Comprehensive Physiology Wiley Online Library

Regulation of the Cerebral Circulation During Development

Full Article on Wiley Online Library



Abstract

The cerebral microcirculation undergoes dynamic changes in parallel with the development of neurons, glia, and their energy metabolism throughout gestation and postnatally. Cerebral blood flow (CBF), oxygen consumption, and glucose consumption are as low as 20% of adult levels in humans born prematurely but eventually exceed adult levels at ages 3 to 11 years, which coincide with the period of continued brain growth, synapse formation, synapse pruning, and myelination. Neurovascular coupling to sensory activation is present but attenuated at birth. By 2 postnatal months, the increase in CBF often is disproportionately smaller than the increase in oxygen consumption, in contrast to the relative hyperemia seen in adults. Vascular smooth muscle myogenic tone increases in parallel with developmental increases in arterial pressure. CBF autoregulatory response to increased arterial pressure is intact at birth but has a more limited range with arterial hypotension. Hypoxia‐induced vasodilation in preterm fetal sheep with low oxygen consumption does not sustain cerebral oxygen transport, but the response becomes better developed for sustaining oxygen transport by term. Nitric oxide tonically inhibits vasomotor tone, and glutamate receptor activation can evoke its release in lambs and piglets. In piglets, astrocyte‐derived carbon monoxide plays a central role in vasodilation evoked by glutamate, ADP, and seizures, and prostanoids play a large role in endothelial‐dependent and hypercapnic vasodilation. Overall, homeostatic mechanisms of CBF regulation in response to arterial pressure, neuronal activity, carbon dioxide, and oxygenation are present at birth but continue to develop postnatally as neurovascular signaling pathways are dynamically altered and integrated. © 2021 American Physiological Society. Compr Physiol 11:2371‐2432, 2021.

Figure 1. Figure 1. Global cerebral blood flow (CBF) is higher at 3 to 11 years of age than in adults. Arterial O2 content, CBF, cerebral O2 transport, cerebral metabolic rate of O2 (CMRO2), and O2 extraction fraction in first quantitative cerebral hemodynamic study in children with the nitrous oxide wash‐in technique. Data from nine children were pooled from ages 3 to 11 years. Children at this age have higher CBF, O2 transport, and CMRO2, with comparable O2 extraction fraction as 12 adults undergoing the same technique. Source: Adapted, with permission, from Kennedy C and Sokoloff L, 1957 221.
Figure 2. Figure 2. Cerebral blood flow (CBF) throughout childhood varies by brain region. Regional CBF was measured by SPECT imaging in 42 children ranging in age from 2 days to 19 years. Data are divided into age bins with 5 to 10 children per bin, and the mean and SD are plotted against the average age for each bin. Blood flow in cortical regions increases in infants and reaches adult levels by 1 year and exceeds adult levels from 3 to 12.5 years. CBF in frontal association cortex lags other cortical regions. In thalamus, CBF is already above adult levels in infants. Source: Adapted, with permission, from Chiron C, et al., 1992 96.
Figure 3. Figure 3. Regional cerebral blood flow (CBF) (A) during development (relative to adult levels) paralleled regional developmental increases in cerebral metabolic rate of O2 (CMRO2) (B) with little change in O2 extraction fraction (C). Measurements were made with PET imaging in children with neurosurgical disorders but with normal psychomotor development. Compared with CBF and CMRO2 values in adults, low values were observed in the frontal association (FA), visual association (VA), and the sensorimotor (SM) areas in the less than 1‐year‐old group (n = 10) and in the FA area in the 1‐ to less than 3‐year‐old group (n = 5). In the 3‐ to less than 8‐year‐old group (n = 7) and in the greater than 8‐year‐old group (9‐17‐years old; n = 8), CBF and CMRO2 exceeded the corresponding adult levels in all regions (*, P < 0.05 by Mann‐Whitney U‐test), except for the FA area. Consistent with delayed maturation in cortical association cortex, CBF and CMRO2 values were lower in FA than in primary SM area, and CBF and CMRO2 in VA cortex were lower than in the primary visual (VC) area (a, P < 0.05 by paired t‐test), except in the ≥8‐year‐old group. P, parietal lobe; T, temporal lobe; Le, lenticulum nuclei; Th, thalamus; Ce, cerebellar hemisphere; BS, brain stem. Source: Adapted, with permission, from Takahashi T, et al., 1999 408, © 1999, American Society of Neuroradiology.
Figure 4. Figure 4. (A) After reaching its peak level in preadolescents, cerebral blood flow (CBF) declines in male adolescents toward levels seen in young male adults. In females, CBF also declines in early adolescence but then increases in specific regions in late adolescence. These regions are denoted by arrows on the MR images; L Inf Parietal, left inferior parietal cortex; R DLPFC, right dorsolateral prefrontal cortex; R insula, right insula cortex; vMPFC, ventromedial prefrontal cortex; R Lat Temporal, right lateral temporal cortex. Source: Reused, with permission, from Satterthwaite TD, et al., 2014 388. © 2014, National Academy of Sciences. Figure 2 from https://www.pnas.org/content/111/23/8643. (B) Structural changes in cerebral cortex during adolescence. Gray matter density (GMD), determined from T1 MRI intensity averaged over the entire brain, increases in adolescence, while gray matter volume (GMV), gray matter mass (GMM = GMD × GMV), and cortical thickness (CT) largely decrease in males and females. Note that the higher CBF per unit mass in females at the end of adolescence is associated with higher GMD and is distributed to a lower gray matter volume and mass in females compared to males, thereby attenuating sex differences in volumetric blood flow to the entire cortical volume. Values are plotted as percentages of the fitted value for males at 8 years of age. Shaded bands correspond to two SE of the fit. Source: Adapted, with permission, from Gennatas ED et al., 156.
Figure 5. Figure 5. Mean arterial pressure (MAP), cerebral blood flow (CBF), and cerebrovascular resistance (CVR) during human development. Source: Adapted, with permission, from Chiron C, et al., 1992 96; De Vis JB, et al., 2014 114, 2013 115; Liu P, et al., 2014 290; Ouyang M, et al., 2017 343.
Figure 6. Figure 6. (A) Global cerebral blood flow (CBF), derived from summation of phase‐contrast MRI of internal carotid and vertebral arteries of 14 unsedated neonates studied about 2 to 3 weeks after birth, increased linearly with gestational age determined by postmenstrual weeks (PMW) at a rate of 1.22 mL/min/100 g/ PMW. Source: Adapted, with permission, from Ouyang M, et al., 2017, 343. (B) Global cerebral metabolic rate of O2 (CMRO2), derived from MRI measurement of sagittal sinus O2 saturation and CBF in 10 unsedated neonates, increased linearly with gestational age. Source: Adapted, with permission, from Liu P, et al., 2014 290, © 2014, John Wiley & Sons.
Figure 7. Figure 7. Postnatal increases in cerebral blood flow are coupled to postnatal increases in cerebral metabolic rate of O2 (CMRO2) as determined by advance near‐infrared spectroscopy (NIRS) modalities in prematurely born neonates over the first 6 weeks after birth. Noninvasive NIRS‐based optical measures of tissue hemoglobin concentration (A), tissue O2 saturation (B), cerebral blood volume (C), a cerebral blood flow index (D), and relative CMRO2 determined with frequency domain NIRS plus diffuse correlation spectroscopy are expressed relative to the group average in human premature neonates born at 25 to 33 weeks PMA brains. The decreases in tissue hemoglobin concentration and tissue oxyhemoglobin saturation without an accompanying decrease in cerebral blood volume are attributed to a postnatal decrease in systemic hematocrit as high‐O2 affinity fetal hemoglobin‐containing red blood cells are cleared faster than low‐O2 affinity adult hemoglobin‐containing red blood cells are produced. Lines are linear regressions that were all statistically significant versus age, except for cerebral blood volume. Source: Reused, with permission, from Roche‐Labarbe N, et al., 2010 379.
Figure 8. Figure 8. Cerebral metabolic rate of glucose (CMRGluc) of various brain regions increases during early human development and then overshoots adult levels especially in forebrain regions. Source: Adapted, with permission, from Chugani HT, et al., 1987 100.
Figure 9. Figure 9. Glucose utilization of the human brain by age in males (A) and females (B) and as a % of total body resting metabolic rate (rmr, solid line) and daily energy requirements (der, dashed line), expressed in glucose equivalents, for males (C) and females (D). Glucose utilization of the entire brain was estimated in units of g/day from the PET‐derived glucose consumption for each brain region and the MRI‐derived volume of each brain region at different stages of development. Through 5 years of age, the increase in total brain glucose utilization outpaces the increase in energy requirements for the entire body, whether based on resting caloric demand or including caloric needs for daily activity. The brain glucose utilization, expressed as a percent of rmr (red dots), is out of phase with the body‐weight growth rate (dw/dt, blue dots) in males (E) and females (F), suggesting an evolutionary adaptation to delay body growth so that limited caloric intake can be diverted to the prolonged period brain growth and development in humans. Glucose utilization as a percent of rmr and weight velocities are plotted as SD scores to allow unitless comparison. Source: Adapted, with permission, from Kuzawa CW, et al., 2014 241, © 2014, National Academy of Sciences. Figures 1A‐D and 2A‐B from https://www.pnas.org/content/111/36/13010.
Figure 10. Figure 10. Neurovascular coupling with visual stimulation. (A) Functional MRI BOLD response to visual stimulation (shaded area) in four children at age 4 to 71 months is directionally opposite of the response in adults, with a negative BOLD response occurring in children. Perfusion MRI (not shown) indicated no increase in blood flow in infants, in contrast to a 20% increase in perfusion in adults. Source: Adapted, with permission, from Born AP, et al., 2002 64, © 2002, Elsevier. (B) In response to visual stimulation, the area of significantly activated voxels in visual cortex displays a sharp transition from positive BOLD in neonates to negative BOLD responses beyond 8 weeks of age. Source: Adapted, with permission, from Yamada H, et al., 2000 468, © 2000, Wolters Kluwer.
Figure 11. Figure 11. Neurovascular coupling with tactile stimulation. (A) Functional MRI BOLD responses to 1‐s somatosensory stimulation of the hand in adults (n = 10), infants at term (n = 15), and preterm infants at 34 weeks (n = 10). Positive BOLD responses are smaller and delayed in infants, and the post‐stimulus undershoot is smaller in preterms. Source: Adapted, with permission, from Arichi T, et al., 2012 14. Licensed Under CCBY 3.0 Unported. (B) In six preterm neonates born at 33 to 34 weeks PMA and studied 2.5 weeks after birth, NIRS‐based measurements during 5 s of tactile stimulation show decreased O2 extraction fraction (OEF) and increased CBF, CMRO2, and cerebral blood volume (CBV), thereby indicating the presence of functional coupling in premature neonates. Source: Adapted, with permission, from Roche‐Labarbe N, et al. 2014 381, © 2014, Elsevier.
Figure 12. Figure 12. Local cerebral blood flow, measured by laser‐speckle imaging, does not increase in response to hindlimb stimulation in the neonatal mouse brain. (A‐C) Averaged time courses of the change in oxyhemoglobin (Δ[HbO]), deoxyhemoglobin (Δ[HbR]), tissue hemoglobin (Δ[HbT]), and the percent change in laser‐speckle flow (%Δ speckle) in the contralateral hindpaw region of the somatosensory cortex of mice in P7 to P8 (A), P10 to P13 (B), and adult (C) age groups (n = 5, 4, 3, respectively). Source: Adapted, with permission, from Kozberg MG, et al., 2016 236.
Figure 13. Figure 13. Schematic diagram of the major signaling pathways involved in neurovascular coupling. With the initiation of neuronal action, the sequence of events proceeds from left to right, resulting in ascending vasodilation, part of which depends on conduction through endothelial gap junctions. The first step occurs with the increase in CMRO2 leading to a transient increase in deoxyhemoglobin that results in an increase in red blood cell (RBC) deformability and velocity in capillaries. The second step involves release of the coneurotransmitter ATP acting on astrocyte P2X1 receptors to stimulate mobilization of arachidonic acid (AA) via phospholipase D2 (PLD2) and diacylglycerol lipase (DGL). PGE2 is synthetized by cyclooxygenase‐1 (COX‐1) and released from the astrocyte foot process to induce relaxation of pericytes via EP4 receptors. Relaxation of circumferential pericytes increases capillary diameter in the vicinity of terminal arterioles, thereby increasing RBC flux. In addition to ATP, some interneurons in close proximity to astrocyte foot processes release GABA and VIP that either directly or indirectly act to increase Ca2+ in the foot processes adjacent to pericytes and smooth muscle and that act to facilitate rapid vasorelaxation signaling. The third step is the action of neurotransmitters on astrocytes adjacent to vascular smooth muscle of intraparenchymal arterioles. In addition to the effects of ATP on astrocyte P2X1 receptors, glutamate can act on metabotropic glutamate receptor 5 (mGluR5) that is thought to be present on astrocytes of immature brain. Extracellular ATP can be metabolized by ectonucleotidases to adenosine, which in sufficiently high concentration can activate the low‐affinity adenosine A2B receptor. Together, these pathways may facilitate Ca2+ wave transmission throughout the astrocyte network leading to mobilization of AA, which can act as a substrate for cytochrome P450 (CYP) epoxygenase to produce epoxyeicosatrienoic acids (EETs). EETs can activate BKCa channels on the astrocyte foot process and release sufficient K+ into the tight extracellular space between the foot process and smooth muscle to activate Kir channels on smooth muscle, resulting in hyperpolarization and relaxation. Furthermore, ATP can be metabolized by a different ectonucleotidase on astrocytes to ADP, which can lead to activation of heme oxygenase‐2 (HO‐2) expressed in astrocytes of immature brain. HO‐2 generates CO, which in swine can facilitate the opening of BKCa channels on smooth muscle and possibly on astrocytes. The intermediate conductance KCa channel 3.1 and the TRPV4 channel on astrocytes also play a less well‐defined role in the coupling process. AA can be released as a coneurotransmitter and serve as an agonist on TRPV4 channels that signal the release of ATP into the extracellular space where they can act on adjacent astrocyte processes to produce a Ca2+ response. This slow spread of Ca2+ waves in surrounding astrocytes presumably augments vasorelaxation signaling converging on smooth muscle. In addition to astrocyte‐mediated vasodilation, neurons can exert direct effects on smooth muscle, including release of PGE2 generated by COX‐2 in select neurons and release of NO by neuronal NO synthase (NOS) mediated by NMDA receptors. The NO can stimulate guanylyl cyclase and increase vasorelaxant cGMP in smooth muscle. NO can also inhibit CYP ω‐hydroxylase activity and the generation of 20‐HETE that normally inhibits BKCa channels. Some GABAergic interneurons have processes in close proximity to arterioles and capillaries and are thought to induce relaxation via an NO‐dependent mechanism. The fourth step is dilation of pial arterioles to maintain the perfusion pressure for the penetrating arterioles serving the activated and nonactivated regions. This dilation involves some of the same mechanisms mediating penetrating arteriolar dilation, including NO generated by neuronal NOS and CO generated by HO‐2 in astrocytes in the glia limitans. One difference is that the astrocyte‐derived adenosine acts directly on pial arteriole smooth muscle A2A receptors to produce dilation.
Figure 14. Figure 14. Developmental increases in nitric oxide synthase (NOS) activity in sheep. (A) NOS catalytic activity, measured in fresh cortical samples, increased fourfold between 0.48 and 0.92 gestation in fetal sheep. CBF (B) and CMRO2 (C) increase threefold between 0.63 and 0.92 gestation, and inhibition of NOS activity with nitro‐L‐arginine methyl ester (L‐NAME) decreases CBF in unanesthetized fetal sheep at both gestational ages without reducing CMRO2. These results imply that NO is tonically generated in quantities sufficient to induce vasodilation by midgestation in fetal sheep. Source: Modified, with permission, from Northington FJ, et al., 1997 335.
Figure 15. Figure 15. Angiogenesis across cerebral cortical layers. In cerebral cortex of PND1 rabbits, capillary density and CBF are lower in deeper layers and increase during postnatal development. Cytochrome oxidase activity also increases in all cortical layers during development, but in relative terms, its spatial gradient is less than the spatial gradient of capillary density. This comparison suggests that increases in capillary density lag the increases in the mitochondrial capacity for energy metabolism. Source: Adapted, with permission, from Tuor UI, et al., 1994 417.
Figure 16. Figure 16. Pressure‐diameter relationship of human infant middle cerebral artery at 37 week gestation. The artery was subjected to changes in transmural pressure in random steps between 30 and 80 mmHg in physiologic salt solution (PSS). With increased transmural pressure, arterial diameter decreased (open circles), indicative of increased myogenic tone. In the presence of PSS containing 127 mmol/liter of KCl to induce depolarization and a maximum contractile state (closed circles), diameter increased slightly with increasing transmural pressure. In the presence of Ca2+‐free PSS containing 2 mmol/liter EGTA (triangles), the diameter increased passively at each pressure value. The difference between the passive and active curves indicates that the human middle cerebral artery is capable of generating a considerable myogenic response at this developmental age. Source: Adapted, with permission, from Bevan RD, et al., 1998 58, © 1998, Springer Nature.
Figure 17. Figure 17. Relationship of cerebral blood flow to cerebral perfusion pressure at different stages of human development. These idealized autoregulation curves are based on the limited number of studies of autoregulation in early human development and of studies of autoregulation in immature animals with extrapolation of the stage of brain maturation of the particular species to human development.
Figure 18. Figure 18. (A) The distribution of the cerebral oximetry index (COx) across binned mean arterial blood pressure (ABP) in children undergoing cardiopulmonary bypass (CPB). Assuming a constant CMRO2 during the sampling period, a low COx indicates a low correlation of tissue HbO2 with variations in ABP (good CBF autoregulation), and a high COx indicates a high correlation of tissue HbO2 with variations in ABP (poor CBF autoregulation). A COx value of 0.4 (horizontal dashed line) is assumed to correspond to the lower limit of autoregulation (LLA) threshold. (B) The distribution of percent of total monitoring time at each ABP before, during, and after CPB. (A, B) Source: Adapted, with permission, from Brady K, et al., 2010 66. (C) The percentage of time spent below an ABP of 40 mmHg was greater during CPB than before or after CPB. (D) The average individual LLA with COx less than 0.4 for 42 pediatric patients was 42 ± 7 mmHg compared with 55 ± 14 mmHg for 37 adults patients undergoing cardiopulmonary bypass (±SD; P < 0.0001 between age groups).
Figure 19. Figure 19. Phase‐angle relationship of the response of ICP to quasi‐sinusoidal variations in arterial blood pressure (ABP) induced by frequency modulation of positive end‐expiratory pressure in two‐week‐old piglets. When ABP was above the lower limit of autoregulation (LLA, open circles), the phase angle decreased when the ABP frequency increased from 1 to 6 cycles per minute (CPM). Assuming that ICP is related to cerebral blood volume, which is related to cerebral vasodilation and vasoconstriction responses, dynamic autoregulation is ineffective at periodicities less than 10 s. When ABP is below the LLA (closed circles), the phase angle is already reduced at low frequencies, confirming that frequency analysis at low ABP frequencies can be used to assess autoregulation. Source: Adapted, with permission, from Fraser CD, 2013 144, © 2013, The American Physiological Society.
Figure 20. Figure 20. Noradrenergic constriction is enhanced in human preterm cerebral arteries. (A) Catecholamine histofluorescence of perivascular nerves is prominent in proximal middle cerebral artery at gestational age 38 weeks (a) and term (b) and in basilar artery at gestational age 32 weeks (c), 5 weeks (d), and 38 weeks (e). (B) Catecholamine histofluorescence in perivascular nerves is also prominent at gestational age of 24 weeks in distal middle cerebral artery (a) and a branch of basilar artery (b). (C) The response of middle cerebral artery from infant of 24 weeks of gestation to norepinephrine before (left) and after (right) endothelium inactivation, which attenuated the relaxation to acetylcholine while preserving norepinephrine‐induced contraction. Source: Adapted, with permission, from Bevan R, et al. 1998 56, © 1998, Springer Nature. Comparison of the dose to produce constriction in 50% of cerebral arteries (ED50) (D) and normalized tension generated in response to norepinephrine (E) and electrical field stimulation (F) of cerebral arteries studied within 2 days of autopsy in eight neonates at 23 to 38 weeks PMA and 21 adults (mean ± SE). Comparison of the mean number of catecholaminergic nerve fibers per millimeter length of cerebral arteries in 2 neonates and 15 adults (G). Source: Adapted, with permission, from Bevan R, et al., 1998 56.
Figure 21. Figure 21. Developmental changes in norepinephrine‐induced constriction of pial arteries. (A) When applied through a closed cranial window, norepinephrine produced dose‐dependent constriction in anesthetized fetal sheep and newborn lambs but not in adult sheep. The effect was most pronounced in fetuses less than 121 days gestation (<0.8 full term). (B) Electrical stimulation of the superior cervical sympathetic ganglion produced constriction of pial arteries that was blocked by the α‐adrenergic antagonist prazosin. Source: Adapted, with permission, from Wagerle LC, et al., 1990 442, © 1990, The American Physiological Society.
Figure 22. Figure 22. Developmental changes in blood‐brain barrier permeability in sheep. The influx transfer constant (Ki) for radiolabeled aminoisobutyric acid shows a progressive decrease from 60% of gestation in fetal sheep through adulthood in various brain regions. However, even at 60% of gestation, the transfer constant is not considered high and indicates that significant diffusion restriction is present for this metabolically inert small molecule. Source: Modified, with permission, from Stonestreet BS, et al., 1996 402, © 1996, The American Physiological Society.
Figure 23. Figure 23. Developmental changes in tight junction protein organization. Immunostaining of occludin and claudin‐5 shows endothelial cytoplasmic staining with some linear cell membrane assembly of occludin present at 12 weeks PMA in human fetal telencephalon (A, B). By 14 weeks, linear beaded staining of occludin and claudin‐5 becomes evident in the cell membrane (C, D). At 18 weeks, a more mature appearance of nearly linear immunostaining of occludin and claudin‐5 occurs. Source: Adapted, with permission, from Virgintino et al. 434.
Figure 24. Figure 24. Endothelial vasodilator function is depressed in fetal sheep carotid arteries. Postnatal maturation is associated with a moderately increased abundance of endothelial nitric oxide synthase (eNOS). However, when normalized by the luminal surface area or by eNOS concentration, the amount of NO generated in response to the calcium ionophore A23187 or to endothelial shear stress is greater in adult than in fetal arteries. In contrast, NO generation by ADP is not significantly different. Thus, eNOS coupling mechanisms are not fully mature in fetal sheep arteries, and maturation may depend on the nature of the stimulus. Mean ± SE, n = 6‐8. Source: Adapted, with permission, from White CR, et al., 2005 451, © 2005, The American Physiological Society.
Figure 25. Figure 25. Role of CO in immature neurovascular unit in glia limitans. Neurotransmitter glutamate binds to astrocyte ionotropic and metabotropic glutamate (GLU) receptors (iGLUR and mGLUR) expressed on immature astrocytes, elevating [Ca2+]i that increases heme oxygenase‐2 (HO‐2 activity) and generates CO. The CO diffuses to pial artery vascular smooth muscle where it binds to a heme element that increases Ca2+ spark‐to‐BKCa channel coupling, thereby elevating BKCa channel activity. In addition, CO increases Ca2+ spark frequency, further increasing BKCa channel activity. The resulting smooth muscle cell hyperpolarization produces vasodilation. Furthermore, endothelial‐derived NO and PGI2 provide a minimal amount of guanylyl cyclase (GC) and cGMP and/or activation of PGI2 receptors (IP), adenylyl cyclase (AC), and cAMP. The resulting protein kinase G (PKG) activity may phosphorylate sarcoplasmic reticulum RyRs, increasing Ca2+ spark frequency and/or the BKCa channel. In the intact vasculature, NO and PGI2 can activate smooth muscle cell GC and AC directly, if a stimulus is sufficient at the endothelial cell. In addition to glutamate, neurons can release ATP from neurotransmitter vesicles, the ATP can be degraded to ADP by an astrocyte endonucleotidase, and the ADP can act on astrocyte P2Y1 receptors to also activate HO‐2 (not shown). IP3, inositol 1,4,5‐trisphosphate. Source: Adapted, with permission, from Leffler CW, et al., 2011 276, © 2011, The American Physiological Society.
Figure 26. Figure 26. Schematic diagram of endothelial‐derived factors in dilation of pial arterioles to hypercapnia. Acidosis is thought to activate cystathionine γ‐lyase (CSE) and generate H2S, which activates KATP channels and induces hyperpolarization and consequent inhibition of VDCC, voltage‐dependent Ca2+ channels (VDCC). H2S also promotes Ca2+ sparks and activation of BKCa channels (not shown), which also play a role in hypercapnic dilation. In addition, acidosis is thought to stimulate arachidonic acid (AA) release and cyclooxygenase (COX) activity and generate PGI2, which then acts on the smooth muscle prostacyclin receptor (IP) to stimulate adenylyl cyclase (AC). Source: Adapted, with permission, from Leffler CW, et al., 2011 274, © 2011, The American Physiological Society.
Figure 27. Figure 27. Developmental changes in the cerebrovascular response to hypoxia in unanesthetized sheep. (A) Over a wide range of arterial O2 content, cerebral blood flow (CBF) increases from 0.6 gestation to 0.9 gestation in fetal sheep, remains elevated after birth, and then decreases in adults. (B) At 0.6 gestation, the increase in CBF is inadequate to maintain cerebral O2 transport, whereas the O2 transport is maintained later in development during hypoxia. (C) O2 extraction fraction, which is the ratio of CMRO2 to O2 transport, is also maintained during hypoxia later in development, although the level is highest in adults and lowest at 0.6 gestation. The levels of O2 extraction fraction differed among groups despite normalization with the different levels of CMRO2 (indicated as μmol O2/min/100 g). (D‐F) Fetal sheep at 0.9 gestation and neonatal lambs underwent a partial exchange transfusion with adult sheep blood having a P50 (PO2 at 50% oxyhemoglobin saturation) of 44 Torr to increase the P50 from 17 to 32 Torr in fetal sheep and from 26 to 37 Torr in neonatal lambs. Compared to the pretransfusion responses to hypoxia, the CBF and O2 transport responses were reduced, and the O2 extraction fraction was elevated to levels closer to those seen in adult sheep. Source: Adapted, with permission, from Koehler RC, et al., 1986 228, 1984 230; Gleason CA, et al., 1990 161; Rosenberg AA, et al., 1986 382.
Figure 28. Figure 28. (A) Example of real‐time monitoring of pressure‐reactivity index (PRx) for 72 h in a child after TBI. Top trace shows arterial blood pressure (ABP, black) and CPP (gray), second trace shows ICP, and third trace shows PRx. Bottom chart shows the PRx within 5 mmHg CPP bins averaged over 72 h for this single patient. An optimal CPP of 57.5 mmHg could be identified wherein PRx had a minimal value. (B) The average PRx for 36 children after TBI displays a U‐shaped curve. (C) The optimal CPP (CPPopt) in 34 TBI patients correlated with the patient's age. Source: Adapted, with permission, from Lewis PM, et al., 2015 278, © 2015, Wolters Kluwer.
Figure 29. Figure 29. Schematic diagram of potential mechanisms of impaired cerebrovascular regulation after hypoxia‐ischemia, stroke, and fluid percussion injury in piglets. Endothelin‐1 (ET‐1) and vasopressin have been implicated in the release of the endogenous opioid nociception/orphanin FQ (NOC/oFQ) that can result in the generation of superoxide through a PKC signaling in concert with activation of cyclooxygenase (COX). Acting through protein tyrosine kinase (PKT) and ERK/MAPK signaling, superoxide generation results in impaired K+ channel function. Focal stroke stimulates endogenous tissue plasminogen activator (tPA) that is known to augment activation of NMDA receptors. This pathway can result in increased ET‐1 and activation of ERK/MAPK. It also produces an inflammatory response leading to increased interleukin‐6 and impaired function of K+ channels. Inhaled nitric oxide can block the increase in ET‐1 and preserve autoregulation. Source: Adapted, with permission, from Pastor P, et al., 2019 361; Armstead, WM, 2005 34.
Figure 30. Figure 30. Synthesis of the relative time course of developmental regulation of the cerebral circulation and energy metabolism. Blood‐brain barrier (BBB) function, CMRO2, CMRGluc, cerebral O2 transport, CBF reactivity to CO2, and neurovascular coupling are normalized relative to an adult value of 1. Autoregulation is displayed in terms of the upper (ULA) and lower (LLA) limits of autoregulation (mmHg) below and above which CBF is pressure passive. All lines are based on human data or, in the cases where human data are not available, on animal data. Interpolation was used in cases where no data are available for a particular developmental stage. Note that energy metabolism and O2 transport overshoot adult values between the infant and adolescent stages, whereas CO2 reactivity does not. Also, O2 transport has a postnatal dip, attributable to transient anemia and the decrease in hemoglobin O2 affinity as fetal hemoglobin is replaced with adult hemoglobin. Neurovascular coupling is thought to have a transient increase near term and then decrease during the period of elevated CMRGluc. The range of autoregulation between the ULA and LLA progressively expands during development.


