rapidly diffusible signal in the development and function of the nervous system. Proc Natl Acad Sci USA 87: 3547–3551, 1990. Gilmore MM, Stone BS, Shepard JA, Czosnyka M, Easley RB, Brady KM. Relationship between cerebrovascular dysautoregulation and arterial blood pressure in the premature infant. J Perinatol 31: 722–729, 2011. Gleason CA, Hamm C, Jones MD Jr. Cerebral blood flow, oxygenation, and carbohydrate metabolism in immature fetal sheep in utero. Am J Physiol Regul Integr Comp Physiol 256: R1264 –R1268, 1989. Goldstein RF, Thompson RJ Jr, Oehler JM, Brazy JE. Influence of acidosis, hypoxemia, and hypotension on neurodevelopmental outcome in very low birth weight infants. Pediatrics 95: 238 –243, 1995. Greisen G. Autoregulation of cerebral blood flow in newborn babies. Early Hum Dev 81: 423–428, 2005. Greisen G. Cerebral blood flow in preterm infants during the first week of life. Acta Paediatr Scand 75: 43–51, 1986. Greisen G. Methods for assessing cerebral blood flow and metabolism of the developing brain. In: Fetal and Neonatal Neurology and Neurosurgery, edited by Levene MI, and Lilford RJ. New York: Churchill Livingstone, 1995, p. 145–162. Greisen G, Borch K. White matter injury in the preterm neonate: the role of perfusion. Dev Neurosci 23: 209 –212, 2001. Greisen G, Hellstrom-Vestas L, Lou H, Rosen I, Svenningsen N. Sleep-waking shifts and cerebral blood flow in stable preterm infants. Pediatr Res 19: 1156 –1159, 1985. Greisen G, Munck H, Lou H. Severe hypocarbia in preterm infants and neurodevelopmental deficit. Acta Paediatr Scand 76: 401–404, 1987. Greisen G, Pryds O. Low CBF, discontinuous EEG activity, and periventricular brain injury in ill, preterm neonates. Brain Dev 11: 164 –168, 1989. Hagen EW, Sadek-Badawi M, Carlton DP, Palta M. Permissive hypercapnia and risk for brain injury and developmental impairment. Pediatrics 122: e583–e589, 2008. Harris AP, Ohata H, Koehler RC. Role of nitric oxide in cerebrovascular reactivity to NMDA and hypercapnia during prenatal development in sheep. Int J Dev Neurosci 26: 47–55, 2008. Harris JJ, Reynell C, Attwell D. The physiology of developmental changes in BOLD functional imaging signals. Dev Cogn Neurosci 1: 199 –216, 2011. Heekeren HR, Obrig H, Wenzel R, Eberle K, Ruben J, Villringer K, Kurth R, Villringer A. Cerebral haemoglobin oxygenation during sustained visual stimulation—a near-infrared spectroscopy study. Philos Trans R Soc Lond B Biol Sci 352: 743–750, 1997. Helou S, Koehler RC, Gleason CA, Jones MD Jr, Traystman RJ. Cerebrovascular autoregulation during fetal development in sheep. Am J Physiol Heart Circ Physiol 266: H1069 –H1074, 1994. Hsu P, Shibata M, Leffler CW. Prostanoid synthesis in response to high CO2 in newborn pig brain microvascular endothelial cells. Am J Physiol Heart Circ Physiol 264: H1485–H1492, 1993. Hudetz AG, Shen H, Kampine JP. Nitric oxide from neuronal NOS plays critical role in cerebral capillary flow response to hypoxia. Am J Physiol Heart Circ Physiol 274: H982–H989, 1998. Hunt RW, Evans N, Rieger I, Kluckow M. Low superior vena cava flow and neurodevelopment at 3 years in very preterm infants. J Pediatr 145: 588 –592, 2004. Iadecola C. Neurovascular regulation in the normal brain and in Alzheimer’s disease. Nat Rev Neurosci 5: 347–360, 2004. Iadecola C, Nedergaard M. Glial regulation of the cerebral microvasculature. Nat Neurosci 10: 1369 –1376, 2007. Ibrahim M, Ho SK, Yeo CL. Restrictive versus liberal red blood cell transfusion thresholds in very low birth weight infants: A systematic review and meta-analysis. J Paediatr Child Health 50: 122–130, 2013. Imamoglu EY, Gursoy T, Karatekin G, Ovali F. Effects of antenatal magnesium sulfate treatment on cerebral blood flow velocities in preterm neonates. J Perinatol 34: 192–196, 2014. Jayasinghe D, Gill AB, Levene MI. CBF reactivity in hypotensive and normotensive preterm infants. Pediatr Res 54: 848 –853, 2003.