Figure 1. Global cerebral blood flow (CBF) is higher at 3 to 11 years of age than in adults. Arterial O2 content, CBF, cerebral O2 transport, cerebral metabolic rate of O2 (CMRO2), and O2 extraction fraction in first quantitative cerebral hemodynamic study in children with the nitrous oxide wash‐in technique. Data from nine children were pooled from ages 3 to 11 years. Children at this age have higher CBF, O2 transport, and CMRO2, with comparable O2 extraction fraction as 12 adults undergoing the same technique. Source: Adapted, with permission, from Kennedy C and Sokoloff L, 1957 221.


Figure 2. Cerebral blood flow (CBF) throughout childhood varies by brain region. Regional CBF was measured by SPECT imaging in 42 children ranging in age from 2 days to 19 years. Data are divided into age bins with 5 to 10 children per bin, and the mean and SD are plotted against the average age for each bin. Blood flow in cortical regions increases in infants and reaches adult levels by 1 year and exceeds adult levels from 3 to 12.5 years. CBF in frontal association cortex lags other cortical regions. In thalamus, CBF is already above adult levels in infants. Source: Adapted, with permission, from Chiron C, et al., 1992 96.


Figure 3. Regional cerebral blood flow (CBF) (A) during development (relative to adult levels) paralleled regional developmental increases in cerebral metabolic rate of O2 (CMRO2) (B) with little change in O2 extraction fraction (C). Measurements were made with PET imaging in children with neurosurgical disorders but with normal psychomotor development. Compared with CBF and CMRO2 values in adults, low values were observed in the frontal association (FA), visual association (VA), and the sensorimotor (SM) areas in the less than 1‐year‐old group (n = 10) and in the FA area in the 1‐ to less than 3‐year‐old group (n = 5). In the 3‐ to less than 8‐year‐old group (n = 7) and in the greater than 8‐year‐old group (9‐17‐years old; n = 8), CBF and CMRO2 exceeded the corresponding adult levels in all regions (*, P < 0.05 by Mann‐Whitney U‐test), except for the FA area. Consistent with delayed maturation in cortical association cortex, CBF and CMRO2 values were lower in FA than in primary SM area, and CBF and CMRO2 in VA cortex were lower than in the primary visual (VC) area (a, P < 0.05 by paired t‐test), except in the ≥8‐year‐old group. P, parietal lobe; T, temporal lobe; Le, lenticulum nuclei; Th, thalamus; Ce, cerebellar hemisphere; BS, brain stem. Source: Adapted, with permission, from Takahashi T, et al., 1999 408, © 1999, American Society of Neuroradiology.


Figure 4. (A) After reaching its peak level in preadolescents, cerebral blood flow (CBF) declines in male adolescents toward levels seen in young male adults. In females, CBF also declines in early adolescence but then increases in specific regions in late adolescence. These regions are denoted by arrows on the MR images; L Inf Parietal, left inferior parietal cortex; R DLPFC, right dorsolateral prefrontal cortex; R insula, right insula cortex; vMPFC, ventromedial prefrontal cortex; R Lat Temporal, right lateral temporal cortex. Source: Reused, with permission, from Satterthwaite TD, et al., 2014 388. © 2014, National Academy of Sciences. Figure 2 from https://www.pnas.org/content/111/23/8643. (B) Structural changes in cerebral cortex during adolescence. Gray matter density (GMD), determined from T1 MRI intensity averaged over the entire brain, increases in adolescence, while gray matter volume (GMV), gray matter mass (GMM = GMD × GMV), and cortical thickness (CT) largely decrease in males and females. Note that the higher CBF per unit mass in females at the end of adolescence is associated with higher GMD and is distributed to a lower gray matter volume and mass in females compared to males, thereby attenuating sex differences in volumetric blood flow to the entire cortical volume. Values are plotted as percentages of the fitted value for males at 8 years of age. Shaded bands correspond to two SE of the fit. Source: Adapted, with permission, from Gennatas ED et al., 156.


Figure 5. Mean arterial pressure (MAP), cerebral blood flow (CBF), and cerebrovascular resistance (CVR) during human development. Source: Adapted, with permission, from Chiron C, et al., 1992 96; De Vis JB, et al., 2014 114, 2013 115; Liu P, et al., 2014 290; Ouyang M, et al., 2017 343.


Figure 6. (A) Global cerebral blood flow (CBF), derived from summation of phase‐contrast MRI of internal carotid and vertebral arteries of 14 unsedated neonates studied about 2 to 3 weeks after birth, increased linearly with gestational age determined by postmenstrual weeks (PMW) at a rate of 1.22 mL/min/100 g/ PMW. Source: Adapted, with permission, from Ouyang M, et al., 2017, 343. (B) Global cerebral metabolic rate of O2 (CMRO2), derived from MRI measurement of sagittal sinus O2 saturation and CBF in 10 unsedated neonates, increased linearly with gestational age. Source: Adapted, with permission, from Liu P, et al., 2014 290, © 2014, John Wiley & Sons.


Figure 7. Postnatal increases in cerebral blood flow are coupled to postnatal increases in cerebral metabolic rate of O2 (CMRO2) as determined by advance near‐infrared spectroscopy (NIRS) modalities in prematurely born neonates over the first 6 weeks after birth. Noninvasive NIRS‐based optical measures of tissue hemoglobin concentration (A), tissue O2 saturation (B), cerebral blood volume (C), a cerebral blood flow index (D), and relative CMRO2 determined with frequency domain NIRS plus diffuse correlation spectroscopy are expressed relative to the group average in human premature neonates born at 25 to 33 weeks PMA brains. The decreases in tissue hemoglobin concentration and tissue oxyhemoglobin saturation without an accompanying decrease in cerebral blood volume are attributed to a postnatal decrease in systemic hematocrit as high‐O2 affinity fetal hemoglobin‐containing red blood cells are cleared faster than low‐O2 affinity adult hemoglobin‐containing red blood cells are produced. Lines are linear regressions that were all statistically significant versus age, except for cerebral blood volume. Source: Reused, with permission, from Roche‐Labarbe N, et al., 2010 379.


Figure 8. Cerebral metabolic rate of glucose (CMRGluc) of various brain regions increases during early human development and then overshoots adult levels especially in forebrain regions. Source: Adapted, with permission, from Chugani HT, et al., 1987 100.


Figure 9. Glucose utilization of the human brain by age in males (A) and females (B) and as a % of total body resting metabolic rate (rmr, solid line) and daily energy requirements (der, dashed line), expressed in glucose equivalents, for males (C) and females (D). Glucose utilization of the entire brain was estimated in units of g/day from the PET‐derived glucose consumption for each brain region and the MRI‐derived volume of each brain region at different stages of development. Through 5 years of age, the increase in total brain glucose utilization outpaces the increase in energy requirements for the entire body, whether based on resting caloric demand or including caloric needs for daily activity. The brain glucose utilization, expressed as a percent of rmr (red dots), is out of phase with the body‐weight growth rate (dw/dt, blue dots) in males (E) and females (F), suggesting an evolutionary adaptation to delay body growth so that limited caloric intake can be diverted to the prolonged period brain growth and development in humans. Glucose utilization as a percent of rmr and weight velocities are plotted as SD scores to allow unitless comparison. Source: Adapted, with permission, from Kuzawa CW, et al., 2014 241, © 2014, National Academy of Sciences. Figures 1A‐D and 2A‐B from https://www.pnas.org/content/111/36/13010.


Figure 10. Neurovascular coupling with visual stimulation. (A) Functional MRI BOLD response to visual stimulation (shaded area) in four children at age 4 to 71 months is directionally opposite of the response in adults, with a negative BOLD response occurring in children. Perfusion MRI (not shown) indicated no increase in blood flow in infants, in contrast to a 20% increase in perfusion in adults. Source: Adapted, with permission, from Born AP, et al., 2002 64, © 2002, Elsevier. (B) In response to visual stimulation, the area of significantly activated voxels in visual cortex displays a sharp transition from positive BOLD in neonates to negative BOLD responses beyond 8 weeks of age. Source: Adapted, with permission, from Yamada H, et al., 2000 468, © 2000, Wolters Kluwer.


Figure 11. Neurovascular coupling with tactile stimulation. (A) Functional MRI BOLD responses to 1‐s somatosensory stimulation of the hand in adults (n = 10), infants at term (n = 15), and preterm infants at 34 weeks (n = 10). Positive BOLD responses are smaller and delayed in infants, and the post‐stimulus undershoot is smaller in preterms. Source: Adapted, with permission, from Arichi T, et al., 2012 14. Licensed Under CCBY 3.0 Unported. (B) In six preterm neonates born at 33 to 34 weeks PMA and studied 2.5 weeks after birth, NIRS‐based measurements during 5 s of tactile stimulation show decreased O2 extraction fraction (OEF) and increased CBF, CMRO2, and cerebral blood volume (CBV), thereby indicating the presence of functional coupling in premature neonates. Source: Adapted, with permission, from Roche‐Labarbe N, et al. 2014 381, © 2014, Elsevier.


Figure 12. Local cerebral blood flow, measured by laser‐speckle imaging, does not increase in response to hindlimb stimulation in the neonatal mouse brain. (A‐C) Averaged time courses of the change in oxyhemoglobin (Δ[HbO]), deoxyhemoglobin (Δ[HbR]), tissue hemoglobin (Δ[HbT]), and the percent change in laser‐speckle flow (%Δ speckle) in the contralateral hindpaw region of the somatosensory cortex of mice in P7 to P8 (A), P10 to P13 (B), and adult (C) age groups (n = 5, 4, 3, respectively). Source: Adapted, with permission, from Kozberg MG, et al., 2016 236.