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS 83. Jones MD Jr, Rosenberg AA, Simmons MA, Molteni RA, Koehler RC, Traystman RJ. Oxygen delivery to the brain before and after birth. Science 216: 324 –325, 1982. 84. Jones MD Jr, Traystman RJ. Cerebral oxygenation of the fetus, newborn, and adult. Sem Perinatol 8: 205–216, 1984. 85. Kaiser JR, Gauss CH, Pont MM, Williams DK. Hypercapnia during the first 3 days of life is associated with severe intraventricular hemorrhage in very low birth weight infants. J Perinatol 26: 279 –285, 2006. 86. Kaiser JR, Gauss CH, Williams DK. The effects of hypercapnia on cerebral autoregulation in ventilated very low birth weight infants. Pediatr Res 58: 931–935, 2005. 87. Kamei A, Houdou S, Mito T, Konomi H, Takashima S. Developmental change in type VI collagen in human cerebral vessels. Pediatr Neurol 8: 183–186, 1992. 88. Kanu A, Leffler CW. Carbon monoxide and Ca2⫹-activated K⫹ channels in cerebral arteriolar responses to glutamate and hypoxia in newborn pigs. Am J Physiol Heart Circ Physiol 293: H3193–H3200, 2007. 89. Kanu A, Whitfield J, Leffler CW. Carbon monoxide contributes to hypotension-induced cerebrovascular vasodilation in piglets. Am J Physiol Heart Circ Physiol 291: H2409 –H2414, 2006. 90. Kapadia VS, Chalak LF, Sparks JE, Allen JR, Savani RC, Wyckoff MH. Resuscitation of preterm neonates with limited versus high oxygen strategy. Pediatrics 132: e1488 –e1496, 2013. 91. Kaur C, Ling EA, Wong WC. Development of the various glial cell types in the cerebral cortex of postnatal rats. Acta Anat (Basel) 136: 204 –210, 1989. 92. Kennedy C, Grave GD, Sokoloff L. Alterations of local cerebral blood flow due to exposure of newborn puppies to 80 –90 per cent oxygen. Eur Neurol 6: 137–140, 1971. 93. Kety SS, Schmidt CF. The effects of active and passive hyperventilation on cerebral blood flow, cerebral oxygen consumption, cardiac output, and blood pressure of normal young men. J Clin Invest 25: 107–119, 1946. 94. Khwaja O, Volpe JJ. Pathogenesis of cerebral white matter injury of prematurity. Arch Dis Child Fetal Neonatal Ed 93: F153–F161, 2008. 95. Kissack CM, Garr R, Wardle SP, Weindling AM. Cerebral fractional oxygen extraction is inversely correlated with oxygen delivery in the sick, newborn, preterm infant. J Cereb Blood Flow Metab 25: 545–553, 2005. 96. Kissack CM, Garr R, Wardle SP, Weindling AM. Postnatal changes in cerebral oxygen extraction in the preterm infant are associated with intraventricular hemorrhage and hemorrhagic parenchymal infarction but not periventricular leukomalacia. Pediatr Res 56: 111–116, 2004. 97. Kluckow M, Evans N. Low superior vena cava flow and intraventricular haemorrhage in preterm infants. Arch Dis Child Fetal Neonatal Ed 82: F188 –F194, 2000. 98. Koehler RC, Gebremedhin D, Harder DR. Role of astrocytes in cerebrovascular regulation. J Appl Physiol (1985) 100: 307–317, 2006. 99. Koller A, Toth P. Contribution of flow-dependent vasomotor mechanisms to the autoregulation of cerebral blood flow. J Vasc Res 49: 375–389, 2012. 100. Kontos HA, Wei EP. Oxygen-dependent mechanisms in cerebral autoregulation. Ann Biomed Eng 13: 329 –334, 1985. 101. Kuban KC, Gilles FH. Human telencephalic angiogenesis. Ann Neurol 17: 539 –548, 1985. 102. Larrivee B, Freitas C, Suchting S, Brunet I, Eichmann A. Guidance of vascular development: lessons from the nervous system. Circ Res 104: 428 –441, 2009. 103. Lassen NA. Control of cerebral circulation in health and disease. Circ Res 34: 749 –760, 1974. 104. Lassen NA, Christensen MS. Physiology of cerebral blood flow. Br J Anaesth 48: 719 –734, 1976. 105. Lavi S, Egbarya R, Lavi R, Jacob G. Role of nitric oxide in the regulation of cerebral blood flow in humans: chemoregulation versus mechanoregulation. Circulation 107: 1901–1905, 2003. 106. Leahy FA, Cates D, MacCallum M, Rigatto H. Effect of CO2 and 100% O2 on cerebral blood flow in preterm infants. J Appl Physiol 48: 468 –472, 1980. 107. LeDoux JE, Thompson ME, Iadecola C, Tucker LW, Reis DJ. Local cerebral blood flow increases during auditory and emotional processing in the conscious rat. Science 221: 576 –578, 1983. 108. Liem KD, Greisen G. Monitoring of cerebral haemodynamics in newborn infants. Early Hum Dev 86: 155–158, 2010. 109. Limperopoulos C, Gauvreau KK, O’Leary H, Moore M, Bassan H, Eichenwald EC, Soul JS, Ringer SA, Di Salvo DN, du Plessis AJ.