Figure 13. Schematic diagram of the major signaling pathways involved in neurovascular coupling. With the initiation of neuronal action, the sequence of events proceeds from left to right, resulting in ascending vasodilation, part of which depends on conduction through endothelial gap junctions. The first step occurs with the increase in CMRO2 leading to a transient increase in deoxyhemoglobin that results in an increase in red blood cell (RBC) deformability and velocity in capillaries. The second step involves release of the coneurotransmitter ATP acting on astrocyte P2X1 receptors to stimulate mobilization of arachidonic acid (AA) via phospholipase D2 (PLD2) and diacylglycerol lipase (DGL). PGE2 is synthetized by cyclooxygenase‐1 (COX‐1) and released from the astrocyte foot process to induce relaxation of pericytes via EP4 receptors. Relaxation of circumferential pericytes increases capillary diameter in the vicinity of terminal arterioles, thereby increasing RBC flux. In addition to ATP, some interneurons in close proximity to astrocyte foot processes release GABA and VIP that either directly or indirectly act to increase Ca2+ in the foot processes adjacent to pericytes and smooth muscle and that act to facilitate rapid vasorelaxation signaling. The third step is the action of neurotransmitters on astrocytes adjacent to vascular smooth muscle of intraparenchymal arterioles. In addition to the effects of ATP on astrocyte P2X1 receptors, glutamate can act on metabotropic glutamate receptor 5 (mGluR5) that is thought to be present on astrocytes of immature brain. Extracellular ATP can be metabolized by ectonucleotidases to adenosine, which in sufficiently high concentration can activate the low‐affinity adenosine A2B receptor. Together, these pathways may facilitate Ca2+ wave transmission throughout the astrocyte network leading to mobilization of AA, which can act as a substrate for cytochrome P450 (CYP) epoxygenase to produce epoxyeicosatrienoic acids (EETs). EETs can activate BKCa channels on the astrocyte foot process and release sufficient K+ into the tight extracellular space between the foot process and smooth muscle to activate Kir channels on smooth muscle, resulting in hyperpolarization and relaxation. Furthermore, ATP can be metabolized by a different ectonucleotidase on astrocytes to ADP, which can lead to activation of heme oxygenase‐2 (HO‐2) expressed in astrocytes of immature brain. HO‐2 generates CO, which in swine can facilitate the opening of BKCa channels on smooth muscle and possibly on astrocytes. The intermediate conductance KCa channel 3.1 and the TRPV4 channel on astrocytes also play a less well‐defined role in the coupling process. AA can be released as a coneurotransmitter and serve as an agonist on TRPV4 channels that signal the release of ATP into the extracellular space where they can act on adjacent astrocyte processes to produce a Ca2+ response. This slow spread of Ca2+ waves in surrounding astrocytes presumably augments vasorelaxation signaling converging on smooth muscle. In addition to astrocyte‐mediated vasodilation, neurons can exert direct effects on smooth muscle, including release of PGE2 generated by COX‐2 in select neurons and release of NO by neuronal NO synthase (NOS) mediated by NMDA receptors. The NO can stimulate guanylyl cyclase and increase vasorelaxant cGMP in smooth muscle. NO can also inhibit CYP ω‐hydroxylase activity and the generation of 20‐HETE that normally inhibits BKCa channels. Some GABAergic interneurons have processes in close proximity to arterioles and capillaries and are thought to induce relaxation via an NO‐dependent mechanism. The fourth step is dilation of pial arterioles to maintain the perfusion pressure for the penetrating arterioles serving the activated and nonactivated regions. This dilation involves some of the same mechanisms mediating penetrating arteriolar dilation, including NO generated by neuronal NOS and CO generated by HO‐2 in astrocytes in the glia limitans. One difference is that the astrocyte‐derived adenosine acts directly on pial arteriole smooth muscle A2A receptors to produce dilation.


Figure 14. Developmental increases in nitric oxide synthase (NOS) activity in sheep. (A) NOS catalytic activity, measured in fresh cortical samples, increased fourfold between 0.48 and 0.92 gestation in fetal sheep. CBF (B) and CMRO2 (C) increase threefold between 0.63 and 0.92 gestation, and inhibition of NOS activity with nitro‐L‐arginine methyl ester (L‐NAME) decreases CBF in unanesthetized fetal sheep at both gestational ages without reducing CMRO2. These results imply that NO is tonically generated in quantities sufficient to induce vasodilation by midgestation in fetal sheep. Source: Modified, with permission, from Northington FJ, et al., 1997 335.


Figure 15. Angiogenesis across cerebral cortical layers. In cerebral cortex of PND1 rabbits, capillary density and CBF are lower in deeper layers and increase during postnatal development. Cytochrome oxidase activity also increases in all cortical layers during development, but in relative terms, its spatial gradient is less than the spatial gradient of capillary density. This comparison suggests that increases in capillary density lag the increases in the mitochondrial capacity for energy metabolism. Source: Adapted, with permission, from Tuor UI, et al., 1994 417.


Figure 16. Pressure‐diameter relationship of human infant middle cerebral artery at 37 week gestation. The artery was subjected to changes in transmural pressure in random steps between 30 and 80 mmHg in physiologic salt solution (PSS). With increased transmural pressure, arterial diameter decreased (open circles), indicative of increased myogenic tone. In the presence of PSS containing 127 mmol/liter of KCl to induce depolarization and a maximum contractile state (closed circles), diameter increased slightly with increasing transmural pressure. In the presence of Ca2+‐free PSS containing 2 mmol/liter EGTA (triangles), the diameter increased passively at each pressure value. The difference between the passive and active curves indicates that the human middle cerebral artery is capable of generating a considerable myogenic response at this developmental age. Source: Adapted, with permission, from Bevan RD, et al., 1998 58, © 1998, Springer Nature.


Figure 17. Relationship of cerebral blood flow to cerebral perfusion pressure at different stages of human development. These idealized autoregulation curves are based on the limited number of studies of autoregulation in early human development and of studies of autoregulation in immature animals with extrapolation of the stage of brain maturation of the particular species to human development.


Figure 18. (A) The distribution of the cerebral oximetry index (COx) across binned mean arterial blood pressure (ABP) in children undergoing cardiopulmonary bypass (CPB). Assuming a constant CMRO2 during the sampling period, a low COx indicates a low correlation of tissue HbO2 with variations in ABP (good CBF autoregulation), and a high COx indicates a high correlation of tissue HbO2 with variations in ABP (poor CBF autoregulation). A COx value of 0.4 (horizontal dashed line) is assumed to correspond to the lower limit of autoregulation (LLA) threshold. (B) The distribution of percent of total monitoring time at each ABP before, during, and after CPB. (A, B) Source: Adapted, with permission, from Brady K, et al., 2010 66. (C) The percentage of time spent below an ABP of 40 mmHg was greater during CPB than before or after CPB. (D) The average individual LLA with COx less than 0.4 for 42 pediatric patients was 42 ± 7 mmHg compared with 55 ± 14 mmHg for 37 adults patients undergoing cardiopulmonary bypass (±SD; P < 0.0001 between age groups).


Figure 19. Phase‐angle relationship of the response of ICP to quasi‐sinusoidal variations in arterial blood pressure (ABP) induced by frequency modulation of positive end‐expiratory pressure in two‐week‐old piglets. When ABP was above the lower limit of autoregulation (LLA, open circles), the phase angle decreased when the ABP frequency increased from 1 to 6 cycles per minute (CPM). Assuming that ICP is related to cerebral blood volume, which is related to cerebral vasodilation and vasoconstriction responses, dynamic autoregulation is ineffective at periodicities less than 10 s. When ABP is below the LLA (closed circles), the phase angle is already reduced at low frequencies, confirming that frequency analysis at low ABP frequencies can be used to assess autoregulation. Source: Adapted, with permission, from Fraser CD, 2013 144, © 2013, The American Physiological Society.


Figure 20. Noradrenergic constriction is enhanced in human preterm cerebral arteries. (A) Catecholamine histofluorescence of perivascular nerves is prominent in proximal middle cerebral artery at gestational age 38 weeks (a) and term (b) and in basilar artery at gestational age 32 weeks (c), 5 weeks (d), and 38 weeks (e). (B) Catecholamine histofluorescence in perivascular nerves is also prominent at gestational age of 24 weeks in distal middle cerebral artery (a) and a branch of basilar artery (b). (C) The response of middle cerebral artery from infant of 24 weeks of gestation to norepinephrine before (left) and after (right) endothelium inactivation, which attenuated the relaxation to acetylcholine while preserving norepinephrine‐induced contraction. Source: Adapted, with permission, from Bevan R, et al. 1998 56, © 1998, Springer Nature. Comparison of the dose to produce constriction in 50% of cerebral arteries (ED50) (D) and normalized tension generated in response to norepinephrine (E) and electrical field stimulation (F) of cerebral arteries studied within 2 days of autopsy in eight neonates at 23 to 38 weeks PMA and 21 adults (mean ± SE). Comparison of the mean number of catecholaminergic nerve fibers per millimeter length of cerebral arteries in 2 neonates and 15 adults (G). Source: Adapted, with permission, from Bevan R, et al., 1998 56.


Figure 21. Developmental changes in norepinephrine‐induced constriction of pial arteries. (A) When applied through a closed cranial window, norepinephrine produced dose‐dependent constriction in anesthetized fetal sheep and newborn lambs but not in adult sheep. The effect was most pronounced in fetuses less than 121 days gestation (<0.8 full term). (B) Electrical stimulation of the superior cervical sympathetic ganglion produced constriction of pial arteries that was blocked by the α‐adrenergic antagonist prazosin. Source: Adapted, with permission, from Wagerle LC, et al., 1990 442, © 1990, The American Physiological Society.


Figure 22. Developmental changes in blood‐brain barrier permeability in sheep. The influx transfer constant (Ki) for radiolabeled aminoisobutyric acid shows a progressive decrease from 60% of gestation in fetal sheep through adulthood in various brain regions. However, even at 60% of gestation, the transfer constant is not considered high and indicates that significant diffusion restriction is present for this metabolically inert small molecule. Source: Modified, with permission, from Stonestreet BS, et al., 1996 402, © 1996, The American Physiological Society.


Figure 23. Developmental changes in tight junction protein organization. Immunostaining of occludin and claudin‐5 shows endothelial cytoplasmic staining with some linear cell membrane assembly of occludin present at 12 weeks PMA in human fetal telencephalon (A, B). By 14 weeks, linear beaded staining of occludin and claudin‐5 becomes evident in the cell membrane (C, D). At 18 weeks, a more mature appearance of nearly linear immunostaining of occludin and claudin‐5 occurs. Source: Adapted, with permission, from Virgintino et al. 434.


Figure 24. Endothelial vasodilator function is depressed in fetal sheep carotid arteries. Postnatal maturation is associated with a moderately increased abundance of endothelial nitric oxide synthase (eNOS). However, when normalized by the luminal surface area or by eNOS concentration, the amount of NO generated in response to the calcium ionophore A23187 or to endothelial shear stress is greater in adult than in fetal arteries. In contrast, NO generation by ADP is not significantly different. Thus, eNOS coupling mechanisms are not fully mature in fetal sheep arteries, and maturation may depend on the nature of the stimulus. Mean ± SE, n = 6‐8. Source: Adapted, with permission, from White CR, et al., 2005 451, © 2005, The American Physiological Society.


Figure 25. Role of CO in immature neurovascular unit in glia limitans. Neurotransmitter glutamate binds to astrocyte ionotropic and metabotropic glutamate (GLU) receptors (iGLUR and mGLUR) expressed on immature astrocytes, elevating [Ca2+]i that increases heme oxygenase‐2 (HO‐2 activity) and generates CO. The CO diffuses to pial artery vascular smooth muscle where it binds to a heme element that increases Ca2+ spark‐to‐BKCa channel coupling, thereby elevating BKCa channel activity. In addition, CO increases Ca2+ spark frequency, further increasing BKCa channel activity. The resulting smooth muscle cell hyperpolarization produces vasodilation. Furthermore, endothelial‐derived NO and PGI2 provide a minimal amount of guanylyl cyclase (GC) and cGMP and/or activation of PGI2 receptors (IP), adenylyl cyclase (AC), and cAMP. The resulting protein kinase G (PKG) activity may phosphorylate sarcoplasmic reticulum RyRs, increasing Ca2+ spark frequency and/or the BKCa channel. In the intact vasculature, NO and PGI2 can activate smooth muscle cell GC and AC directly, if a stimulus is sufficient at the endothelial cell. In addition to glutamate, neurons can release ATP from neurotransmitter vesicles, the ATP can be degraded to ADP by an astrocyte endonucleotidase, and the ADP can act on astrocyte P2Y1 receptors to also activate HO‐2 (not shown). IP3, inositol 1,4,5‐trisphosphate. Source: Adapted, with permission, from Leffler CW, et al., 2011 276, © 2011, The American Physiological Society.


Figure 26. Schematic diagram of endothelial‐derived factors in dilation of pial arterioles to hypercapnia. Acidosis is thought to activate cystathionine γ‐lyase (CSE) and generate H2S, which activates KATP channels and induces hyperpolarization and consequent inhibition of VDCC, voltage‐dependent Ca2+ channels (VDCC). H2S also promotes Ca2+ sparks and activation of BKCa channels (not shown), which also play a role in hypercapnic dilation. In addition, acidosis is thought to stimulate arachidonic acid (AA) release and cyclooxygenase (COX) activity and generate PGI2, which then acts on the smooth muscle prostacyclin receptor (IP) to stimulate adenylyl cyclase (AC). Source: Adapted, with permission, from Leffler CW, et al., 2011 274, © 2011, The American Physiological Society.


Figure 27. Developmental changes in the cerebrovascular response to hypoxia in unanesthetized sheep. (A) Over a wide range of arterial O2 content, cerebral blood flow (CBF) increases from 0.6 gestation to 0.9 gestation in fetal sheep, remains elevated after birth, and then decreases in adults. (B) At 0.6 gestation, the increase in CBF is inadequate to maintain cerebral O2 transport, whereas the O2 transport is maintained later in development during hypoxia. (C) O2 extraction fraction, which is the ratio of CMRO2 to O2 transport, is also maintained during hypoxia later in development, although the level is highest in adults and lowest at 0.6 gestation. The levels of O2 extraction fraction differed among groups despite normalization with the different levels of CMRO2 (indicated as μmol O2/min/100 g). (D‐F) Fetal sheep at 0.9 gestation and neonatal lambs underwent a partial exchange transfusion with adult sheep blood having a P50 (PO2 at 50% oxyhemoglobin saturation) of 44 Torr to increase the P50 from 17 to 32 Torr in fetal sheep and from 26 to 37 Torr in neonatal lambs. Compared to the pretransfusion responses to hypoxia, the CBF and O2 transport responses were reduced, and the O2 extraction fraction was elevated to levels closer to those seen in adult sheep. Source: Adapted, with permission, from Koehler RC, et al., 1986 228, 1984 230; Gleason CA, et al., 1990 161; Rosenberg AA, et al., 1986 382.


Figure 28. (A) Example of real‐time monitoring of pressure‐reactivity index (PRx) for 72 h in a child after TBI. Top trace shows arterial blood pressure (ABP, black) and CPP (gray), second trace shows ICP, and third trace shows PRx. Bottom chart shows the PRx within 5 mmHg CPP bins averaged over 72 h for this single patient. An optimal CPP of 57.5 mmHg could be identified wherein PRx had a minimal value. (B) The average PRx for 36 children after TBI displays a U‐shaped curve. (C) The optimal CPP (CPPopt) in 34 TBI patients correlated with the patient's age. Source: Adapted, with permission, from Lewis PM, et al., 2015 278, © 2015, Wolters Kluwer.


Figure 29. Schematic diagram of potential mechanisms of impaired cerebrovascular regulation after hypoxia‐ischemia, stroke, and fluid percussion injury in piglets. Endothelin‐1 (ET‐1) and vasopressin have been implicated in the release of the endogenous opioid nociception/orphanin FQ (NOC/oFQ) that can result in the generation of superoxide through a PKC signaling in concert with activation of cyclooxygenase (COX). Acting through protein tyrosine kinase (PKT) and ERK/MAPK signaling, superoxide generation results in impaired K+ channel function. Focal stroke stimulates endogenous tissue plasminogen activator (tPA) that is known to augment activation of NMDA receptors. This pathway can result in increased ET‐1 and activation of ERK/MAPK. It also produces an inflammatory response leading to increased interleukin‐6 and impaired function of K+ channels. Inhaled nitric oxide can block the increase in ET‐1 and preserve autoregulation. Source: Adapted, with permission, from Pastor P, et al., 2019 361; Armstead, WM, 2005 34.


Figure 30. Synthesis of the relative time course of developmental regulation of the cerebral circulation and energy metabolism. Blood‐brain barrier (BBB) function, CMRO2, CMRGluc, cerebral O2 transport, CBF reactivity to CO2, and neurovascular coupling are normalized relative to an adult value of 1. Autoregulation is displayed in terms of the upper (ULA) and lower (LLA) limits of autoregulation (mmHg) below and above which CBF is pressure passive. All lines are based on human data or, in the cases where human data are not available, on animal data. Interpolation was used in cases where no data are available for a particular developmental stage. Note that energy metabolism and O2 transport overshoot adult values between the infant and adolescent stages, whereas CO2 reactivity does not. Also, O2 transport has a postnatal dip, attributable to transient anemia and the decrease in hemoglobin O2 affinity as fetal hemoglobin is replaced with adult hemoglobin. Neurovascular coupling is thought to have a transient increase near term and then decrease during the period of elevated CMRGluc. The range of autoregulation between the ULA and LLA progressively expands during development.
References
 1.Abrams RM, Ito M, Frisinger JE, Patlak CS, Pettigrew KD, Kennedy C. Local cerebral glucose utilization in fetal and neonatal sheep. Am J Physiol Regul Integr Comp Physiol 246: R608‐R618, 1984.
 2.Acosta S, Penny DJ, Brady KM, Rusin CG. An effective model of cerebrovascular pressure reactivity and blood flow autoregulation. Microvasc Res 115: 34‐43, 2018. DOI: 10.1016/j.mvr.2017.08.006.
 3.Adachi K, Takahashi S, Melzer P, Campos KL, Nelson T, Kennedy C, Sokoloff L. Increases in local cerebral blood flow associated with somatosensory activation are not mediated by NO. Am J Physiol Heart Circ Physiol 267: H2155‐H2162, 1994.
 4.Adams RH, Eichmann A. Axon guidance molecules in vascular patterning. Cold Spring Harb Perspect Biol 2: a001875, 2010. DOI: 10.1101/cshperspect.a001875.
 5.Akopov SE, Zhang L, Pearce WJ. Regulation of Ca2+ sensitization by PKC and rho proteins in ovine cerebral arteries: Effects of artery size and age. Am J Physiol 275: H930‐H939, 1998.
 6.Alkayed NJ, Birks EK, Narayanan J, Petrie KA, Kohler‐Cabot AE, Harder DR. Role of P‐450 arachidonic acid expoygenase in the response of cerebral blood flow to glutamate in rats. Stroke 28: 1066‐1072, 1997.
 7.Alkayed NJ, Narayanan J, Gebremedhin D, Medhora M, Roman RJ, Harder DR. Molecular characterization of an arachidonic acid epoxygenase in rat brain astrocytes. Stroke 27: 971‐979, 1996.
 8.Allievi AG, Arichi T, Tusor N, Kimpton J, Arulkumaran S, Counsell SJ, Edwards AD, Burdet E. Maturation of sensori‐motor functional responses in the preterm brain. Cereb Cortex 26: 402‐413, 2016. DOI: 10.1093/cercor/bhv203.
 9.Alonso‐Galicia M, Hudetz AG, Shen H, Harder DR, Roman RJ. Contribution of 20‐HETE to vasodilator actions of nitric oxide in the cerebral microcirculation. Stroke 30: 2727‐2734, 1999.
 10.Altman DI, Perlman JM, Volpe JJ, Powers WJ. Cerebral oxygen metabolism in newborns. Pediatrics 92: 99‐104, 1993.
 11.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.
 12.Anderson AW, Marois R, Colson ER, Peterson BS, Duncan CC, Ehrenkranz RA, Schneider KC, Gore JC, Ment LR. Neonatal auditory activation detected by functional magnetic resonance imaging. Magn Reson Imaging 19: 1‐5, 2001.
 13.Andresen JJ, Shafi NI, Durante W, Bryan RM Jr. Effects of carbon monoxide and heme oxygenase inhibitors in cerebral vessels of rats and mice. Am J Physiol Heart Circ Physiol 291: H223‐H230, 2006. DOI: 10.1152/ajpheart.00058.2006.
 14.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. DOI: 10.1016/j.neuroimage.2012.06.054.
 15.Arichi T, Moraux A, Melendez A, Doria V, Groppo M, Merchant N, Combs S, Burdet E, Larkman DJ, Counsell SJ, Beckmann CF, Edwards AD. Somatosensory cortical activation identified by functional MRI in preterm and term infants. NeuroImage 49: 2063‐2071, 2010. DOI: 10.1016/j.neuroimage.2009.10.038.
 16.Armstead WM. The contribution of delta 1‐ and delta 2‐opioid receptors to hypoxia‐induced pial artery dilation in the newborn pig. J Cereb Blood Flow Metab 15: 539‐546, 1995. DOI: 10.1038/jcbfm.1995.67.
 17.Armstead WM. Opioids and nitric oxide contribute to hypoxia‐induced pial arterial vasodilation in newborn pigs. Am J Physiol Heart Circ Physiol 268: H226‐H232, 1995.
 18.Armstead WM. Role of activation of calcium‐sensitive K+ channels and cAMP in opioid‐induced pial artery dilation. Brain Res 747: 252‐258, 1997.
 19.Armstead WM. Role of activation of calcium‐sensitive K+ channels in NO‐ and hypoxia‐induced pial artery vasodilation. Am J Physiol Heart Circ Physiol 272: H1785‐H1790, 1997. DOI: 10.1152/ajpheart.1997.272.4.H1785.
 20.Armstead WM. Role of nitric oxide, cyclic nucleotides, and the activation of ATP‐sensitive K+ channels in the contribution of adenosine to hypoxia‐induced pial artery dilation. J Cereb Blood Flow Metab 17: 100‐108, 1997. DOI: 10.1097/00004647‐199701000‐00013.
 21.Armstead WM. Contribution of KCa channel activation to hypoxic cerebrovasodilation does not involve NO. Brain Res 799: 44‐48, 1998.
 22.Armstead WM. Nitric oxide contributes to opioid release from glia during hypoxia. Brain Res 813: 398‐401, 1998.
 23.Armstead WM. Relationship among NO, the KATP channel, and opioids in hypoxic pial artery dilation. Am J Physiol Heart Circ Physiol 275: H988‐H994, 1998.
 24.Armstead WM. Role of opioids in hypoxic pial artery dilation is stimulus duration dependent. Am J Physiol Heart Circ Physiol 275: H861‐H867, 1998.
 25.Armstead WM. Age‐dependent impairment of K(ATP) channel function following brain injury. J Neurotrauma 16: 391‐402, 1999. DOI: 10.1089/neu.1999.16.391.
 26.Armstead WM. Endothelin‐1 contributes to normocapnic hyperoxic pial artery vasoconstriction. Brain Res 842: 252‐255, 1999.
 27.Armstead WM. Hypotension dilates pial arteries by KATP and KCa channel activation. Brain Res 816: 158‐164, 1999.
 28.Armstead WM. Role of nociceptin/orphanin FQ in age‐dependent cerebral hemodynamic effects of brain injury. J Neurotrauma 17: 751‐764, 2000. DOI: 10.1089/neu.2000.17.751.
 29.Armstead WM. Endothelin‐induced cyclooxygenase‐dependent superoxide generation contributes to K+ channel functional impairment after brain injury. J Neurotrauma 18: 1039‐1048, 2001. DOI: 10.1089/08977150152693737.
 30.Armstead WM. Vasopressin‐induced protein kinase C‐dependent superoxide generation contributes to ATP‐sensitive potassium channel but not calcium‐sensitive potassium channel function impairment after brain injury. Stroke 32: 1408‐1414, 2001.
 31.Armstead WM. Vasopressin induced cyclooxygenase dependent superoxide generation contributes to K(+) channel function impairment after brain injury. Brain Res 910: 19‐28, 2001.
 32.Armstead WM. Age‐dependent NOC/oFQ contribution to impaired hypotensive cerebral hemodynamics after brain injury. J Neurotrauma 19: 1193‐1202, 2002. DOI: 10.1089/08977150260337994.
 33.Armstead WM. Protein tyrosine kinase and mitogen‐activated protein kinase activation contribute to K(ATP) and K(ca) channel impairment after brain injury. Brain Res 943: 276‐282, 2002.
 34.Armstead WM. Age and cerebral circulation. Pathophysiology 12: 5‐15, 2005. DOI: 10.1016/j.pathophys.2005.01.002.
 35.Armstead WM, Hekierski H, Pastor P, Yarovoi S, Higazi AA, Cines DB. Release of IL‐6 after stroke contributes to impaired cerebral autoregulation and hippocampal neuronal necrosis through NMDA receptor activation and upregulation of ET‐1 and JNK. Transl Stroke Res 10: 104‐111, 2019. DOI: 10.1007/s12975‐018‐0617‐z.
 36.Armstead WM, Hekierski H, Yarovoi S, Higazi AA, Cines DB. tPA variant tPA‐A(296‐299) prevents impairment of cerebral autoregulation and necrosis of hippocampal neurons after stroke by inhibiting upregulation of ET‐1. J Neurosci Res 96: 128‐137, 2018. DOI: 10.1002/jnr.24112.
 37.Armstead WM, Mirro R, Busija DW, Leffler CW. Permissive role of prostanoids in acetylcholine‐induced cerebral vasoconstriction. J Pharmacol Exp Ther 251: 1012‐1019, 1989.
 38.Armstead WM, Riley J, Vavilala MS. TBI sex dependently upregulates ET‐1 to impair autoregulation, which is aggravated by phenylephrine in males but is abrogated in females. J Neurotrauma 29: 1483‐1490, 2012. DOI: 10.1089/neu.2011.2248.
 39.Armstead WM, Riley J, Vavilala MS. Dopamine prevents impairment of autoregulation after traumatic brain injury in the newborn pig through inhibition of Up‐regulation of endothelin‐1 and extracellular signal‐regulated kinase mitogen‐activated protein kinase. Pediatr Crit Care Med 14: e103‐e111, 2013. DOI: 10.1097/PCC.0b013e3182712b44.
 40.Armstead WM, Riley J, Vavilala MS. Norepinephrine protects cerebral autoregulation and reduces hippocampal necrosis after traumatic brain injury via blockade of ERK MAPK and IL‐6 in juvenile pigs. J Neurotrauma 33: 1761‐1767, 2016. DOI: 10.1089/neu.2015.4290.
 41.Armstead WM, Riley J, Vavilala MS. Preferential protection of cerebral autoregulation and reduction of hippocampal necrosis with norepinephrine after traumatic brain injury in female piglets. Pediatr Crit Care Med 17: e130‐e137, 2016. DOI: 10.1097/PCC.0000000000000603.
 42.Armstead WM, Riley J, Vavilala MS. K channel impairment determines sex and age differences in epinephrine‐mediated outcomes after brain injury. J Neurosci Res 95: 1917‐1926, 2017. DOI: 10.1002/jnr.24063.
 43.Armstead WM, Riley J, Vavilala MS. Sex and age differences in epinephrine mechanisms and outcomes after brain injury. J Neurotrauma 34: 1666‐1675, 2017. DOI: 10.1089/neu.2016.4770.
 44.Armstead WM, Vavilala MS. Translational approach towards determining the role of cerebral autoregulation in outcome after traumatic brain injury. Exp Neurol 317: 291‐297, 2019. DOI: 10.1016/j.expneurol.2019.03.015.
 45.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. DOI: 10.1038/jcbfm.1994.142.
 46.Attwell D, Mishra A, Hall CN, O'Farrell FM, Dalkara T. What is a pericyte? J Cereb Blood Flow Metab 36: 451‐455, 2016. DOI: 10.1177/0271678X15610340.
 47.Baerts W, van Bel F, Thewissen L, Derks JB, Lemmers PM. Tocolytic indomethacin: Effects on neonatal haemodynamics and cerebral autoregulation in the preterm newborn. Arch Dis Child Fetal Neonatal Ed 98: F419‐F423, 2013. DOI: 10.1136/archdischild‐2012‐302532.
 48.Baranov D, Armstead WM. Selective blockade of AT1 receptor attenuates impairment of hypotensive autoregulation and improves cerebral blood flow after brain injury in the newborn pig. Anesthesiology 99: 1118‐1124, 2003.
 49.Bari F, Louis TM, Meng W, Busija DW. Global ischemia impairs ATP‐sensitive K+ channel function in cerebral arterioles in piglets. Stroke 27: 1874‐1880; discussion 1880‐1871, 1996.
 50.Barkoudah E, Jaggar JH, Leffler CW. The permissive role of endothelial NO in CO‐induced cerebrovascular dilation. Am J Physiol Heart Circ Physiol 287: H1459‐H1465, 2004.
 51.Barkovich AJ, Latal‐Hajnal B, Partridge JC, Sola A, Ferriero DM. MR contrast enhancement of the normal neonatal brain. AJNR Am J Neuroradiol 18: 1713‐1717, 1997.
 52.Barkovich AJ, Westmark K, Partridge C, Sola A, Ferriero DM. Perinatal asphyxia: MR findings in the first 10 days. Am J Neuroradiol 16: 427‐438, 1995.
 53.Baumbach GL, Heistad DD. Effects of sympathetic stimulation and changes in arterial pressure on segmental resistance of cerebral vessels in rabbits and cats. Circ Res 52: 527‐533, 1983.
 54.Bevan JA. Sites of transition between functional systemic and cerebral arteries of rabbits occur at embryological junctional sites. Science 204: 635‐637, 1979.
 55.Bevan JA, Duckworth J, Laher I, Oriowo MA, McPherson GA, Bevan RD. Sympathetic control of cerebral arteries: Specialization in receptor type, reserve, affinity, and distribution. FASEB J 1: 193‐198, 1987.
 56.Bevan R, Dodge J, Nichols P, Poseno T, Vijayakumaran E, Wellman T, Bevan JA. Responsiveness of human infant cerebral arteries to sympathetic nerve stimulation and vasoactive agents. Pediatr Res 44: 730‐739, 1998. DOI: 10.1203/00006450‐199811000‐00016.
 57.Bevan RD, Dodge J, Nichols P, Penar PL, Walters CL, Wellman T, Bevan JA. Weakness of sympathetic neural control of human pial compared with superficial temporal arteries reflects low innervation density and poor sympathetic responsiveness. Stroke 29: 212‐221, 1998.
 58.Bevan RD, Vijayakumaran E, Gentry A, Wellman T, Bevan JA. Intrinsic tone of cerebral artery segments of human infants between 23 weeks of gestation and term. Pediatr Res 43: 20‐27, 1998. DOI: 10.1203/00006450‐199804001‐00125.
 59.Bishai JM, Blood AB, Hunter CJ, Longo LD, Power GG. Fetal lamb cerebral blood flow (CBF) and oxygen tensions during hypoxia: A comparison of laser Doppler and microsphere measurements of CBF. J Physiol 546: 869‐878, 2003.
 60.Boedtkjer E. Acid‐base regulation and sensing: Accelerators and brakes in metabolic regulation of cerebrovascular tone. J Cereb Blood Flow Metab 38: 588‐602, 2018. DOI: 10.1177/0271678X17733868.
 61.Bohlen HG. Nitric oxide and the cardiovascular system. Compr Physiol 5: 808‐823, 2015. DOI: 10.1002/c140052.
 62.Bohlen HG, Harper SL. Evidence of myogenic vascular control in the rat cerebral cortex. Circ Res 55: 554‐559, 1984.
 63.Borel CO, Backofen JE, Koehler RC, Jones MD Jr, Traystman RJ. Cerebral blood flow autoregulation during intracranial hypertension in hypoxic lambs. Am J Physiol Heart Circ Physiol 253: H1342‐H1348, 1987.
 64.Born AP, Rostrup E, Miranda MJ, Larsson HB, Lou HC. Visual cortex reactivity in sedated children examined with perfusion MRI (FAIR). Magn Reson Imaging 20: 199‐205, 2002.
 65.Born P, Leth H, Miranda MJ, Rostrup E, Stensgaard A, Peitersen B, Larsson HB, Lou HC. Visual activation in infants and young children studied by functional magnetic resonance imaging. Pediatr Res 44: 578‐583, 1998. DOI: 10.1203/00006450‐199810000‐00018.
 66.Brady K, Joshi B, Zweifel C, Smielewski P, Czosnyka M, Easley RB, Hogue CW Jr. Real‐time continuous monitoring of cerebral blood flow autoregulation using near‐infrared spectroscopy in patients undergoing cardiopulmonary bypass. Stroke 41: 1951‐1956, 2010.
 67.Brady KM, Easley RB, Kibler K, Kaczka DW, Andropoulos D, Fraser CD 3rd, Smielewski P, Czosnyka M, Adams GJ, Rhee CJ, Rusin CG. Positive end‐expiratory pressure oscillation facilitates brain vascular reactivity monitoring. J Appl Physiol 113: 1362‐1368, 2012. DOI: 10.1152/japplphysiol.00853.2012.
 68.Brady KM, Lee JK, Kibler KK, Easley RB, Koehler RC, Czosnyka M, Smielewski P, Shaffner DH. The lower limit of cerebral blood flow autoregulation is increased with elevated intracranial pressure. Anesth Analg 108: 1278‐1283, 2009.
 69.Brady KM, Lee JK, Kibler KK, Easley RB, Koehler RC, Shaffner DH. Continuous measurement of autoregulation by spontaneous fluctuations in cerebral perfusion pressure: Comparison of 3 methods. Stroke 39: 2531‐2537, 2008.
 70.Brady KM, Lee JK, Kibler KK, Smielewski P, Czosnyka M, Easley RB, Koehler RC, Shaffner DH. Continuous time‐domain analysis of cerebrovascular autoregulation using near‐infrared spectroscopy. Stroke 38: 2818‐2825, 2007.
 71.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.
 72.Brady KM, Shaffner DH, Lee JK, Easley RB, Smielewski P, Czosnyka M, Jallo GI, Guerguerian AM. Continuous monitoring of cerebrovascular pressure reactivity after traumatic brain injury in children. Pediatrics 124: e1205‐e1212, 2009.
 73.Brian JE Jr, Heistad DD, Faraci FM. Effect of carbon monoxide on rabbit cerebral arteries. Stroke 25: 639‐643, 1994.
 74.Brubakk AM, Bratlid D, Oh W, Yao AC, Stonestreet BS. Atropine prevents increases in brain blood flow during hypertension in newborn piglets. Pediatr Res 18: 1121‐1126, 1984. DOI: 10.1203/00006450‐198411000‐00013.
 75.Bucher HU, Edwards AD, Lipp AE, Duc G. Comparison between near infrared spectroscopy and 133Xenon clearance for estimation of cerebral blood flow in critically ill preterm infants. Pediatr Res 33: 56‐60, 1993. DOI: 10.1203/00006450‐199301000‐00012.
 76.Buerk DG, Ances BM, Greenberg JH, Detre JA. Temporal dynamics of brain tissue nitric oxide during functional forepaw stimulation in rats. NeuroImage 18: 1‐9, 2003.
 77.Burton VJ, Gerner G, Cristofalo E, Chung SE, Jennings JM, Parkinson C, Koehler RC, Chavez‐Valdez R, Johnston MV, Northington FJ, Lee JK. A pilot cohort study of cerebral autoregulation and 2‐year neurodevelopmental outcomes in neonates with hypoxic‐ischemic encephalopathy who received therapeutic hypothermia. BMC Neurol 15: 209, 2015. DOI: 10.1186/s12883‐015‐0464‐4.
 78.Busija DW, Leffler CW. Exogenous norepinephrine constricts cerebral arterioles via alpha 2‐adrenoceptors in newborn pigs. J Cereb Blood Flow Metab 7: 184‐188, 1987.
 79.Busija DW, Leffler CW. Postjunctional alpha 2‐adrenoceptors in pial arteries of anesthetized newborn pigs. Dev Pharmacol Ther 10: 36‐46, 1987.
 80.Busija DW, Leffler CW. Role of prostanoids in cerebrovascular responses during seizures in piglets. Am J Physiol Heart Circ Physiol 256: H120‐H125, 1989.
 81.Busija DW, Leffler CW, Wagerle LC. Responses of newborn pig pial arteries to sympathetic nervous stimulation and exogenous norepinephrine. Pediatr Res 19: 1210‐1214, 1985.
 82.Busija DW, Marcus ML, Heistad DD. Pial artery diameter and blood flow velocity during sympathetic stimulation in cats. J Cereb Blood Flow Metab 2: 363‐367, 1982.