110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

120.

121.

122.

123.

124. 125. 126.

127.

128.

129.

130.

131.

R783

Cerebral hemodynamic changes during intensive care of preterm infants. Pediatrics 122: e1006 –e1013, 2008. Lin TN, Wang CK, Cheung WM, Hsu CY. Induction of angiopoietin and Tie receptor mRNA expression after cerebral ischemia-reperfusion. J Cereb Blood Flow Metab 20: 387–395, 2000. Loos N, Grant DA, Wild J, Paul S, Barfield C, Zoccoli G, Franzini C, Walker AM. Sympathetic nervous control of the cerebral circulation in sleep. J Sleep Res 14: 275–283, 2005. Lou HC, Lassen NA, Friis-Hansen B. Impaired autoregulation of cerebral blood flow in the distressed newborn infant. J Pediatr 94: 118 –121, 1979. Lucitti JL, Mackey JK, Morrison JC, Haigh JJ, Adams RH, Faber JE. Formation of the collateral circulation is regulated by vascular endothelial growth factor-A and a disintegrin and metalloprotease family members 10 and 17. Circ Res 111: 1539 –1550, 2012. Lundstrom KE, Pryds O, Greisen G. Oxygen at birth and prolonged cerebral vasoconstriction in preterm infants. Arch Dis Child Fetal Neonatal Ed 73: F81–F86, 1995. Mandala M, Pedatella AL, Morales Palomares S, Cipolla MJ, Osol G. Maturation is associated with changes in rat cerebral artery structure, biomechanical properties and tone. Acta Physiol (Oxf) 205: 363–371, 2012. Marlow N, Greenough A, Peacock JL, Marston L, Limb ES, Johnson AH, Calvert SA. Randomised trial of high frequency oscillatory ventilation or conventional ventilation in babies of gestational age 28 weeks or less: respiratory and neurological outcomes at 2 years. Arch Dis Child Fetal Neonatal Ed 91: F320 –F326, 2006. Martin JA, Kung HC, Mathews TJ, Hoyert DL, Strobino DM, Guyer B, Sutton SR. Annual summary of vital statistics: 2006. Pediatrics 121: 788 –801, 2008. McCallum J, Smith N, Schwab M, Coksaygan T, Reinhardt B, Nathanielsz P, Richardson BS. Effects of antenatal glucocorticoids on cerebral substrate metabolism in the preterm ovine fetus. Am J Obstet Gynecol 198: 105 e101–e109, 2008. McPherson RW, Kirsch JR, Ghaly RF, Traystman RJ. Effect of nitric oxide synthase inhibition on the cerebral vascular response to hypercapnia in primates. Stroke 26: 682–687, 1995. Meek JH, Firbank M, Elwell CE, Atkinson J, Braddick O, Wyatt JS. Regional hemodynamic responses to visual stimulation in awake infants. Pediatr Res 43: 840 –843, 1998. Meek JH, Tyszczuk L, Elwell CE, Wyatt JS. Cerebral blood flow increases over the first three days of life in extremely preterm neonates. Arch Dis Child Fetal Neonatal Ed 78: F33–F37, 1998. Meek JH, Tyszczuk L, Elwell CE, Wyatt JS. Low cerebral blood flow is a risk factor for severe intraventricular haemorrhage. Arch Dis Child Fetal Neonatal Ed 81: F15–F18, 1999. Menon DK, Coles JP, Gupta AK, Fryer TD, Smielewski P, Chatfield DA, Aigbirhio F, Skepper JN, Minhas PS, Hutchinson PJ, Carpenter TA, Clark JC, Pickard JD. Diffusion limited oxygen delivery following head injury. Crit Care Med 32: 1384 –1390, 2004. Miller JD, Carlo WA. Safety and effectiveness of permissive hypercapnia in the preterm infant. Curr Opin Pediatr 19: 142–144, 2007. Milligan DW. Failure of autoregulation and intraventricular haemorrhage in preterm infants. Lancet 1: 896 –898, 1980. Miranda MJ, Olofsson K, Sidaros K. Noninvasive measurements of regional cerebral perfusion in preterm and term neonates by magnetic resonance arterial spin labeling. Pediatr Res 60: 359 –363, 2006. Moore T, Hennessy EM, Myles J, Johnson SJ, Draper ES, Costeloe KL, Marlow N. Neurological and developmental outcome in extremely preterm children born in England in 1995 and 2006: the EPICure studies. Br Med J 345: e7961, 2012. Morita Y, Hardebo JE, Bouskela E. Influence of cerebrovascular parasympathetic nerves on resting cerebral blood flow, spontaneous vasomotion, autoregulation, hypercapnic vasodilation and sympathetic vasoconstriction. J Auton Nerv Syst 49 Suppl. S9 –S14, 1994. Munro MJ, Walker AM, Barfield CP. Hypotensive extremely low birth weight infants have reduced cerebral blood flow. Pediatrics 114: 1591–1596, 2004. Nelson MD Jr, Gonzalez-Gomez I, Gilles FH. Dyke Award The search for human telencephalic ventriculofugal arteries AJNR. Am J Neuroradiol 12: 215–222, 1991. Nih LR, Deroide N, Lere-Dean C, Lerouet D, Soustrat M, Levy BI, Silvestre JS, Merkulova-Rainon T, Pocard M, Margaill I, Kubis N. Neuroblast survival depends on mature vascular network formation after

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review R784

132.

133.

134.

135.

136.

137.

138.

139.

140.

141.

142. 143. 144.

145.

146.

147. 148.

149.

150.

151.

152.

153.

CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

mouse stroke: role of endothelial and smooth muscle progenitor cell co-administration. Eur J Neurosci 35: 1208 –1217, 2012. Nongena P, Ederies A, Azzopardi DV, Edwards AD. Confidence in the prediction of neurodevelopmental outcome by cranial ultrasound and MRI in preterm infants. Arch Dis Child Fetal Neonatal Ed 95: F388 – F390, 2010. Noone MA, Sellwood M, Meek JH, Wyatt JS. Postnatal adaptation of cerebral blood flow using near infrared spectroscopy in extremely preterm infants undergoing high-frequency oscillatory ventilation. Acta Paediatr 92: 1079 –1084, 2003. Norman MG, O’Kusky JR. The growth and development of microvasculature in human cerebral cortex. J Neuropathol Exp Neurol 45: 222– 232, 1986. O’Leary H, Gregas MC, Limperopoulos C, Zaretskaya I, Bassan H, Soul JS, Di Salvo DN, du Plessis AJ. Elevated cerebral pressure passivity is associated with prematurity-related intracranial hemorrhage. Pediatrics 124: 302–309, 2009. Ohlsson A, Shah SS. Ibuprofen for the prevention of patent ductus arteriosus in preterm and/or low birth weight infants. Cochrane Database Syst Rev CD004213, 2011. Ohta T, Kikuta K, Imamura H, Takagi Y, Nishimura M, Arakawa Y, Hashimoto N, Nozaki K. Administration of ex vivo-expanded bone marrow-derived endothelial progenitor cells attenuates focal cerebral ischemia-reperfusion injury in rats. Neurosurgery 59: 679 –686; discussion 679 – 686, 2006. Osborn DA, Evans N, Kluckow M, Bowen JR, Rieger I. Low superior vena cava flow and effect of inotropes on neurodevelopment to 3 years in preterm infants. Pediatrics 120: 372–380, 2007. Pamphlett R, Raisanen J, Kum-Jew S. Vertebral artery compression resulting from head movement: a possible cause of the sudden infant death syndrome. Pediatrics 103: 460 –468, 1999. Patel J, Pryds O, Roberts I, Harris D, Edwards AD. Limited role for nitric oxide in mediating cerebrovascular control of newborn piglets. Arch Dis Child Fetal Neonatal Ed 75: F82–F86, 1996. Patel J, Roberts I, Azzopardi D, Hamilton P, Edwards AD. Randomized double-blind controlled trial comparing the effects of ibuprofen with indomethacin on cerebral hemodynamics in preterm infants with patent ductus arteriosus. Pediatr Res 47: 36 –42, 2000. Paulson OB, Strandgaard S, Edvinsson L. Cerebral autoregulation. Cerebrovasc Brain Metab Rev 2: 161–192, 1990. Pearce W. Hypoxic regulation of the fetal cerebral circulation. J Appl Physiol 100: 731–738, 2006. Peeters LL, Sheldon RE, Jones MD Jr, Makowski EL, Meschia G. Blood flow to fetal organs as a function of arterial oxygen content. Am J Obstet Gynecol 135: 637–646, 1979. Pellicer A, Aparicio M, Cabanas F, Valverde E, Quero J, Stiris TA. Effect of the cyclo-oxygenase blocker ibuprofen on cerebral blood volume and cerebral blood flow during normocarbia and hypercarbia in newborn piglets. Acta Paediatr 88: 82–88, 1999. Pellicer A, Bravo MC, Madero R, Salas S, Quero J, Cabanas F. Early systemic hypotension and vasopressor support in low birth weight infants: impact on neurodevelopment. Pediatrics 123: 1369 –1376, 2009. Pellicer A, Bravo Mdel C. Near-infrared spectroscopy: a methodologyfocused review. Semin Fetal Neonatal Med 16: 42–49, 2011. Pellicer A, Valverde E, Elorza MD, Madero R, Gaya F, Quero J, Cabanas F. Cardiovascular support for low birth weight infants and cerebral hemodynamics: a randomized, blinded, clinical trial. Pediatrics 115: 1501–1512, 2005. Pena JP, Tomimatsu T, Hatran DP, McGill LL, Longo LD. Cerebral blood flow and oxygenation in ovine fetus: responses to superimposed hypoxia at both low and high altitude. J Physiol 578: 359 –370, 2007. Perlman JM, Herscovitch P, Kreusser KL, Raichle ME, Volpe JJ. Positron emission tomography in the newborn: effect of seizure on regional cerebral blood flow in an asphyxiated infant. Neurology 35: 244 –247, 1985. Perlman JM, McMenamin JB, Volpe JJ. Fluctuating cerebral bloodflow velocity in respiratory-distress syndrome. Relation to the development of intraventricular hemorrhage. N Engl J Med 309: 204 –209, 1983. Phillis JW, Lungu CL, Barbu DE, O’Regan MH. Adenosine’s role in hypercapnia-evoked cerebral vasodilation in the rat. Neurosci Lett 365: 6 –9, 2004. Pichler G, Binder C, Avian A, Beckenbach E, Schmolzer GM, Urlesberger B. Reference ranges for regional cerebral tissue oxygen