 83.Busija DW, Meng W, Bari F, McGough PS, Errico RA, Tobin JR, Louis TM. Effects of ischemia on cerebrovascular responses to N‐methyl‐d‐aspartate in piglets. Am J Physiol Heart Circ Physiol 270: H1225‐H1230, 1996. DOI: 10.1152/ajpheart.1996.270.4.H1225.
 84.Busija DW, Rutkai I, Dutta S, Katakam PV. Role of mitochondria in cerebral vascular function: energy production, cellular protection, and regulation of vascular tone. Compr Physiol 6: 1529‐1548, 2016. DOI: 10.1002/c150051.
 85.Busija DW, Wagerle LC, Pourcyrous M, Leffler CW. Acetylcholine dramatically increases prostanoid synthesis in piglet parietal cortex. Brain Res 439: 122‐126, 1988.
 86.Busija DW, Wei M. Altered cerebrovascular responsiveness to N‐methyl‐d‐aspartate after asphyxia in piglets. Am J Physiol Heart Circ Physiol 265: H389‐H394, 1993.
 87.Cai C, Fordsmann JC, Jensen SH, Gesslein B, Lonstrup M, Hald BO, Zambach SA, Brodin B, Lauritzen MJ. Stimulation‐induced increases in cerebral blood flow and local capillary vasoconstriction depend on conducted vascular responses. Proc Natl Acad Sci U S A 115: E5796‐E5804, 2018. DOI: 10.1073/pnas.1707702115.
 88.Carratu P, Pourcyrous M, Fedinec A, Leffler CW, Parfenova H. Endogenous heme oxygenase prevents impairment of cerebral vascular functions caused by seizures. Am J Physiol Heart Circ Physiol 285: H1148‐H1157, 2003.
 89.Cauli B, Hamel E. Revisiting the role of neurons in neurovascular coupling. Front Neuroenergetics 2: 9, 2010. DOI: 10.3389/fnene.2010.00009.
 90.Cauli B, Tong XK, Rancillac A, Serluca N, Lambolez B, Rossier J, Hamel E. Cortical GABA interneurons in neurovascular coupling: Relays for subcortical vasoactive pathways. J Neurosci 24: 8940‐8949, 2004. DOI: 10.1523/JNEUROSCI.3065‐04.2004.
 91.Cavazutti M, Duffy TE. Regulation of local cerebral blood flow in normal and hypoxic newborn dogs. Ann Neurol 11: 247‐257, 1982.
 92.Charles SM, Zhang L, Longo LD, Buchholz JN, Pearce WJ. Postnatal maturation attenuates pressure‐evoked myogenic tone and stretch‐induced increases in Ca2+ in rat cerebral arteries. Am J Physiol Regul Integr Comp Physiol 293: R737‐R744, 2007. DOI: 10.1152/ajpregu.00869.2006.
 93.Charpie JR, Schreur KD, Papadopoulos SM, Webb RC. Endothelium dependency of contractile activity differs in infant and adult vertebral arteries. J Clin Invest 93: 1339‐1343, 1994. DOI: 10.1172/JCI117093.
 94.Chen BR, Kozberg MG, Bouchard MB, Shaik MA, Hillman EM. A critical role for the vascular endothelium in functional neurovascular coupling in the brain. J Am Heart Assoc 3: e000787, 2014. DOI: 10.1161/JAHA.114.000787.
 95.Chen S, Tisch N, Kegel M, Yerbes R, Hermann R, Hudalla H, Zuliani C, Gulculer GS, Zwadlo K, von Engelhardt J, Ruiz de Almodovar C, Martin‐Villalba A. CNS macrophages control neurovascular development via CD95L. Cell Rep 19: 1378‐1393, 2017. DOI: 10.1016/j.celrep.2017.04.056.
 96.Chiron C, Raynaud C, Maziere B, Zilbovicius M, Laflamme L, Masure MC, Dulac O, Bourguignon M, Syrota A. Changes in regional cerebral blood flow during brain maturation in children and adolescents. J Nucl Med 33: 696‐703, 1992.
 97.Chugani HT. Biological basis of emotions: Brain systems and brain development. Pediatrics 102: 1225‐1229, 1998.
 98.Chugani HT, Hovda DA, Villablanca JR, Phelps ME, Xu WF. Metabolic maturation of the brain: A study of local cerebral glucose utilization in the developing cat. J Cereb Blood Flow Metab 11: 35‐47, 1991. DOI: 10.1038/jcbfm.1991.4.
 99.Chugani HT, Phelps ME. Maturational changes in cerebral function in infants determined by 18FDG positron emission tomography. Science 231: 840‐843, 1986.
 100.Chugani HT, Phelps ME, Mazziotta JC. Positron emission tomography study of human brain functional development. Ann Neurol 22: 487‐497, 1987. DOI: 10.1002/ana.410220408.
 101.Cikic S, Chandra PK, Harman JC, Rutkai I, Katakam PV, Guidry JJ, Gidday JM, Busija DW. Sexual differences in mitochondrial and related proteins in rat cerebral microvessels: A proteomic approach. J Cereb Blood Flow Metab 41: 397‐412, 2021. DOI: 10.1177/0271678X20915127.
 102.Clough GF. Developmental conditioning of the vasculature. Compr Physiol 5: 397‐438, 2015. DOI: 10.1002/c140037.
 103.Colonnese MT, Phillips MA, Constantine‐Paton M, Kaila K, Jasanoff A. Development of hemodynamic responses and functional connectivity in rat somatosensory cortex. Nat Neurosci 11: 72‐79, 2008. DOI: 10.1038/nn2017.
 104.Craigie EH. Postnatal changes in vascularity in the cerebral cortex of the male albino rat. J Comp Neurol 39: 301‐324, 1925.
 105.Cremer JE. Nutrients for the brain: Problems in supply. Early Hum Dev 5: 117‐132, 1981.
 106.Cremer JE, Braun LD, Oldendorf WH. Changes during development in transport processes of the blood‐brain barrier. Biochim Biophys Acta 448: 633‐637, 1976.
 107.Cremer JE, Cunningham VJ, Pardridge WM, Braun LD, Oldendorf WH. Kinetics of blood‐brain barrier transport of pyruvate, lactate and glucose in suckling, weanling and adult rats. J Neurochem 33: 439‐445, 1979.
 108.Curvello V, Hekierski H, Pastor P, Vavilala MS, Armstead WM. Dopamine protects cerebral autoregulation and prevents hippocampal necrosis after traumatic brain injury via block of ERK MAPK in juvenile pigs. Brain Res 1670: 118‐124, 2017. DOI: 10.1016/j.brainres.2017.06.010.
 109.Curvello V, Hekierski H, Riley J, Vavilala M, Armstead WM. Sex and age differences in phenylephrine mechanisms and outcomes after piglet brain injury. Pediatr Res 82: 108‐113, 2017. DOI: 10.1038/pr.2017.83.
 110.Curvello V, Pastor P, Hekierski H, Armstead WM. Inhaled nitric oxide protects cerebral autoregulation and reduces hippocampal necrosis after traumatic brain injury through inhibition of ET‐1, ERK MAPK and IL‐6 upregulation in pigs. Neurocrit Care 30: 467‐477, 2019. DOI: 10.1007/s12028‐018‐0638‐1.
 111.Dacey RG Jr, Duling BR. Effect of norepinephrine on penetrating arterioles of rat cerebral cortex. Am J Physiol Heart Circ Physiol 246: H380‐H385, 1984. DOI: 10.1152/ajpheart.1984.246.3.H380.
 112.De Vis JB, Alderliesten T, Hendrikse J, Petersen ET, Benders MJ. Magnetic resonance imaging based noninvasive measurements of brain hemodynamics in neonates: A review. Pediatr Res 80: 641‐650, 2016. DOI: 10.1038/pr.2016.146.
 113.De Vis JB, Hendrikse J, Petersen ET, de Vries LS, van Bel F, Alderliesten T, Negro S, Groenendaal F, Benders MJ. Arterial spin‐labelling perfusion MRI and outcome in neonates with hypoxic‐ischemic encephalopathy. Eur Radiol 25: 113‐121, 2015. DOI: 10.1007/s00330‐014‐3352‐1.
 114.De Vis JB, Petersen ET, Alderliesten T, Groenendaal F, de Vries LS, van Bel F, Benders MJ, Hendrikse J. Non‐invasive MRI measurements of venous oxygenation, oxygen extraction fraction and oxygen consumption in neonates. NeuroImage 95: 185‐192, 2014. DOI: 10.1016/j.neuroimage.2014.03.060.
 115.De Vis JB, Petersen ET, de Vries LS, Groenendaal F, Kersbergen KJ, Alderliesten T, Hendrikse J, Benders MJ. Regional changes in brain perfusion during brain maturation measured non‐invasively with Arterial Spin Labeling MRI in neonates. Eur J Radiol 82: 538‐543, 2013. DOI: 10.1016/j.ejrad.2012.10.013.
 116.De Vis JB, Petersen ET, Kersbergen KJ, Alderliesten T, de Vries LS, van Bel F, Groenendaal F, Lemmers PM, Hendrikse J, Benders MJ. Evaluation of perinatal arterial ischemic stroke using noninvasive arterial spin labeling perfusion MRI. Pediatr Res 74: 307‐313, 2013. DOI: 10.1038/pr.2013.111.
 117.de Vries LS, Groenendaal F. Patterns of neonatal hypoxic‐ischaemic brain injury. Neuroradiology 52: 555‐566, 2010. DOI: 10.1007/s00234‐010‐0674‐9.
 118.Domoki F, Perciaccante JV, Veltkamp R, Bari F, Busija DW. Mitochondrial potassium channel opener diazoxide preserves neuronal‐vascular function after cerebral ischemia in newborn pigs. Stroke 30: 2713‐2718; discussion 2718‐2719, 1999.
 119.Donahue S, Pappas GD. The fine structure of capillaries in the cerebral cortex of the rat at various stages of development. Am J Anat 108: 331‐347, 1961. DOI: 10.1002/aja.1001080307.
 120.Donegan JH, Traystman RJ, Koehler RC, Jones MD Jr, Rogers MC. Cerebrovascular hypoxic and autoregulatory responses during reduced brain metabolism. Am J Physiol Heart Circ Physiol 249: H421‐H429, 1985.
 121.Dumont I, Peri KG, Hardy P, Hou X, Martinez‐Bermudez AK, Molotchnikoff S, Varma DR, Chemtob S. PGE2, via EP3 receptors, regulates brain nitric oxide synthase in the perinatal period. Am J Physiol Regul Integr Comp Physiol 275: R1812‐R1821, 1998.
 122.Dunn KM, Hill‐Eubanks DC, Liedtke WB, Nelson MT. TRPV4 channels stimulate Ca2+‐induced Ca2+ release in astrocytic endfeet and amplify neurovascular coupling responses. Proc Natl Acad Sci U S A 110: 6157‐6162, 2013. DOI: 10.1073/pnas.1216514110.
 123.Dziegielewska KM, Evans CA, Lorscheider FL, Malinowska DH, Mollgard K, Reynolds ML, Saunders NR. Plasma proteins in fetal sheep brain: Blood‐brain barrier and intracerebral distribution. J Physiol 318: 239‐250, 1981.
 124.Dziegielewska KM, Evans CAN, Malinowska DH, Mollgard K, Reynolds JM, Reynolds ML, Saunders NR. Studies of the development of brain barrier systems to lipid insoluble molecules in fetal sheep. J Physiol Lond 292: 207‐231, 1979.
 125.Easley RB, Kibler KK, Brady KM, Joshi B, Ono M, Brown C, Hogue CW. Continuous cerebrovascular reactivity monitoring and autoregulation monitoring identify similar lower limits of autoregulation in patients undergoing cardiopulmonary bypass. Neurol Res 35: 344‐354, 2013. DOI: 10.1179/1743132812Y.0000000145.
 126.Easley RB, Marino BS, Jennings J, Cassedy AE, Kibler KK, Brady KM, Andropoulos DB, Brunetti M, Hogue CW, Heitmiller ES, Lee JK, Spaeth J, Everett AD. Impaired cerebral autoregulation and elevation in plasma glial fibrillary acidic protein level during cardiopulmonary bypass surgery for CHD. Cardiol Young 28: 55‐65, 2018. DOI: 10.1017/S1047951117001573.
 127.Eidson TH, Edrington JL, Albuquerque ML, Zuckerman SL, Leffler CW. Light/dye microvascular injury eliminates pial arteriolar dilation in hypotensive piglets. Pediatr Res 37: 10‐14, 1995.
 128.Elliott CF, Pearce WJ. Effects of maturation on cell water, protein, and DNA content in ovine cerebral arteries. J Appl Physiol (1985) 79: 831‐837, 1995. DOI: 10.1152/jappl.1995.79.3.831.
 129.Elliott SR, Pearce WJ. Effects of maturation on alpha‐adrenergic receptor affinity and occupancy in small cerebral arteries. Am J Physiol Heart Circ Physiol 267: H757‐H763, 1994. DOI: 10.1152/ajpheart.1994.267.2.H757.
 130.Enager P, Piilgaard H, Offenhauser N, Kocharyan A, Fernandes P, Hamel E, Lauritzen M. Pathway‐specific variations in neurovascular and neurometabolic coupling in rat primary somatosensory cortex. J Cereb Blood Flow Metab 29: 976‐986, 2009. DOI: 10.1038/jcbfm.2009.23.
 131.Engl E, Jolivet R, Hall CN, Attwell D. Non‐signalling energy use in the developing rat brain. J Cereb Blood Flow Metab 37: 951‐966, 2017. DOI: 10.1177/0271678X16648710.
 132.Erberich SG, Friedlich P, Seri I, Nelson MD Jr, Bluml S. Functional MRI in neonates using neonatal head coil and MR compatible incubator. NeuroImage 20: 683‐692, 2003. DOI: 10.1016/S1053‐8119(03)00370‐7.
 133.Erberich SG, Panigrahy A, Friedlich P, Seri I, Nelson MD, Gilles F. Somatosensory lateralization in the newborn brain. NeuroImage 29: 155‐161, 2006. DOI: 10.1016/j.neuroimage.2005.07.024.
 134.Evans CAN, Reynolds JM, Reynolds ML, Saunders NR, Segal MB. The development of a blood‐brain barrier mechanism in fetal sheep. J Physiol Lond 238: 371‐386, 1974.
 135.Faraci FM, Taugher RJ, Lynch C, Fan R, Gupta S, Wemmie JA. Acid‐sensing ion channels: Novel mediators of cerebral vascular responses. Circ Res 125: 907‐920, 2019. DOI: 10.1161/CIRCRESAHA.119.315024.
 136.Farzam P, Buckley EM, Lin PY, Hagan K, Grant PE, Inder TE, Carp SA, Franceschini MA. Shedding light on the neonatal brain: Probing cerebral hemodynamics by diffuse optical spectroscopic methods. Sci Rep 7: 15786, 2017. DOI: 10.1038/s41598‐017‐15995‐1.
 137.Faustino JV, Wang X, Johnson CE, Klibanov A, Derugin N, Wendland MF, Vexler ZS. Microglial cells contribute to endogenous brain defenses after acute neonatal focal stroke. J Neurosci 31: 12992‐13001, 2011. DOI: 10.1523/JNEUROSCI.2102‐11.2011.
 138.Ferguson RK, Woodbury DM. Penetration of 14C‐inulin and 14C‐sucrose into brain, cerebrospinal fluid, and skeletal muscle of developing rats. Exp Brain Res 7: 181‐194, 1969.
 139.Fernandez‐Lopez D, Faustino J, Klibanov AL, Derugin N, Blanchard E, Simon F, Leib SL, Vexler ZS. Microglial cells prevent hemorrhage in neonatal focal arterial stroke. J Neurosci 36: 2881‐2893, 2016. DOI: 10.1523/JNEUROSCI.0140‐15.2016.
 140.Figaji AA, Zwane E, Fieggen AG, Argent AC, Le Roux PD, Siesjo P, Peter JC. Pressure autoregulation, intracranial pressure, and brain tissue oxygenation in children with severe traumatic brain injury. J Neurosurg Pediatr 4: 420‐428, 2009. DOI: 10.3171/2009.6.PEDS096.
 141.Filosa JA, Bonev AD, Straub SV, Meredith AL, Wilkerson MK, Aldrich RW, Nelson MT. Local potassium signaling couples neuronal activity to vasodilation in the brain. Nat Neurosci 9: 1397‐1403, 2006.
 142.Fiumana E, Parfenova H, Jagger JH, Leffler CW. Carbon monoxide mediates vasodilator effects of glutamate in isolated pressurized cerebral arterioles of newborn pigs. Am J Physiol Heart Circ Physiol 284: H1073‐H1079, 2003.
 143.Franceschini MA, Thaker S, Themelis G, Krishnamoorthy KK, Bortfeld H, Diamond SG, Boas DA, Arvin K, Grant PE. Assessment of infant brain development with frequency‐domain near‐infrared spectroscopy. Pediatr Res 61: 546‐551, 2007. DOI: 10.1203/pdr.0b013e318045be99.
 144.Fraser CD 3rd, Brady KM, Rhee CJ, Easley RB, Kibler K, Smielewski P, Czosnyka M, Kaczka DW, Andropoulos DB, Rusin C. The frequency response of cerebral autoregulation. J Appl Physiol 115: 52‐56, 2013. DOI: 10.1152/japplphysiol.00068.2013.
 145.Freeman SS, Udomphorn Y, Armstead WM, Fisk DM, Vavilala MS. Young age as a risk factor for impaired cerebral autoregulation after moderate to severe pediatric traumatic brain injury. Anesthesiology 108: 588‐595, 2008. DOI: 10.1097/ALN.0b013e31816725d7.
 146.Fujimoto K. 'Medial defects' in the prenatal human cerebral arteries: an electron microscopic study. Stroke 27: 706‐708, 1996.
 147.Furuta A, Martin LJ. Laminar segregation of the cortical plate during corticogenesis is accompanied by changes in glutamate receptor expression. J Neurobiol 39: 67‐80, 1999.
 148.Galea E, Reis DJ, Xu H, Feinstein DL. Transient expression of calcium‐independent nitric oxide synthase in blood vessels during brain development. FASEB J 9: 1632‐1637, 1995.
 149.Garfunkel JM, Baird HW 3rd, Ziegler J. The relationship of oxygen consumption of cerebral functional activity. J Pediatr 44: 64‐72, 1954.
 150.Geary GG, Buchholz JN, Pearce WJ. Maturation depresses mouse cerebrovascular tone through endothelium‐dependent mechanisms. Am J Physiol Regul Integr Comp Physiol 284: R734‐R741, 2003. DOI: 10.1152/ajpregu.00510.2002.
 151.Gebremedhin D, Bonnet P, Greene AS, England SK, Rusch NJ, Lombard JH, Harder DR. Hypoxia increases the activity of Ca2+‐sensitive K+ channels in cat cerebral arterial muscle cell membranes. Pflugers Arch 428: 621‐630, 1994.
 152.Gebremedhin D, Lange AR, Lowry TF, Taheri MR, Birks EK, Hudetz AG, Narayanan J, Falck JR, Okamoto H, Roman RJ, Nithipatikom K, Campbell WB, Harder DR. Production of 20‐HETE and its role in autoregulation of cerebral blood flow. Circ Res 87: 60‐65, 2000.
 153.Gebremedhin D, Ma YH, Falck JR, Roman RJ, VanRollins M, Harder DR. Mechanism of action of cerebral epoxyeicosatrienoic acids on cerebral arterial smooth muscle. Am J Physiol Heart Circ Physiol 263: H519‐H525, 1992.
 154.Gebremedhin D, Yamaura K, Zhang C, Bylund J, Koehler RC, Harder DR. Metabotropic glutamate receptor activation enhances the activities of two types of Ca2+ ‐activated K+ channels in rat hippocampal astrocytes. J Neurosci 23: 1678‐1687, 2003.
 155.Gebremedhin D, Zhang DX, Carver KA, Rau N, Rarick KR, Roman RJ, Harder DR. Expression of CYP 4A omega‐hydroxylase and formation of 20‐hydroxyeicosatetreanoic acid (20‐HETE) in cultured rat brain astrocytes. Prostaglandins Other Lipid Mediat 124: 16‐26, 2016. DOI: 10.1016/j.prostaglandins.2016.04.003.
 156.Gennatas ED, Avants BB, Wolf DH, Satterthwaite TD, Ruparel K, Ciric R, Hakonarson H, Gur RE, Gur RC. Age‐related effects and sex differences in gray matter density, volume, mass, and cortical thickness from childhood to young adulthood. J Neurosci 37: 5065‐5073, 2017. DOI: 10.1523/JNEUROSCI.3550‐16.2017.
 157.Gerrits RJ, Stein EA, Greene AS. Anesthesia alters NO‐mediated functional hyperemia. Brain Res 907: 20‐26, 2001.
 158.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.
 159.Girouard H, Bonev AD, Hannah RM, Meredith A, Aldrich RW, Nelson MT. Astrocytic endfoot Ca2+ and BK channels determine both arteriolar dilation and constriction. Proc Natl Acad Sci U S A 107: 3811‐3816, 2010.
 160.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.
 161.Gleason CA, Hamm C, Jones MD Jr. Effect of acute hypoxemia on brain blood flow and oxygen metabolism in immature fetal sheep. Am J Physiol Heart Circ Physiol 258: H1064‐H1069, 1990.
 162.Gleason CA, Jones MD Jr, Traystman RJ, Notter RH. Fetal cerebral responses to ventilation and oxygenation in utero. Am J Physiol Regul Integr Comp Physiol 255: R1049‐R1054, 1988.
 163.Gollasch M, Wellman GC, Knot HJ, Jaggar JH, Damon DH, Bonev AD, Nelson MT. Ontogeny of local sarcoplasmic reticulum Ca2+ signals in cerebral arteries: Ca2+ sparks as elementary physiological events. Circ Res 83: 1104‐1114, 1998.
 164.Goplerud JM, Wagerle LC, Delivoria‐Papadopoulos M. Sympathetic nerve modulation of regional cerebral blood flow during asphyxia in newborn piglets. Am J Physiol Heart Circ Physiol 260: H1575‐H1580, 1991. DOI: 10.1152/ajpheart.1991.260.5.H1575.
 165.Gotoh J, Kuang TY, Nakao Y, Cohen DM, Melzer P, Itoh Y, Pak H, Pettigrew K, Sokoloff L. Regional differences in mechanisms of cerebral circulatory response to neuronal activation. Am J Physiol Heart Circ Physiol 280: H821‐H829, 2001.
 166.Gregoire NM, Gjedde A, Plum F, Duffy TE. Cerebral blood flow and cerebral metabolic rates for oxygen, glucose, and ketone bodies in newborn dogs. J Neurochem 30: 63‐69, 1978.
 167.Guiza F, Meyfroidt G, Lo TY, Jones PA, Van den Berghe G, Depreitere B. Continuous optimal CPP based on minute‐by‐minute monitoring data: A study of a pediatric population. Acta Neurochir Suppl 122: 187‐191, 2016. DOI: 10.1007/978‐3‐319‐22533‐3_38.
 168.Haggendal E, Lofgren J, Nilsson NJ, Zwetnow NN. Effects of varied cerebrospinal fluid pressure on cerebral blood flow in dogs. Acta Physiol Scand 79: 262‐271, 1970.
 169.Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA, O'Farrell FM, Buchan AM, Lauritzen M, Attwell D. Capillary pericytes regulate cerebral blood flow in health and disease. Nature 508: 55‐60, 2014. DOI: 10.1038/nature13165.
 170.Hansen NB, Brubakk AM, Bratlid D, Oh W, Stonestreet BS. The effects of variations in PaCO2 on brain blood flow and cardiac output in the newborn piglet. Pediatr Res 18: 1132‐1136, 1984. DOI: 10.1203/00006450‐198411000‐00015.
 171.Hansen NB, Nowicki PT, Miller RR, Malone T, Bickers RG, Menke JA. Alterations in cerebral blood flow and oxygen consumption during prolonged hypocarbia. Pediatr Res 20: 147‐150, 1986. DOI: 10.1203/00006450‐198602000‐00010.
 172.Harder DR, Abel PW, Hermsmeyer K. Membrane electrical mechanism of basilar artery constriction and pial artery dilation by norepinephrine. Circ Res 49: 1237‐1242, 1981.
 173.Harder DR, Gebremedhin D, Narayanan J, Jefcoat C, Falck JR, Campbell WB, Roman R. Formation and action of a p‐450 4A metabolite of arachidonic acid in cat cerebral microvessels. Am J Physiol Heart Circ Physiol 266: H2098‐H2107, 1994.
 174.Harder DR, Rarick KR, Gebremedhin D, Cohen SS. Regulation of cerebral blood flow: Response to cytochrome P450 lipid metabolites. Compr Physiol 8: 801‐821, 2018. DOI: 10.1002/c170025.
 175.Harris AP, Helou S, Gleason CA, Traystman RJ, Koehler RC. Fetal cerebral and peripheral circulatory responses to hypoxia after nitric oxide synthase inhibition. Am J Physiol Regul Integr Comp Physiol 281: R381‐R390, 2001.
 176.Harris AP, Koehler RC, Gleason CA, Jones MD Jr, Traystman RJ. Cerebral and peripheral circulatory responses to intracranial hypertension in fetal sheep. Circ Res 64: 991‐1000, 1989.
 177.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.
 178.Harris AP, Robinson R, Koehler RC, Traystman RJ, Gleason CA. Blood‐brain barrier permeability during dopamine‐induced hypertension in fetal sheep. J Appl Physiol 91: 123‐129, 2001.
 179.Harris JJ, Jolivet R, Attwell D. Synaptic energy use and supply. Neuron 75: 762‐777, 2012. DOI: 10.1016/j.neuron.2012.08.019.
 180.Hayashi S, Park MK, Kuehl TJ. Higher sensitivity of cerebral arteries isolated from premature and newborn baboons to adrenergic and cholinergic stimulation. Life Sci 35: 253‐260, 1984.
 181.Hayashi S, Park MK, Kuehl TJ. Relaxant and contractile responses to prostaglandins in premature, newborn and adult baboon cerebral arteries. J Pharmacol Exp Ther 233: 628‐635, 1985.
 182.Hekierski H, Pastor P, Curvello V, Armstead WM. Inhaled nitric oxide protects cerebral autoregulation and reduces hippocampal neuronal cell necrosis after traumatic brain injury in newborn and juvenile pigs. J Neurotrauma 36: 630‐638, 2019. DOI: 10.1089/neu.2018.5824.
 183.Helou SM, Hudak ML, Jones MD Jr. Cerebral blood flow response to hypercapnia in immature fetal sheep. Am J Physiol Heart Circ Physiol 261: H1366‐H1370, 1991.
 184.Helou SM, 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.
 185.Hernandez MJ, Brennan RW, Bowman GS. Autoregulation of cerebral blood flow in the newborn dog. Brain Res 184: 199‐202, 1980.
 186.Hernandez MJ, Brennan RW, Vannucci RC, Bowman GS. Cerebral blood flow and oxygen consumption in the newborn dog. Am J Physiol Regul Integr Comp Physiol 234: R209‐R215, 1978.
 187.Hernandez MJ, Vannucci RC, Salcedo A, Brennan RW. Cerebral blood flow and metabolism during hypoglycemia in newborn dogs. J Neurochem 35: 622‐628, 1980.
 188.Higashimori H, Blanco VM, Tuniki VR, Falck JR, Filosa JA. Role of epoxyeicosatrienoic acids as autocrine metabolites in glutamate‐mediated K+ signaling in perivascular astrocytes. Am J Physiol Cell Physiol 299: C1068‐C1078, 2010.
 189.Hill RA, Tong L, Yuan P, Murikinati S, Gupta S, Grutzendler J. Regional blood flow in the normal and ischemic brain is controlled by arteriolar smooth muscle cell contractility and not by capillary pericytes. Neuron 87: 95‐110, 2015. DOI: 10.1016/j.neuron.2015.06.001.
 190.Himwich HE, Fazekas JF. Comparative studies of the metabolism of the brain of infant and adult dogs. Am J Phys 132: 454‐459, 1941.
 191.Hohimer AR, Bissonnette JM. Effects of cephalic hypotension, hypertension, and barbiturates on fetal cerebral flood flow and metabolism. Am J Obstet Gynecol 161: 1344‐1351, 1989.
 192.Hoiland RL, Bain AR, Rieger MG, Bailey DM, Ainslie PN. Hypoxemia, oxygen content, and the regulation of cerebral blood flow. Am J Physiol Regul Integr Comp Physiol 310: R398‐R413, 2016. DOI: 10.1152/ajpregu.00270.2015.
 193.Hoiland RL, Caldwell HG, Howe CA, Nowak‐Fluck D, Stacey BS, Bailey DM, Paton JFR, Green DJ, Sekhon MS, Macleod DB, Ainslie PN. Nitric oxide is fundamental to neurovascular coupling in humans. J Physiol 598: 4927‐4939, 2020. DOI: 10.1113/JP280162.
 194.Hoiland RL, Fisher JA, Ainslie PN. Regulation of the cerebral circulation by arterial carbon dioxide. Compr Physiol 9: 1101‐1154, 2019. DOI: 10.1002/c180021.
 195.Holt DC, Fedinec AL, Vaughn AN, Leffler CW. Age and species dependence of pial arteriolar responses to topical carbon monoxide in vivo. Exp Biol Med (Maywood) 232: 1465‐1469, 2007.
 196.Hovda DA, Chugani HT, Villablanca JR, Badie B, Sutton RL. Maturation of cerebral oxidative metabolism in the cat: A cytochrome oxidase histochemistry study. J Cereb Blood Flow Metab 12: 1039‐1048, 1992. DOI: 10.1038/jcbfm.1992.141.
 197.Hovda DA, Villablanca JR, Chugani HT, Barrio JR. Metabolic maturation of the brain: A study of local cerebral protein synthesis in the developing cat. Brain Res 1113: 54‐63, 2006. DOI: 10.1016/j.brainres.2006.07.083.
 198.Howarth C, Gleeson P, Attwell D. Updated energy budgets for neural computation in the neocortex and cerebellum. J Cereb Blood Flow Metab 32: 1222‐1232, 2012. DOI: 10.1038/jcbfm.2012.35.
 199.Hsu P, Albuquerque ML, Leffler CW. Mechanisms of hypercapnia‐stimulated PG production in piglet cerebral microvascular endothelial cells. Am J Physiol Heart Circ Physiol 268: H591‐H603, 1995. DOI: 10.1152/ajpheart.1995.268.2.H591.
 200.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. DOI: 10.1152/ajpheart.1993.264.5.H1485.
 201.Hudak ML, Koehler RC, Rosenberg AA, Traystman RJ, Jones MD Jr. Effect of hematocrit on cerebral blood flow. Am J Physiol Heart Circ Physiol 251: H63‐H70, 1986.
 202.Hudak ML, Tang YL, Massik J, Koehler RC, Traystman RJ, Jones MD Jr. Base‐line O2 extraction influences cerebral blood flow response to hematocrit. Am J Physiol Heart Circ Physiol 254: H156‐H162, 1988.
 203.Hunter CJ, Blood AB, White CR, Pearce WJ, Power GG. Role of nitric oxide in hypoxic cerebral vasodilatation in the ovine fetus. J Physiol 549: 625‐633, 2003. DOI: 10.1113/jphysiol.2002.038034.
 204.Ioroi T, Yonetani M, Nakamura H. Effects of hypoxia and reoxygenation on nitric oxide production and cerebral blood flow in developing rat striatum. Pediatr Res 43: 733‐737, 1998.
 205.Ishikawa M, Kajimura M, Adachi T, Maruyama K, Makino N, Goda N, Yamaguchi T, Sekizuka E, Suematsu M. Carbon monoxide from heme oxygenase‐2 is a tonic regulator against NO‐dependent vasodilatation in the adult rat cerebral microcirculation. Circ Res 97: e104‐e114, 2005. DOI: 10.1161/01.RES.0000196681.34485.ec.
 206.Iwamoto HS, Kaufman T, Keil LC, Rudolph AM. Responses to acute hypoxemia in fetal sheep at 0.6‐0.7 gestation. Am J Physiol Heart Circ Physiol 256: H613‐H620, 1989.
 207.Jacobs B, Chugani HT, Allada V, Chen S, Phelps ME, Pollack DB, Raleigh MJ. Developmental changes in brain metabolism in sedated rhesus macaques and vervet monkeys revealed by positron emission tomography. Cereb Cortex 5: 222‐233, 1995.
 208.Jaggar JH, Leffler CW, Cheranov SY, Tcheranova D, Shuyu E, Cheng X. Carbon monoxide dilates cerebral arterioles by enhancing the coupling of Ca2+ sparks to Ca2+‐activated K+ channels. Circ Res 91: 610‐617, 2002.
 209.Jaggar JH, Li A, Parfenova H, Liu J, Umstot ES, Dopico AM, Leffler CW. Heme is a carbon monoxide receptor for large‐conductance Ca2+‐activated K+ channels. Circ Res 97: 805‐812, 2005.
 210.Jones M Jr, Sheldon RE, Peeters LL, Meschia G, Battaglia FC, Makowski EL. Fetal cerebral oxygen consumption at different levels of oxygenation. J Appl Physiol 43: 1080‐1084, 1977. DOI: 10.1152/jappl.1977.43.6.1080.
 211.Jones MD Jr, Sheldon RE, Peeters LL, Makowski EL, Meschia G. Regulation of cerebral blood flow in the ovine fetus. Am J Physiol Heart Circ Physiol 235: H162‐H166, 1978.
 212.Jones MD Jr, Traystman RJ, Simmons MA, Molteni RA. Effects of changes in arterial O2 content on cerebral blood flow in the lamb. Am J Physiol Heart Circ Physiol 240: H209‐H215, 1981.
 213.Joshi B, Ono M, Brown C, Brady K, Easley RB, Yenokyan G, Gottesman RF, Hogue CW. Predicting the limits of cerebral autoregulation during cardiopulmonary bypass. Anesth Analg 114: 503‐510, 2012. DOI: 10.1213/ANE.0b013e31823d292a.
 214.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.
 215.Kanu A, Leffler CW. Roles of glia limitans astrocytes and carbon monoxide in adenosine diphosphate‐induced pial arteriolar dilation in newborn pigs. Stroke 40: 930‐935, 2009.
 216.Kanu A, Leffler CW. Arachidonic acid‐ and prostaglandin E2‐induced cerebral vasodilation is mediated by carbon monoxide, independent of reactive oxygen species in piglets. Am J Physiol Heart Circ Physiol 301: H2482‐H2487, 2011.
 217.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.
 218.Karen T, Morren G, Haensse D, Bauschatz AS, Bucher HU, Wolf M. Hemodynamic response to visual stimulation in newborn infants using functional near‐infrared spectroscopy. Hum Brain Mapp 29: 453‐460, 2008. DOI: 10.1002/hbm.20411.
 219.Keep RF, Ennis SR, Beer ME, Betz AL. Developmental changes in blood‐brain barrier potassium permeability in the rat: Relation to brain growth. J Physiol 488 (Pt 2): 439‐448, 1995.
 220.Kennedy C, Grave GD, Juhle JW, Sokoloff L. Changes in blood flow in the component structures of the dog brain during postnatal maturation. J Neurochem 19: 2423‐2433, 1972.
 221.Kennedy C, Sokoloff L. An adaptation of the nitrous oxide method to the study of the cerebral circulation in children; normal values for cerebral blood flow and cerebral metabolic rate in childhood. J Clin Invest 36: 1130‐1137, 1957. DOI: 10.1172/JCI103509.
 222.Kety SS, Schmidt CF. The nitrous nxide method for the quantitative determination of cerebral bloodflow in man: Theory, procedure and normal values. J Clin Invest 27: 476‐483, 1948. DOI: 10.1172/JCI101994.
 223.Khennouf L, Gesslein B, Brazhe A, Octeau JC, Kutuzov N, Khakh BS, Lauritzen M. Active role of capillary pericytes during stimulation‐induced activity and spreading depolarization. Brain 141: 2032‐2046, 2018. DOI: 10.1093/brain/awy143.
 224.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. DOI: 10.1038/sj.jcbfm.9600046.
 225.Knecht KR, Milam S, Wilkinson DA, Fedinec AL, Leffler CW. Time‐dependent action of carbon monoxide on the newborn cerebrovascular circulation. Am J Physiol Heart Circ Physiol 299: H70‐H75, 2010.
 226.Kocharyan A, Fernandes P, Tong XK, Vaucher E, Hamel E. Specific subtypes of cortical GABA interneurons contribute to the neurovascular coupling response to basal forebrain stimulation. J Cereb Blood Flow Metab 28: 221‐231, 2008. DOI: 10.1038/sj.jcbfm.9600558.
 227.Koehler RC, Jones MD Jr, Traystman RJ. Cerebral circulatory response to carbon monoxide and hypoxic hypoxia in the lamb. Am J Physiol Heart Circ Physiol 243: H27‐H32, 1982.
 228.Koehler RC, Traystman RJ, Jones MD Jr. Influence of reduced oxygemoglobin affinity on cerebrovascular response to hypoxic hypoxia. Am J Physiol Heart Circ Physiol 251: H756‐H763, 1986.
 229.Koehler RC, Traystman RJ, Rosenberg AA, Hudak ML, Jones MD Jr. Role of O2‐hemoglobin affinity on cerebrovascular response to carbon monoxide hypoxia. Am J Physiol Heart Circ Physiol 245: H1019‐H1023, 1983.
 230.Koehler RC, Traystman RJ, Zeger S, Rogers MC, Jones MD Jr. Comparison of cerebrovascular response to hypoxic and carbon monoxide hypoxia in newborn and adult sheep. J Cereb Blood Flow Metab 4: 115‐122, 1984.
 231.Koneru P, Leffler CW. Role of cGMP in carbon monoxide‐induced cerebral vasodilation in piglets. Am J Physiol Heart Circ Physiol 286: H304‐H309, 2004.
 232.Kontos HA, Wei EP, Navari RM, Levasseur JE, Rosenblum WI, Patterson JL Jr. Responses of cerebral arteries and arterioles to acute hypotension and hypertension. Am J Physiol Heart Circ Physiol 234: H371‐H383, 1978.
 233.Kosty J, Riley J, Liang J, Armstead WM. Influence of sex and ERK MAPK on the pressure reactivity index in newborn piglets after fluid percussion injury. Transl Stroke Res 3: 460‐465, 2012. DOI: 10.1007/s12975‐012‐0196‐3.
 234.Kozberg M, Hillman E. Neurovascular coupling and energy metabolism in the developing brain. Prog Brain Res 225: 213‐242, 2016. DOI: 10.1016/bs.pbr.2016.02.002.
 235.Kozberg MG, Chen BR, DeLeo SE, Bouchard MB, Hillman EM. Resolving the transition from negative to positive blood oxygen level‐dependent responses in the developing brain. Proc Natl Acad Sci U S A 110: 4380‐4385, 2013. DOI: 10.1073/pnas.1212785110.
 236.Kozberg MG, Ma Y, Shaik MA, Kim SH, Hillman EM. Rapid postnatal expansion of neural networks occurs in an environment of altered neurovascular and neurometabolic coupling. J Neurosci 36: 6704‐6717, 2016. DOI: 10.1523/JNEUROSCI.2363‐15.2016.
 237.Kratzer I, Chip S, Vexler ZS. Barrier mechanisms in neonatal stroke. Front Neurosci 8: 359, 2014. DOI: 10.3389/fnins.2014.00359.
 238.Kurth CD, Wagerle LC. Cerebrovascular reactivity to adenosine analogues in 0.6‐0.7 gestation and near‐term fetal sheep. Am J Physiol Heart Circ Physiol 262: H1338‐H1342, 1992.
 239.Kurth CD, Wagerle LC, Delivoria‐Papadopoulos M. Sympathetic regulation of cerebral blood flow during seizures in newborn lambs. Am J Physiol Heart Circ Physiol 255: H563‐H568, 1988. DOI: 10.1152/ajpheart.1988.255.3.H563.
 240.Kusaka T, Kawada K, Okubo K, Nagano K, Namba M, Okada H, Imai T, Isobe K, Itoh S. Noninvasive optical imaging in the visual cortex in young infants. Hum Brain Mapp 22: 122‐132, 2004. DOI: 10.1002/hbm.20020.
 241.Kuzawa CW, Chugani HT, Grossman LI, Lipovich L, Muzik O, Hof PR, Wildman DE, Sherwood CC, Leonard WR, Lange N. Metabolic costs and evolutionary implications of human brain development. Proc Natl Acad Sci U S A 111: 13010‐13015, 2014. DOI: 10.1073/pnas.1323099111.
 242.Lacoste B, Comin CH, Ben‐Zvi A, Kaeser PS, Xu X, Costa Lda F, Gu C. Sensory‐related neural activity regulates the structure of vascular networks in the cerebral cortex. Neuron 83: 1117‐1130, 2014. DOI: 10.1016/j.neuron.2014.07.034.
 243.Lacroix A, Toussay X, Anenberg E, Lecrux C, Ferreiros N, Karagiannis A, Plaisier F, Chausson P, Jarlier F, Burgess SA, Hillman EM, Tegeder I, Murphy TH, Hamel E, Cauli B. COX‐2‐derived prostaglandin E2 produced by pyramidal neurons contributes to neurovascular coupling in the rodent cerebral cortex. J Neurosci 35: 11791‐11810, 2015. DOI: 10.1523/JNEUROSCI.0651‐15.2015.
 244.Laptook AR, Stonestreet BS, Oh W. Brain blood flow and O2 delivery during hemorrhagic hypotension in the piglet. Pediatr Res 17: 77‐80, 1983.
 245.Larson AC, Jamrogowicz JL, Kulikowicz E, Wang B, Yang ZJ, Shaffner DH, Koehler RC, Lee JK. Cerebrovascular autoregulation after rewarming from hypothermia in a neonatal swine model of asphyxic brain injury. J Appl Physiol 115: 1433‐1442, 2013. DOI: 10.1152/japplphysiol.00238.2013.
 246.Laudignon N, Farri E, Beharry K, Rex J, Aranda JV. Influence of adenosine on cerebral blood flow during hypoxic hypoxia in the newborn piglet. J Appl Physiol 68: 1534‐1541, 1990.
 247.Lecrux C, Toussay X, Kocharyan A, Fernandes P, Neupane S, Levesque M, Plaisier F, Shmuel A, Cauli B, Hamel E. Pyramidal neurons are "neurogenic hubs" in the neurovascular coupling response to whisker stimulation. J Neurosci 31: 9836‐9847, 2011.