154.

155. 156. 157. 158. 159. 160. 161. 162. 163. 164.

165.

166. 167. 168.

169.

170. 171.

172.

173. 174.

175.

saturation and fractional oxygen extraction in neonates during immediate transition after birth. J Pediatr 163: 1558 –1563, 2013. Powers WJ, Grubb RL Jr, Darriet D, Raichle ME. Cerebral blood flow and cerebral metabolic rate of oxygen requirements for cerebral function and viability in humans. J Cereb Blood Flow Metab 5: 600 –608, 1985. Pryds O. Control of cerebral circulation in the high-risk neonate. Ann Neurol 30: 321–329, 1991. Pryds O, Andersen GE, Friis-Hansen B. Cerebral blood flow reactivity in spontaneously breathing, preterm infants shortly after birth. Acta Paediatr Scand 79: 391–396, 1990. Pryds O, Greisen G. Effect of PaCO2 and haemoglobin concentration on day to day variation of CBF in preterm neonates. Acta Paediatr Scand Suppl 360: 33–36, 1989. Pryds O, Greisen G, Lou H, Friis-Hansen B. Heterogeneity of cerebral vasoreactivity in preterm infants supported by mechanical ventilation. J Pediatr 115: 638 –645, 1989. Pryds O, Greisen G, Lou H, Friis-Hansen B. Vasoparalysis associated with brain damage in asphyxiated term infants. J Pediatr 117: 119 –125, 1990. Rabi Y, Singhal N, Nettel-Aguirre A. Room-air versus oxygen administration for resuscitation of preterm infants: the ROAR study. Pediatrics 128: e374 –e381, 2011. Rahilly PM. Effects of sleep state and feeding on cranial blood flow of the human neonate. Arch Dis Child 55: 265–270, 1980. Raju TN. Some animal models for the study of perinatal asphyxia. Biol Neonate 62: 202–214, 1992. Ribatti D. Genetic and epigenetic mechanisms in the early development of the vascular system. J Anat 208: 139 –152, 2006. Richardson BS, Carmichael L, Homan J, Gagnon R. Cerebral oxidative metabolism in lambs during perinatal period: relationship to electrocortical state. Am J Physiol Regul Integr Comp Physiol 257: R1251– R1257, 1989. Riddle A, Luo NL, Manese M, Beardsley DJ, Green L, Rorvik DA, Kelly KA, Barlow CH, Kelly JJ, Hohimer AR, Back SA. Spatial heterogeneity in oligodendrocyte lineage maturation and not cerebral blood flow predicts fetal ovine periventricular white matter injury. J Neurosci 26: 3045–3055, 2006. Risser L, Plouraboue F, Cloetens P, Fonta C. A 3D-investigation shows that angiogenesis in primate cerebral cortex mainly occurs at capillary level. Int J Dev Neurosci 27: 185–196, 2009. Roberts G, Burnett AC, Lee KJ, Cheong J, Wood SJ, Anderson PJ, Doyle LW. Quality of life at age 18 years after extremely preterm birth in the post-surfactant era. J Pediatr 163: 1008 –1013 e1001, 2013. Robinson JM, Lancaster JR Jr. Hemoglobin-mediated, hypoxiainduced vasodilation via nitric oxide: mechanism(s) and physiologic versus pathophysiologic relevance. Am J Respir Cell Mol Biol 32: 257–261, 2005. Saigal S, den Ouden L, Wolke D, Hoult L, Paneth N, Streiner DL, Whitaker A, Pinto-Martin J. School-age outcomes in children who were extremely low birth weight from four international populationbased cohorts. Pediatrics 112: 943–950, 2003. Saigal S, Doyle LW. An overview of mortality and sequelae of preterm birth from infancy to adulthood. Lancet 371: 261–269, 2008. Saito M, Matsuda T, Okuyama K, Kobayashi Y, Kitanishi R, Hanita T, Okamura K. Effect of intrauterine inflammation on fetal cerebral hemodynamics and white-matter injury in chronically instrumented fetal sheep. Am J Obstet Gynecol 200: 663 e661–e611, 2009. Sandal G, Oguz SS, Erdeve O, Akar M, Uras N, Dilmen U. Assessment of red blood cell transfusion and transfusion duration on cerebral and mesenteric oxygenation using near-infrared spectroscopy in preterm infants with symptomatic anemia. Transfusion In press. Sassano-Higgins S, Friedlich P, Seri I. A meta-analysis of dopamine use in hypotensive preterm infants: blood pressure and cerebral hemodynamics. J Perinatol 31: 647–655, 2011. Sentilhes L, Michel C, Lecourtois M, Catteau J, Bourgeois P, Laudenbach V, Marret S, Laquerriere A. Vascular endothelial growth factor and its high-affinity receptor (VEGFR-2) are highly expressed in the human forebrain and cerebellum during development. J Neuropathol Exp Neurol 69: 111–128, 2010. Seo JH, Yu JH, Suh H, Kim MS, Cho SR. Fibroblast growth factor-2 induced by enriched environment enhances angiogenesis and motor function in chronic hypoxic-ischemic brain injury. PLos One 8: e74405, 2013.