 248.Lee JK, Brady KM, Chung SE, Jennings JM, Whitaker EE, Aganga D, Easley RB, Heitmiller K, Jamrogowicz JL, Larson AC, Lee JH, Jordan LC, Hogue CW, Lehmann CU, Bembea MM, Hunt EA, Koehler RC, Shaffner DH. A pilot study of cerebrovascular reactivity autoregulation after pediatric cardiac arrest. Resuscitation 85: 1387‐1393, 2014. DOI: 10.1016/j.resuscitation.2014.07.006.
 249.Lee JK, Brady KM, Mytar JO, Kibler KK, Carter EL, Hirsch KG, Hogue CW, Easley RB, Jordan LC, Smielewski P, Czosnyka M, Shaffner DH, Koehler RC. Cerebral blood flow and cerebrovascular autoregulation in a swine model of pediatric cardiac arrest and hypothermia. Crit Care Med 39: 2337‐2345, 2011.
 250.Lee JK, Kibler KK, Benni PB, Easley RB, Czosnyka M, Smielewski P, Koehler RC, Shaffner DH, Brady KM. Cerebrovascular reactivity measured by near‐infrared spectroscopy. Stroke 40: 1820‐1826, 2009.
 251.Lee JK, Poretti A, Perin J, Huisman T, Parkinson C, Chavez‐Valdez R, O'Connor M, Reyes M, Armstrong J, Jennings JM, Gilmore MM, Koehler RC, Northington FJ, Tekes A. Optimizing cerebral autoregulation may decrease neonatal regional hypoxic‐ischemic brain injury. Dev Neurosci 39: 248‐256, 2017. DOI: 10.1159/000452833.
 252.Lee JK, Yang ZJ, Wang B, Larson AC, Jamrogowicz JL, Kulikowicz E, Kibler KK, Mytar JO, Carter EL, Burman HT, Brady KM, Smielewski P, Czosnyka M, Koehler RC, Shaffner DH. Noninvasive autoregulation monitoring in a swine model of pediatric cardiac arrest. Anesth Analg 114: 825‐836, 2012.
 253.Lee TJ, Kinkead LR, Sarwinski S. Norepinephrine and acetylcholine transmitter mechanisms in large cerebral arteries of the pig. J Cereb Blood Flow Metab 2: 439‐450, 1982. DOI: 10.1038/jcbfm.1982.50.
 254.Leffler CW, Balabanova L, Fedinec AL, Parfenova H. Nitric oxide increases carbon monoxide production by piglet cerebral microvessels. Am J Physiol Heart Circ Physiol 289: H1442‐H1447, 2005.
 255.Leffler CW, Balabanova L, Fedinec AL, Waters CM, Parfenova H. Mechanism of glutamate stimulation of CO production in cerebral microvessels. Am J Physiol Heart Circ Physiol 285: H74‐H80, 2003.
 256.Leffler CW, Balabanova L, Sullivan D, Wang X, Fedinec AL, Parfenova H. Regulation of CO production in cerebral microvessels of newborn pigs. Am J Physiol Heart Circ Physiol 285: H292‐H297, 2003.
 257.Leffler CW, Balabanova L, Williams KK. cAMP production by piglet cerebral vascular smooth muscle cells: pH(o), pH(i), and permissive action of PGI(2). Am J Physiol Heart Circ Physiol 277: H1878‐H1883, 1999.
 258.Leffler CW, Beasley DG, Busija DW. Cerebral ischemia alters cerebral microvascular reactivity in newborn pigs. Am J Physiol Heart Circ Physiol 257: H266‐H271, 1989.
 259.Leffler CW, Busija DW. Prostanoids and pial arteriolar diameter in hypotensive newborn pigs. Am J Physiol Heart Circ Physiol 252: H687‐H691, 1987.
 260.Leffler CW, Busija DW, Armstead WM, Mirro R, Beasley DG. Ischemia alters cerebral vascular responses to hypercapnia and acetylcholine in piglets. Pediatr Res 25: 180‐183, 1989.
 261.Leffler CW, Busija DW, Beasley DG, Armstead WM, Mirro R. Postischemic cerebral microvascular responses to norepinephrine and hypotension in newborn pigs. Stroke 20: 541‐546, 1989.
 262.Leffler CW, Busija DW, Beasley DG, Fletcher AM. Maintenance of cerebral circulation during hemorrhagic hypotension in newborn pigs: Role of prostanoids. Circ Res 59: 562‐567, 1986.
 263.Leffler CW, Busija DW, Mirro R, Armstead WM, Beasley DG. Effects of ischemia on brain blood flow and oxygen consumption of newborn pigs. Am J Physiol Heart Circ Physiol 257: H1917‐H1926, 1989.
 264.Leffler CW, Fedinec AL. Newborn piglet cerebral microvascular responses to epoxyeicosatrienoic acids. Am J Physiol Heart Circ Physiol 273: H333‐H338, 1997. DOI: 10.1152/ajpheart.1997.273.1.H333.
 265.Leffler CW, Fedinec AL, Shibata M. Prostacyclin receptor activation and pial arteriolar dilation after endothelial injury in piglets. Stroke 26: 2103‐2110; discussion 2110‐2111, 1995.
 266.Leffler CW, Mirro R, Armstead WM, Busija DW, Thelin O. Prostanoid synthesis and vascular responses to exogenous arachidonic acid following cerebral ischemia in piglets. Prostaglandins 40: 241‐248, 1990.
 267.Leffler CW, Mirro R, Armstead WM, Shibata M. Topical arachidonic acid restores pial arteriolar dilation to hypercapnia of postischemic newborn pig brain. Am J Physiol Heart Circ Physiol 263: H746‐H751, 1992.
 268.Leffler CW, Mirro R, Pharris LJ, Shibata M. Permissive role of prostacyclin in cerebral vasodilation to hypercapnia in newborn pigs. Am J Physiol Heart Circ Physiol 267: H285‐H291, 1994.
 269.Leffler CW, Mirro R, Shanklin DR, Armstead WM, Shibata M. Light/dye microvascular injury selectively eliminates hypercapnia‐induced pial arteriolar dilation in newborn pigs. Am J Physiol Heart Circ Physiol 266: H623‐H630, 1994. DOI: 10.1152/ajpheart.1994.266.2.H623.
 270.Leffler CW, Mirro R, Shibata M, Parfenova H, Armstead WM, Zuckerman S. Effects of indomethacin on cerebral vasodilator responses to arachidonic acid and hypercapnia in newborn pigs. Pediatr Res 33: 609‐614, 1993. DOI: 10.1203/00006450‐199306000‐00016.
 271.Leffler CW, Nasjletti A, Johnson RA, Fedinec AL. Contributions of prostacyclin and nitric oxide to carbon monoxide‐induced cerebrovascular dilation in piglets. Am J Physiol Heart Circ Physiol 280: H1490‐H1495, 2001.
 272.Leffler CW, Nasjletti A, Yu C, Johnson RA, Fedinec AL, Walker N. Carbon monoxide and cerebral microvascular tone in newborn pigs. Am J Physiol Heart Circ Physiol 276: H1641‐H1646, 1999.
 273.Leffler CW, Parfenova H. Cerebral arteriolar dilation to hypoxia: Role of prostanoids. Am J Physiol Heart Circ Physiol 272: H418‐H424, 1997. DOI: 10.1152/ajpheart.1997.272.1.H418.
 274.Leffler CW, Parfenova H, Basuroy S, Jaggar JH, Umstot ES, Fedinec AL. Hydrogen sulfide and cerebral microvascular tone in newborn pigs. Am J Physiol Heart Circ Physiol 300: H440‐H447, 2011.
 275.Leffler CW, Parfenova H, Fedinec AL, Basuroy S, Tcheranova D. Contributions of astrocytes and CO to pial arteriolar dilation to glutamate in newborn pigs. Am J Physiol Heart Circ Physiol 291: H2897‐H2904, 2006.
 276.Leffler CW, Parfenova H, Jaggar JH. Carbon monoxide as an endogenous vascular modulator. Am J Physiol Heart Circ Physiol 301: H1‐H11, 2011.
 277.Leffler CW, Smith JS, Edrington JL, Zuckerman SL, Parfenova H. Mechanisms of hypoxia‐induced cerebrovascular dilation in the newborn pig. Am J Physiol Heart Circ Physiol 272: H1323‐H1332, 1997. DOI: 10.1152/ajpheart.1997.272.3.H1323.
 278.Lewis PM, Czosnyka M, Carter BG, Rosenfeld JV, Paul E, Singhal N, Butt W. Cerebrovascular pressure reactivity in children with traumatic brain injury. Pediatr Crit Care Med 16: 739‐749, 2015. DOI: 10.1097/PCC.0000000000000471.
 279.Li A, Adebiyi A, Leffler CW, Jaggar JH. KCa channel insensitivity to Ca2+ sparks underlies fractional uncoupling in newborn cerebral artery smooth muscle cells. Am J Physiol Heart Circ Physiol 291: H1118‐H1125, 2006.
 280.Li A, Xi Q, Umstot ES, Bellner L, Schwartzman ML, Jaggar JH, Leffler CW. Astrocyte‐derived CO is a diffusible messenger that mediates glutamate‐induced cerebral arteriolar dilation by activating smooth muscle cell KCa channels. Circ Res 102: 234‐241, 2008.
 281.Liang GH, Adebiyi A, Leo MD, McNally EM, Leffler CW, Jaggar JH. Hydrogen sulfide dilates cerebral arterioles by activating smooth muscle cell plasma membrane KATP channels. Am J Physiol Heart Circ Physiol 300: H2088‐H2095, 2011.
 282.Liang GH, Xi Q, Leffler CW, Jaggar JH. Hydrogen sulfide activates Ca(2)(+) sparks to induce cerebral arteriole dilatation. J Physiol 590: 2709‐2720, 2012.
 283.Liao SM, Gregg NM, White BR, Zeff BW, Bjerkaas KA, Inder TE, Culver JP. Neonatal hemodynamic response to visual cortex activity: High‐density near‐infrared spectroscopy study. J Biomed Opt 15: 026010, 2010. DOI: 10.1117/1.3369809.
 284.Lin MT, Hessinger DA, Pearce WJ, Longo LD. Developmental differences in Ca2+‐activated K+ channel activity in ovine basilar artery. Am J Physiol Heart Circ Physiol 285: H701‐H709, 2003. DOI: 10.1152/ajpheart.00138.2003.
 285.Lin MT, Longo LD, Pearce WJ, Hessinger DA. Ca2+‐activated K+ channel‐associated phosphatase and kinase activities during development. Am J Physiol Heart Circ Physiol 289: H414‐H425, 2005. DOI: 10.1152/ajpheart.01079.2004.
 286.Lin PY, Roche‐Labarbe N, Dehaes M, Fenoglio A, Grant PE, Franceschini MA. Regional and hemispheric asymmetries of cerebral hemodynamic and oxygen metabolism in newborns. Cereb Cortex 23: 339‐348, 2013. DOI: 10.1093/cercor/bhs023.
 287.Lind BL, Brazhe AR, Jessen SB, Tan FC, Lauritzen MJ. Rapid stimulus‐evoked astrocyte Ca2+ elevations and hemodynamic responses in mouse somatosensory cortex in vivo. Proc Natl Acad Sci U S A 110: E4678‐E4687, 2013. DOI: 10.1073/pnas.1310065110.
 288.Lind BL, Jessen SB, Lonstrup M, Josephine C, Bonvento G, Lauritzen M. Fast Ca(2+) responses in astrocyte end‐feet and neurovascular coupling in mice. Glia 66: 348‐358, 2018. DOI: 10.1002/glia.23246.
 289.Lindauer U, Megow D, Matsuda H, Dirnagl U. Nitric oxide: A modulator, but not a mediator, of neurovascular coupling in rat somatosensory cortex. Am J Physiol Heart Circ Physiol 277: H799‐H811, 1999.
 290.Liu P, Huang H, Rollins N, Chalak LF, Jeon T, Halovanic C, Lu H. Quantitative assessment of global cerebral metabolic rate of oxygen (CMRO2) in neonates using MRI. NMR Biomed 27: 332‐340, 2014. DOI: 10.1002/nbm.3067.
 291.Liu X, Gebremedhin D, Harder DR, Koehler RC. Contribution of epoxyeicosatrienoic acids to the cerebral blood flow response to hypoxemia. J Appl Physiol 119: 1202‐1209, 2015. DOI: 10.1152/japplphysiol.01043.2014.
 292.Liu X, Li C, Falck JR, Harder DR, Koehler RC. Relative contribution of cyclooxygenases, epoxyeicosatrienoic acids, and pH to the cerebral blood flow response to vibrissal stimulation. Am J Physiol Heart Circ Physiol 302: H1075‐H1085, 2012.
 293.Liu X, Li C, Falck JR, Roman RJ, Harder DR, Koehler RC. Interaction of nitric oxide, 20‐HETE, and EETs during functional hyperemia in whisker barrel cortex. Am J Physiol Heart Circ Physiol 295: H619‐H631, 2008.
 294.Liu X, Li C, Gebremedhin D, Hwang SH, Hammock BD, Falck JR, Roman RJ, Harder DR, Koehler RC. Epoxyeicosatrienoic acid‐dependent cerebral vasodilation evoked by metabotropic glutamate receptor activation in vivo. Am J Physiol Heart Circ Physiol 301: H373‐H381, 2011.
 295.Lizasoain I, Weiner CP, Knowles RG, Moncada S. The ontogeny of cerebral and cerebellar nitric oxide synthase in the guinea pig and rat. Pediatr Res 39: 779‐783, 1996. DOI: 10.1203/00006450‐199605000‐00006.
 296.Logsdon AF, Lucke‐Wold BP, Turner RC, Huber JD, Rosen CL, Simpkins JW. Role of microvascular disruption in brain damage from traumatic brain injury. Compr Physiol 5: 1147‐1160, 2015. DOI: 10.1002/c140057.
 297.Lohn M, Lauterbach B, Haller H, Pongs O, Luft FC, Gollasch M. Beta(1)‐subunit of BK channels regulates arterial wall[Ca(2+)] and diameter in mouse cerebral arteries. J Appl Physiol 91: 1350‐1354, 2001. DOI: 10.1152/jappl.2001.91.3.1350.
 298.Longden TA, Dunn KM, Draheim HJ, Nelson MT, Weston AH, Edwards G. Intermediate‐conductance calcium‐activated potassium channels participate in neurovascular coupling. Br J Pharmacol 164: 922‐933, 2011. DOI: 10.1111/j.1476‐5381.2011.01447.x.
 299.Longo LD, Ueno N, Zhao Y, Pearce WJ, Zhang L. Developmental changes in alpha 1‐adrenergic receptors, IP3 responses, and NE‐induced contraction in cerebral arteries. Am J Physiol Heart Circ Physiol 271: H2313‐H2319, 1996. DOI: 10.1152/ajpheart.1996.271.6.H2313.
 300.Lu H, Xu F, Rodrigue KM, Kennedy KM, Cheng Y, Flicker B, Hebrank AC, Uh J, Park DC. Alterations in cerebral metabolic rate and blood supply across the adult lifespan. Cereb Cortex 21: 1426‐1434, 2011. DOI: 10.1093/cercor/bhq224.
 301.Ma J, Ayata C, Huang PL, Fishman MC, Moskowitz MA. Regional cerebral blood flow response to vibrissal stimulation in mice lacking type I NOS gene expression. Am J Physiol Heart Circ Physiol 270: H1085‐H1090, 1996.
 302.Martin E, Joeri P, Loenneker T, Ekatodramis D, Vitacco D, Hennig J, Marcar VL. Visual processing in infants and children studied using functional MRI. Pediatr Res 46: 135‐140, 1999.
 303.Martin LJ, Brambrink A, Koehler RC, Traystman RJ. Neonatal asphyxic brain injury is neural system preferential and targets sensory‐motor networks. In: Stevenson DK, Sunshine P, editors. Fetal and Neonatal Brain Injury: Mechanisms, Management and the Risks of Practice. Oxford: Oxford University Press, 1997, p. 374‐399.
 304.Martin LJ, Brambrink A, Koehler RC, Traystman RJ. Primary sensory and forebrain motor systems in the newborn brain are preferentially damaged by hypoxia‐ischemia. J Comp Neurol 377: 262‐285, 1997.
 305.Martin LJ, Brambrink AM, Price AC, Kaiser A, Agnew DM, Ichord RN, Traystman RJ. Neuronal death in newborn striatum after hypoxia‐ischemia is necrosis and evolves with oxidative stress. Neurobiol Dis 7: 169‐191, 2000.
 306.Martinez‐Orgado J, Gonzalez R, Alonso MJ, Rodriguez‐Martinez MA, Sanchez‐Ferrer CF, Marin J. Endothelial factors and autoregulation during pressure changes in isolated newborn piglet cerebral arteries. Pediatr Res 44: 161‐167, 1998. DOI: 10.1203/00006450‐199808000‐00004.
 307.Massaro AN, Govindan RB, Vezina G, Chang T, Andescavage NN, Wang Y, Al‐Shargabi T, Metzler M, Harris K, du Plessis AJ. Impaired cerebral autoregulation and brain injury in newborns with hypoxic‐ischemic encephalopathy treated with hypothermia. J Neurophysiol 114: 818‐824, 2015. DOI: 10.1152/jn.00353.2015.
 308.Massik J, Tang YL, Hudak ML, Koehler RC, Traystman RJ, Jones MD Jr. Effect of hematocrit on cerebral blood flow with induced polycythemia. J Appl Physiol 62: 1090‐1096, 1987.
 309.McAdams RM, Fleiss B, Traudt C, Schwendimann L, Snyder JM, Haynes RL, Natarajan N, Gressens P, Juul SE. Long‐term neuropathological changes associated with cerebral palsy in a nonhuman primate model of hypoxic‐ischemic encephalopathy. Dev Neurosci 39: 124‐140, 2017. DOI: 10.1159/000470903.
 310.McCrabb GJ, Harding R. Role of nitric oxide in the regulation of cerebral blood flow in the ovine foetus. Clin Exp Pharmacol Physiol 23: 855‐860, 1996.
 311.McCrabb GJ, Harding R. Cerebral oxygen delivery is reduced during the acidaemia associated with prolonged hypoxaemia in the immature ovine fetus. Biol Neonate 71: 385‐394, 1997. DOI: 10.1159/000244440.
 312.McKenna MC, Scafidi S, Robertson CL. Metabolic alterations in developing brain after injury: Knowns and unknowns. Neurochem Res 40: 2527‐2543, 2015. DOI: 10.1007/s11064‐015‐1600‐7.
 313.McPherson RW, Koehler RC, Traystman RJ. Effect of jugular venous pressure on cerebral autoregulation in dogs. Am J Physiol Heart Circ Physiol 255: H1516‐H1524, 1988.
 314.Meno JR, Crum AV, Winn HR. Effect of adenosine receptor blockade on pial arteriolar dilation during sciatic nerve stimulation. Am J Physiol Heart Circ Physiol 281: H2018‐H2027, 2001.
 315.Miekisiak G, Kulik T, Kusano Y, Kung D, Chen JF, Winn HR. Cerebral blood flow response in adenosine 2a receptor knockout mice during transient hypoxic hypoxia. J Cereb Blood Flow Metab 28: 1656‐1664, 2008.
 316.Miller SP, Ramaswamy V, Michelson D, Barkovich AJ, Holshouser B, Wycliffe N, Glidden DV, Deming D, Partridge JC, Wu YW, Ashwal S, Ferriero DM. Patterns of brain injury in term neonatal encephalopathy. J Pediatr 146: 453‐460, 2005.
 317.Mirro R, Pharris LJ, Armstead WM, Shibata M, Leffler CW. Effects of indomethacin on newborn pig pial arteriolar responses to PCO2. J Appl Physiol 75: 1300‐1305, 1993. DOI: 10.1152/jappl.1993.75.3.1300.
 318.Mishra A, Reynolds JP, Chen Y, Gourine AV, Rusakov DA, Attwell D. Astrocytes mediate neurovascular signaling to capillary pericytes but not to arterioles. Nat Neurosci 19: 1619‐1627, 2016. DOI: 10.1038/nn.4428.
 319.Mollgard K, Saunders NR. The development of the human blood‐brain and blood‐CSF barriers. Neuropathol Appl Neurobiol 12: 337‐358, 1986.
 320.Montecot C, Seylaz J, Pinard E. Carbon monoxide regulates cerebral blood flow in epileptic seizures but not in hypercapnia. Neuroreport 9: 2341‐2346, 1998.
 321.Moore LE, Kirsch JR, Helfaer MA, Greenberg RS, Traystman RJ. Hypercapnic blood flow reactivity not increased by alpha‐blockade or cordotomy in piglets. Am J Physiol Heart Circ Physiol 262: H1884‐H1890, 1992.
 322.Moore TJ, Lione AP, Sugden MC, Regen DM. Beta‐hydroxybutyrate transport in rat brain: Developmental and dietary modulations. Am J Phys 230: 619‐630, 1976. DOI: 10.1152/ajplegacy.1976.230.3.619.
 323.Muizelaar JP, Ward JD, Marmarou A, Newlon PG, Wachi A. Cerebral blood flow and metabolism in severely head‐injured children. Part 2: Autoregulation. J Neurosurg 71: 72‐76, 1989.
 324.Muramoto S, Yamada H, Sadato N, Kimura H, Konishi Y, Kimura K, Tanaka M, Kochiyama T, Yonekura Y, Ito H. Age‐dependent change in metabolic response to photic stimulation of the primary visual cortex in infants: Functional magnetic resonance imaging study. J Comput Assist Tomogr 26: 894‐901, 2002.
 325.Myers RE. Two patterns of perinatal brain damage and their conditions of occurrence. Am J Obstet Gynecol 112: 246‐276, 1972.
 326.Nagel C, Diedler J, Gerbig I, Heimberg E, Schuhmann MU, Hockel K. State of cerebrovascular autoregulation correlates with outcome in severe infant/pediatric traumatic brain injury. Acta Neurochir Suppl 122: 239‐244, 2016. DOI: 10.1007/978‐3‐319‐22533‐3_48.