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS 176. Seri I, Rudas G, Bors Z, Kanyicska B, Tulassay T. Effects of low-dose dopamine infusion on cardiovascular and renal functions, cerebral blood flow, and plasma catecholamine levels in sick preterm neonates. Pediatr Res 34: 742–749, 1993. 177. Shankaran S, Langer JC, Kazzi SN, Laptook AR, Walsh M. Cumulative index of exposure to hypocarbia and hyperoxia as risk factors for periventricular leukomalacia in low birth weight infants. Pediatrics 118: 1654 –1659, 2006. 178. Shapiro HM, Greenberg JH, Naughton KV, Reivich M. Heterogeneity of local cerebral blood flow—PaCO2 sensitivity in neonatal dogs. J Appl Physiol 49: 113–118, 1980. 179. Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, Vincent P, Pumiglia K, Temple S. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science 304: 1338 –1340, 2004. 180. Shinozuka T, Nemoto EM, Winter PM. Mechanisms of cerebrovascular O2 sensitivity from hyperoxia to moderate hypoxia in the rat. J Cereb Blood Flow Metab 9: 187–195, 1989. 181. Sirc J, Dempsey EM, Miletin J. Cerebral tissue oxygenation index, cardiac output and superior vena cava flow in infants with birth weight less than 1250 grams in the first 48 hours of life. Early Hum Dev 89: 449 –452, 2013. 182. Soul JS, Hammer PE, Tsuji M, Saul JP, Bassan H, Limperopoulos C, Disalvo DN, Moore M, Akins P, Ringer S, Volpe JJ, Trachtenberg F, du Plessis AJ. Fluctuating pressure-passivity is common in the cerebral circulation of sick premature infants. Pediatr Res 61: 467–473, 2007. 183. Soul JS, Taylor GA, Wypij D, Duplessis AJ, Volpe JJ. Noninvasive detection of changes in cerebral blood flow by near-infrared spectroscopy in a piglet model of hydrocephalus. Pediatr Res 48: 445–449, 2000. 184. Steggerda SJ, De Bruine FT, van den Berg-Huysmans AA, Rijken M, Leijser LM, Walther FJ, van Wezel-Meijler G. Small cerebellar hemorrhage in preterm infants: perinatal and postnatal factors and outcome. Cerebellum 12: 794 –801, 2013. 185. Stenson BJ, Tarnow-Mordi WO, Darlow BA, Simes J, Juszczak E, Askie L, Battin M, Bowler U, Broadbent R, Cairns P, Davis PG, Deshpande S, Donoghoe M, Doyle L, Fleck BW, Ghadge A, Hague W, Halliday HL, Hewson M, King A, Kirby A, Marlow N, Meyer M, Morley C, Simmer K, Tin W, Wardle SP, Brocklehurst P. Oxygen saturation and outcomes in preterm infants. N Engl J Med 368: 2094 – 2104, 2013. 186. Sun H, Cheng R, Kang W, Xiong H, Zhou C, Zhang Y, Wang X, Zhu C. High-frequency oscillatory ventilation versus synchronized intermittent mandatory ventilation plus pressure support in preterm infants with severe respiratory distress syndrome. Respir Care 59: 159 –169, 2014. 187. Sun Y, Jin K, Xie L, Childs J, Mao XO, Logvinova A, Greenberg DA. VEGF-induced neuroprotection, neurogenesis, and angiogenesis after focal cerebral ischemia. J Clin Invest 111: 1843–1851, 2003. 188. Szymonowicz W, Walker AM, Cussen L, Cannata J, Yu VY. Developmental changes in regional cerebral blood flow in fetal and newborn lambs. Am J Physiol Heart Circ Physiol 254: H52–H58, 1988. 189. Szymonowicz W, Walker AM, Yu VY, Stewart ML, Cannata J, Cussen L. Regional cerebral blood flow after hemorrhagic hypotension in the preterm, near-term, and newborn lamb. Pediatr Res 28: 361–366, 1990. 190. Takei Y, Edwards AD, Lorek A, Peebles DM, Belai A, Cope M, Delpy DT, Reynolds EO. Effects of N-omega-nitro-L-arginine methyl ester on the cerebral circulation of newborn piglets quantified in vivo by near-infrared spectroscopy. Pediatr Res 34: 354 –359, 1993. 191. Talman WT, Dragon DN, Ohta H. Baroreflexes influence autoregulation of cerebral blood flow during hypertension. Am J Physiol Heart Circ Physiol 267: H1183–H1189, 1994. 192. Tan CO, Hamner JW, Taylor JA. The role of myogenic mechanisms in human cerebrovascular regulation. J Physiol 591: 5095–5105, 2013. 193. Tanner SF, Cornette L, Ramenghi LA, Miall LS, Ridgway JP, Smith MA, Levene MI. Cerebral perfusion in infants and neonates: preliminary results obtained using dynamic susceptibility contrast enhanced magnetic resonance imaging. Arch Dis Child Fetal Neonatal Ed 88: F525–F530, 2003. 194. Tavazoie M, Van der Veken L, Silva-Vargas V, Louissaint M, Colonna L, Zaidi B, Garcia-Verdugo JM, Doetsch F. A specialized vascular niche for adult neural stem cells. Cell Stem Cell 3: 279 –288, 2008. 195. Thau-Zuchman O, Shohami E, Alexandrovich AG, Leker RR. Vascular endothelial growth factor increases neurogenesis after traumatic brain injury. J Cereb Blood Flow Metab 30: 1008 –1016, 2010.

R785

196. Thome UH, Carroll W, Wu TJ, Johnson RB, Roane C, Young D, Carlo WA. Outcome of extremely preterm infants randomized at birth to different PaCO2 targets during the first seven days of life. Biol Neonate 90: 218 –225, 2006. 197. Tokumaru AM, Barkovich AJ, O’Uchi T, Matsuo T, Kusano S. The evolution of cerebral blood flow in the developing brain: evaluation with iodine-123 iodoamphetamine SPECT and correlation with MR imaging. Am J Neuroradiol 20: 845–852, 1999. 198. Tolcos M, Harding R, Loeliger M, Breen S, Cock M, Duncan J, Rees S. The fetal brainstem is relatively spared from injury following intrauterine hypoxemia. Brain Res Dev Brain Res 143: 73–81, 2003. 199. Tomimatsu T, Lee SJ, Pena JP, Ross JM, Lang JA, Longo LD. Maternal caffeine administration and cerebral oxygenation in near-term fetal sheep. Reprod Sci 14: 588 –594, 2007. 200. Toth P, Rozsa B, Springo Z, Doczi T, Koller A. Isolated human and rat cerebral arteries constrict to increases in flow: role of 20-HETE and TP receptors. J Cereb Blood Flow Metab 31: 2096 –2105, 2011. 201. Tsuji M, Saul JP, du Plessis A, Eichenwald E, Sobh J, Crocker R, Volpe JJ. Cerebral intravascular oxygenation correlates with mean arterial pressure in critically ill premature infants. Pediatrics 106: 625–632, 2000. 202. Vaucher YE, Peralta-Carcelen M, Finer NN, Carlo WA, Gantz MG, Walsh MC, Laptook AR, Yoder BA, Faix RG, Das A, Schibler K, Rich W, Newman NS, Vohr BR, Yolton K, Heyne RJ, Wilson-Costello DE, Evans PW, Goldstein RF, Acarregui MJ, Adams-Chapman I, Pappas A, Hintz SR, Poindexter B, Dusick AM, McGowan EC, Ehrenkranz RA, Bodnar A, Bauer CR, Fuller J, O’Shea TM, Myers GJ, Higgins RD. Neurodevelopmental outcomes in the early CPAP and pulse oximetry trial. N Engl J Med 367: 2495–2504, 2012. 203. Victor S, Marson AG, Appleton RE, Beirne M, Weindling AM. Relationship between blood pressure, cerebral electrical activity, cerebral fractional oxygen extraction, and peripheral blood flow in very low birth weight newborn infants. Pediatr Res 59: 314 –319, 2006. 204. Vinukonda G, Dummula K, Malik S, Hu F, Thompson CI, Csiszar A, Ungvari Z, Ballabh P. Effect of prenatal glucocorticoids on cerebral vasculature of the developing brain. Stroke 41: 1766 –1773, 2010. 205. Virgintino D, Errede M, Robertson D, Girolamo F, Masciandaro A, Bertossi M. VEGF expression is developmentally regulated during human brain angiogenesis. Histochem Cell Biol 119: 227–232, 2003. 206. Volpe JJ. Hypoxic-ischaemic encephalopathy: biochemical and physiological aspects. In: Neurology of the Newborn, edited by Volpe JJ. Philadelphia, PA: Saunders, 2008, p. 245–480. 207. Wang CL, Anderson C, Leone TA, Rich W, Govindaswami B, Finer NN. Resuscitation of preterm neonates by using room air or 100% oxygen. Pediatrics 121: 1083–1089, 2008. 208. Wang Y, Kilic E, Kilic U, Weber B, Bassetti CL, Marti HH, Hermann DM. VEGF overexpression induces post-ischaemic neuroprotection, but facilitates haemodynamic steal phenomena. Brain 128: 52–63, 2005. 209. Wardle SP, Yoxall CW, Weindling AM. Determinants of cerebral fractional oxygen extraction using near infrared spectroscopy in preterm neonates. J Cereb Blood Flow Metab 20: 272–279, 2000. 210. Wei EP, Randad RS, Levasseur JE, Abraham DJ, Kontos HA. Effect of local change in O2 saturation of hemoglobin on cerebral vasodilation from hypoxia and hypotension. Am J Physiol Heart Circ Physiol 265: H1439 –H1443, 1993. 211. Wei L, Keogh CL, Whitaker VR, Theus MH, Yu SP. Angiogenesis and stem cell transplantation as potential treatments of cerebral ischemic stroke. Pathophysiology 12: 47–62, 2005. 212. West CR, Groves AM, Williams CE, Harding JE, Skinner JR, Kuschel CA, Battin MR. Early low cardiac output is associated with compromised electroencephalographic activity in very preterm infants. Pediatr Res 59: 610 –615, 2006. 213. Whiteus C, Freitas C, Grutzendler J. Perturbed neural activity disrupts cerebral angiogenesis during a postnatal critical period. Nature 505: 407–411, 2013. 214. Willie CK, Macleod DB, Shaw AD, Smith KJ, Tzeng YC, Eves ND, Ikeda K, Graham J, Lewis NC, Day TA, Ainslie PN. Regional brain blood flow in man during acute changes in arterial blood gases. J Physiol 590: 3261–3275, 2012. 215. Willis AP, Leffler CW. NO and prostanoids: age dependence of hypercapniaand histamine-induced dilations of pig pial arterioles. Am J Physiol Heart Circ Physiol 277: H299 –H307, 1999. 216. Winn HR, Welsh JE, Rubio R, Berne RM. Brain adenosine production in rat during sustained alteration in systemic blood pressure. Am J Physiol Heart Circ Physiol 239: H636 –H641, 1980.

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Review R786

CEREBRAL VASOREGULATION AND BRAIN INJURY IN PRETERM INFANTS

217. Wintermark P, Tolsa JF, Van Melle G, Forcada-Guex M, Moessinger AC. Long-term outcome of preterm infants treated with nasal continuous positive airway pressure. Eur J Pediatr 166: 473–483, 2007. 218. Wolke D, Samara M, Bracewell M, Marlow N. Specific language difficulties and school achievement in children born at 25 weeks of gestation or less. J Pediatr 152: 256 –262, 2008. 219. Wong FY, Barfield CP, Horne RS, Walker AM. Dopamine therapy promotes cerebral flow-metabolism coupling in preterm infants. Intensive Care Med 35: 1777–1782, 2009. 220. Wong FY, Leung TS, Austin T, Wilkinson M, Meek JH, Wyatt JS, Walker AM. Impaired autoregulation in preterm infants identified by using spatially resolved spectroscopy. Pediatrics 121: e604 –e611, 2008. 221. Wong FY, Silas R, Hew S, Samarasinghe T, Walker AM. Cerebral oxygenation is highly sensitive to blood pressure variability in sick preterm infants. PloS One 7: e43165, 2012.

222. Wong FY, Witcombe NB, Yiallourou SR, Yorkston S, Dymowski AR, Krishnan L, Walker AM, Horne RS. Cerebral oxygenation is depressed during sleep in healthy term infants when they sleep prone. Pediatrics 127: e558 –e565, 2011. 223. 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, 2006. 224. Yoxall CW, Weindling AM. Measurement of cerebral oxygen consumption in the human neonate using near infrared spectroscopy: cerebral oxygen consumption increases with advancing gestational age. Pediatr Res 44: 283–290, 1998. 225. Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann KA, Pozzan T, Carmignoto G. Neuron-to-astrocyte signaling is central to the dynamic control of brain microcirculation. Nat Neurosci 6: 43–50, 2003.

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00487.2013 • www.ajpregu.org

Copyright of American Journal of Physiology: Regulatory, Integrative & Comparative Physiology is the property of American Physiological Society and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

Cerebral vascular regulation and brain injury in preterm infants.

Cerebrovascular lesions, mainly germinal matrix hemorrhage and ischemic injury to the periventricular white matter, are major causes of adverse neurod...
674KB Sizes 2 Downloads 3 Views