 327.Nakao Y, Itoh Y, Kuang TY, Cook M, Jehle J, Sokoloff L. Effects of anesthesia on functional activation of cerebral blood flow and metabolism. Proc Natl Acad Sci U S A 98: 7593‐7598, 2001.
 328.Nauli SM, Ally A, Zhang L, Gerthoffer WT, Pearce WJ. Maturation attenuates the effects of cGMP on contraction, [Ca2+]i and Ca2+ sensitivity in ovine basilar arteries. Gen Pharmacol 35: 107‐118, 2000.
 329.Nauli SM, Williams JM, Akopov SE, Zhang L, Pearce WJ. Developmental changes in ryanodine‐ and IP(3)‐sensitive Ca(2+) pools in ovine basilar artery. Am J Physiol Cell Physiol 281: C1785‐C1796, 2001. DOI: 10.1152/ajpcell.2001.281.6.C1785.
 330.Nehlig A, de Vasconcelos AP, Boyet S. Quantitative autoradiographic measurement of local cerebral glucose utilization in freely moving rats during postnatal development. J Neurosci 8: 2321‐2333, 1988.
 331.Nehlig A, Pereira de Vasconcelos A, Boyet S. Postnatal changes in local cerebral blood flow measured by the quantitative autoradiographic [14C]iodoantipyrine technique in freely moving rats. J Cereb Blood Flow Metab 9: 579‐588, 1989. DOI: 10.1038/jcbfm.1989.83.
 332.Niwa K, Araki E, Morham SG, Ross ME, Iadecola C. Cyclooxygenase‐2 contributes to functional hyperemia in whisker‐barrel cortex. J Neurosci 20: 763‐770, 2000.
 333.Nnorom CC, Davis C, Fedinec AL, Howell K, Jaggar JH, Parfenova H, Pourcyrous M, Leffler CW. Contributions of KATP and KCa channels to cerebral arteriolar dilation to hypercapnia in neonatal brain. Physiol Rep 2: e12127, 2014. DOI: 10.14814/phy2.12127.
 334.Northington FJ, Koehler RC, Traystman RJ, Martin LJ. Nitric oxide synthase 1 and nitric oxide synthase 3 protein expression is regionally and temporally regulated in fetal brain. Brain Res Dev Brain Res 95: 1‐14, 1996.
 335.Northington FJ, Tobin JR, Harris AP, Traystman RJ, Koehler RC. Developmental and regional differences in nitric oxide synthase activity and blood flow in the sheep brain. J Cerebr Blood Flow Metab 17: 109‐115, 1997.
 336.Northington FJ, Tobin JR, Koehler RC, Traystman RJ. In vivo production of nitric oxide correlates with NMDA‐induced cerebral hyperemia in newborn sheep. Am J Physiol Heart Circ Physiol 269: H215‐H221, 1995.
 337.Northington FJ, Traystman RJ, Koehler RC, Martin LJ. GLT1, glial glutamate transporter, is transiently expressed in neurons and develops astrocyte specificity only after midgestation in the ovine fetal brain. J Neurobiol 39: 515‐526, 1999.
 338.Numan T, Bain AR, Hoiland RL, Smirl JD, Lewis NC, Ainslie PN. Static autoregulation in humans: A review and reanalysis. Med Eng Phys 36: 1487‐1495, 2014. DOI: 10.1016/j.medengphy.2014.08.001.
 339.Ohata H, Cao S, Koehler RC. Contribution of adenosine A2A and A2B receptors and heme oxygenase to AMPA‐induced dilation of pial arterioles in rats. Am J Physiol Regul Integr Comp Physiol 291: R728‐R735, 2006.
 340.Ohata H, Gebremedhin D, Narayanan J, Harder DR, Koehler RC. Onset of pulmonary ventilation in fetal sheep produces pial arteriolar constriction dependent on cytochrome p450 omega‐hydroxylase activity. J Appl Physiol 109: 412‐417, 2010.
 341.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. DOI: 10.1542/peds.2008‐2004.
 342.O'Neill JT, Golden SM, Franklin GA, Alden ER. Cerebral vascular response to hemorrhagic hypotension in newborn lambs: The influence of developing anemia. Proc Soc Exp Biol Med 205: 132‐139, 1994.
 343.Ouyang M, Liu P, Jeon T, Chalak L, Heyne R, Rollins NK, Licht DJ, Detre JA, Roberts TP, Lu H, Huang H. Heterogeneous increases of regional cerebral blood flow during preterm brain development: Preliminary assessment with pseudo‐continuous arterial spin labeled perfusion MRI. NeuroImage 147: 233‐242, 2017. DOI: 10.1016/j.neuroimage.2016.12.034.
 344.Papile LA, Rudolph AM, Heymann MA. Autoregulation of cerebral blood flow in the preterm fetal lamb. Pediatr Res 19: 159‐161, 1985.
 345.Parfenova H, Carratu P, Tcheranova D, Fedinec A, Pourcyrous M, Leffler CW. Epileptic seizures cause extended postictal cerebral vascular dysfunction that is prevented by HO‐1 overexpression. Am J Physiol Heart Circ Physiol 288: H2843‐H2850, 2005.
 346.Parfenova H, Eidson TH, Leffler CW. Upregulation of COX‐2 in cerebral microvascular endothelial cells by smooth muscle cell signals. Am J Physiol Cell Physiol 273: C277‐C288, 1997.
 347.Parfenova H, Fedinec A, Leffler CW. Ionotropic glutamate receptors in cerebral microvascular endothelium are functionally linked to heme oxygenase. J Cereb Blood Flow Metab 23: 190‐197, 2003.
 348.Parfenova H, Hsu P, Leffler CW. Dilator prostanoid‐induced cyclic AMP formation and release by cerebral microvascular smooth muscle cells: Inhibition by indomethacin. J Pharmacol Exp Ther 272: 44‐52, 1995.
 349.Parfenova H, Leffler CW. Functional study on vasodilator effects of prostaglandin E2 in the newborn pig cerebral circulation. Eur J Pharmacol 278: 133‐142, 1995.
 350.Parfenova H, Levine V, Gunther WM, Pourcyrous M, Leffler CW. COX‐1 and COX‐2 contributions to basal and IL‐1 beta‐stimulated prostanoid synthesis in human neonatal cerebral microvascular endothelial cells. Pediatr Res 52: 342‐348, 2002.
 351.Parfenova H, Massie V, Leffler CW. Developmental changes in endothelium‐derived vasorelaxant factors in cerebral circulation. Am J Physiol Heart Circ Physiol 278: H780‐H788, 2000.
 352.Parfenova H, Neff RA III, Alonso JS, Shlopov BV, Jamal CN, Sarkisova SA, Leffler CW. Cerebral vascular endothelial heme oxygenase: Expression, localization, and activation by glutamate. Am J Physiol Cell Physiol 281: C1954‐C1963, 2001.
 353.Parfenova H, Pourcyrous M, Fedinec AL, Liu J, Basuroy S, Leffler CW. Astrocyte‐produced carbon monoxide and the carbon monoxide donor CORM‐A1 protect against cerebrovascular dysfunction caused by prolonged neonatal asphyxia. Am J Physiol Heart Circ Physiol 315: H978‐H988, 2018. DOI: 10.1152/ajpheart.00140.2018.
 354.Parfenova H, Shibata M, Zuckerman S, Leffler CW. CO2 and cerebral circulation in newborn pigs: Cyclic nucleotides and prostanoids in vascular regulation. Am J Physiol Heart Circ Physiol 266: H1494‐H1501, 1994. DOI: 10.1152/ajpheart.1994.266.4.H1494.
 355.Parfenova H, Shibata M, Zuckerman S, Mirro R, Leffler CW. Cyclic nucleotides and cerebrovascular tone in newborn pigs. Am J Physiol Heart Circ Physiol 265: H1972‐H1982, 1993.
 356.Parfenova H, Zuckerman S, Leffler CW. Inhibitory effect of indomethacin on prostacyclin receptor‐mediated cerebral vascular responses. Am J Physiol Heart Circ Physiol 268: H1884‐H1890, 1995. DOI: 10.1152/ajpheart.1995.268.5.H1884.
 357.Park TS, Van Wylen DG, Rubio R, Berne RM. Increased brain interstitial fluid adenosine concentration during hypoxia in newborn piglet. J Cereb Blood Flow Metab 7: 178‐183, 1987. DOI: 10.1038/jcbfm.1987.41.
 358.Park TS, Van Wylen DG, Rubio R, Berne RM. Brain interstitial adenosine and sagittal sinus blood flow during systemic hypotension in piglet. J Cereb Blood Flow Metab 8: 822‐828, 1988. DOI: 10.1038/jcbfm.1988.138.
 359.Parkinson PA, Parfenova H, Leffler CW. Phospholipase C activation by prostacyclin receptor agonist in cerebral microvascular smooth muscle cells. Proc Soc Exp Biol Med 223: 53‐58, 2000.
 360.Pasternak JF, Groothuis DR. Autoregulation of cerebral blood flow in the newborn beagle puppy. Biol Neonate 48: 100‐109, 1985. DOI: 10.1159/000242160.
 361.Pastor P, Curvello V, Hekierski H, Armstead WM. Inhaled nitric oxide protects cerebral autoregulation through prevention of impairment of ATP and calcium sensitive K channel mediated cerebrovasodilation after traumatic brain injury. Brain Res 1711: 1‐6, 2019. DOI: 10.1016/j.brainres.2019.01.008.
 362.Pearce WJ, Ashwal S. Developmental changes in thickness, contractility, and hypoxic sensitivity of newborn lamb cerebral arteries. Pediatr Res 22: 192‐196, 1987. DOI: 10.1203/00006450‐198708000‐00019.
 363.Pearce WJ, Elliott SR. Maturation enhances the sensitivity of ovine cerebral arteries to the ATP‐sensitive potassium channel activator lemakalim. Pediatr Res 35: 729‐732, 1994. DOI: 10.1203/00006450‐199406000‐00021.
 364.Pearce WJ, Hull AD, Long DM, Longo LD. Developmental changes in ovine cerebral artery composition and reactivity. Am J Physiol Regul Integr Comp Physiol 261: R458‐R465, 1991. DOI: 10.1152/ajpregu.1991.261.2.R458.
 365.Pearce WJ, Khorram O. Maturation and differentiation of the fetal vasculature. Clin Obstet Gynecol 56: 537‐548, 2013. DOI: 10.1097/GRF.0b013e31829e5bc9.
 366.Peng X, Carhuapoma JR, Bhardwaj A, Alkayed NJ, Falck JR, Harder DR, Traystman RJ, Koehler RC. Suppression of cortical functional hyperemia to vibrissal stimulation in the rat by epoxygenase inhibitors. Am J Physiol Heart Circ Physiol 283: H2029‐H2037, 2002.
 367.Peng X, Zhang C, Alkayed NJ, Harder DR, Koehler RC. Dependency of cortical functional hyperemia to forepaw stimulation on epoxygenase and nitric oxide synthase activities in rats. J Cereb Blood Flow Metab 24: 509‐517, 2004.
 368.Peppiatt CM, Howarth C, Mobbs P, Attwell D. Bidirectional control of CNS capillary diameter by pericytes. Nature 443: 700‐704, 2006.
 369.Pourcyrous M, Bada HS, Parfenova H, Daley ML, Korones SB, Leffler CW. Cerebrovasodilatory contribution of endogenous carbon monoxide during seizures in newborn pigs. Pediatr Res 51: 579‐585, 2002.
 370.Qin X, Kwansa H, Bucci E, Dore S, Boehning D, Shugar D, Koehler RC. Role of heme oxygenase‐2 in pial arteriolar response to acetylcholine in mice with and without transfusion of cell‐free hemoglobin polymers. Am J Physiol Regul Integr Comp Physiol 295: R498‐R504, 2008.
 371.Rama GP, Parfenova H, Leffler CW. Protein kinase Cs and tyrosine kinases in permissive action of prostacyclin on cerebrovascular regulation in newborn pigs. Pediatr Res 41: 83‐89, 1997. DOI: 10.1203/00006450‐199704001‐00506.
 372.Rebel A, Cao S, Kwansa H, Dore S, Bucci E, Koehler RC. Dependence of acetylcholine and ADP dilation of pial arterioles on heme oxygenase after transfusion of cell‐free polymeric hemoglobin. Am J Physiol Heart Circ Physiol 290: H1027‐H1037, 2006.
 373.Rebich S, Devine JO, Armstead WM. Role of nitric oxide and cAMP in beta‐adrenoceptor‐induced pial artery vasodilation. Am J Phys 268: H1071‐H1076, 1995. DOI: 10.1152/ajpheart.1995.268.3.H1071.
 374.Reivich M, Brann AW Jr, Shapiro H, Rawson J, Sano N. Reactivity of cerebral vessels to CO2 in the newborn rhesus monkey. Eur Neurol 6: 132‐136, 1971. DOI: 10.1159/000114481.
 375.Reynolds JD, Zeballos GA, Penning DH, Kimura KA, Atkins B, Brien JF. Nitrate and nitrite anion concentration in the intact cerebral cortex of preterm and nearterm fetal sheep: Indirect index of in vivo nitric oxide formation. J Pharmacol Toxicol Methods 39: 125‐128, 1998.
 376.Rickards CA. Cerebral blood‐flow regulation during hemorrhage. Compr Physiol 5: 1585‐1621, 2015. DOI: 10.1002/c140058.
 377.Roach MR. The static elastic properties of carotid arteries from fetal sheep. Can J Physiol Pharmacol 48: 694‐708, 1970.
 378.Robinson JS, Fedinec AL, Leffler CW. Role of carbon monoxide in glutamate receptor‐induced dilation of newborn pig pial arterioles. Am J Physiol Heart Circ Physiol 282: H2371‐H2376, 2002.
 379.Roche‐Labarbe N, Carp SA, Surova A, Patel M, Boas DA, Grant PE, Franceschini MA. Noninvasive optical measures of CBV, StO(2), CBF index, and rCMRO(2) in human premature neonates' brains in the first six weeks of life. Hum Brain Mapp 31: 341‐352, 2010. DOI: 10.1002/hbm.20868.
 380.Roche‐Labarbe N, Fenoglio A, Aggarwal A, Dehaes M, Carp SA, Franceschini MA, Grant PE. Near‐infrared spectroscopy assessment of cerebral oxygen metabolism in the developing premature brain. J Cereb Blood Flow Metab 32: 481‐488, 2012. DOI: 10.1038/jcbfm.2011.145.
 381.Roche‐Labarbe N, Fenoglio A, Radhakrishnan H, Kocienski‐Filip M, Carp SA, Dubb J, Boas DA, Grant PE, Franceschini MA. Somatosensory evoked changes in cerebral oxygen consumption measured non‐invasively in premature neonates. NeuroImage 85 Pt 1: 279‐286, 2014. DOI: 10.1016/j.neuroimage.2013.01.035.
 382.Rosenberg AA, Harris AP, Koehler RC, Hudak ML, Traystman RJ, Jones MD Jr. Role of O2‐hemoglobin affinity in the regulation of cerebral blood flow in fetal sheep. Am J Physiol Heart Circ Physiol 251: H56‐H62, 1986.
 383.Rosenberg AA, Jones MD Jr, Traystman RJ, Simmons MA, Molteni RA. Response of cerebral blood flow to changes in PCO2 in fetal, newborn, and adult sheep. Am J Physiol Heart Circ Physiol 242: H862‐H866, 1982.
 384.Rosenegger DG, Tran CH, Wamsteeker Cusulin JI, Gordon GR. Tonic local brain blood flow control by astrocytes independent of phasic neurovascular coupling. J Neurosci 35: 13463‐13474, 2015. DOI: 10.1523/JNEUROSCI.1780‐15.2015.
 385.Rossberg MI, Armstead WM. Relationship between vasopressin and opioids in hypoxia induced pial artery vasodilation. Am J Physiol Heart Circ Physiol 271: H521‐H527, 1996. DOI: 10.1152/ajpheart.1996.271.2.H521.
 386.Rossberg MI, Armstead WM. Role of cyclic nucleotides in vasopressin‐induced piglet pial artery dilation and opioid release. Pediatr Res 41: 498‐504, 1997. DOI: 10.1203/00006450‐199704000‐00008.
 387.Sandoval RJ, Injeti ER, Williams JM, Georthoffer WT, Pearce WJ. Myogenic contractility is more dependent on myofilament calcium sensitization in term fetal than adult ovine cerebral arteries. Am J Physiol Heart Circ Physiol 293: H548‐H556, 2007. DOI: 10.1152/ajpheart.00134.2007.
 388.Satterthwaite TD, Shinohara RT, Wolf DH, Hopson RD, Elliott MA, Vandekar SN, Ruparel K, Calkins ME, Roalf DR, Gennatas ED, Jackson C, Erus G, Prabhakaran K, Davatzikos C, Detre JA, Hakonarson H, Gur RC, Gur RE. Impact of puberty on the evolution of cerebral perfusion during adolescence. Proc Natl Acad Sci U S A 111: 8643‐8648, 2014. DOI: 10.1073/pnas.1400178111.
 389.Schleien CL, Eberle B, Shaffner DH, Koehler RC, Traystman RJ. Reduced blood‐brain barrier permeability after cardiac arrest by conjugated superoxide dismutase and catalase in piglets. Stroke 25: 1830‐1834, 1994.
 390.Schleien CL, Koehler RC, Shaffner DH, Eberle B, Traystman RJ. Blood‐brain barrier disruption after cardiopulmonary resuscitation in immature swine. Stroke 22: 477‐483, 1991.
 391.Sciotti VM, Park TS, Berne RM, Van Wylen DG. Changes in extracellular adenosine during chemical or electrical brain stimulation. Brain Res 613: 16‐20, 1993.
 392.Settergren G, Lindblad BS, Persson B. Cerebral blood flow and exchange of oxygen, glucose, ketone bodies, lactate, pyruvate and amino acids in infants. Acta Paediatr Scand 65: 343‐353, 1976.
 393.Settergren G, Lindblad BS, Persson B. Cerebral blood flow and exchange of oxygen, glucose ketone bodies, lactate, pyruvate and amino acids in anesthetized children. Acta Paediatr Scand 69: 457‐465, 1980.
 394.Shankar V, Armstead WM. Opioids contribute to hypoxia‐induced pial artery dilation through activation of ATP‐sensitive K+ channels. Am J Physiol Heart Circ Physiol 269: H997‐H1002, 1995.
 395.Sharon G, Sampson TR, Geschwind DH, Mazmanian SK. The central nervous system and the gut microbiome. Cell 167: 915‐932, 2016. DOI: 10.1016/j.cell.2016.10.027.
 396.Shi Y, Liu X, Gebremedhin D, Falck JR, Harder DR, Koehler RC. Interaction of mechanisms involving epoxyeicosatrienoic acids, adenosine receptors, and metabotropic glutamate receptors in neurovascular coupling in rat whisker barrel cortex. J Cereb Blood Flow Metab 28: 111‐125, 2008.
 397.Shibasaki K, Ikenaka K, Tamalu F, Tominaga M, Ishizaki Y. A novel subtype of astrocytes expressing TRPV4 (transient receptor potential vanilloid 4) regulates neuronal excitability via release of gliotransmitters. J Biol Chem 289: 14470‐14480, 2014. DOI: 10.1074/jbc.M114.557132.
 398.Shibata M, Parfenova H, Zuckerman SL, Seyer JM, Krueger JM, Leffler CW. Interleukin‐1 beta peptides induce cerebral pial arteriolar dilation in anesthetized newborn pigs. Am J Physiol Regul Integr Comp Physiol 270: R1044‐R1050, 1996. DOI: 10.1152/ajpregu.1996.270.5.R1044.
 399.Skov L, Pryds O, Greisen G. Estimating cerebral blood flow in newborn infants: Comparison of near infrared spectroscopy and 133Xe clearance. Pediatr Res 30: 570‐573, 1991. DOI: 10.1203/00006450‐199112000‐00016.
 400.Stefanovic M, Puchulu‐Campanella E, Kodippili G, Low PS. Oxygen regulates the band 3‐ankyrin bridge in the human erythrocyte membrane. Biochem J 449: 143‐150, 2013. DOI: 10.1042/BJ20120869.
 401.Stonestreet BS, Barefield ES, Piva D, Goldstein M. Effects of hypercarbia on autoregulation of brain blood flow and cerebral metabolism in newborn piglets. Reprod Fertil Dev 7: 1381‐1387, 1995.
 402.Stonestreet BS, Patlak CS, Pettigrew KD, Reilly CB, Cserr HF. Ontogeny of blood‐brain barrier function in ovine fetuses, lambs, and adults. Am J Physiol Regul Integr Comp Physiol 271: R1594‐R1601, 1996. DOI: 10.1152/ajpregu.1996.271.6.R1594.
 403.Sun W, McConnell E, Pare JF, Xu Q, Chen M, Peng W, Lovatt D, Han X, Smith Y, Nedergaard M. Glutamate‐dependent neuroglial calcium signaling differs between young and adult brain. Science 339: 197‐200, 2013. DOI: 10.1126/science.1226740.
 404.Symon L, Held K, Dorsch NW. On the myogenic nature of the autoregulatory mechanism in the cerebral circulation. Eur Neurol 6: 11‐18, 1971.
 405.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. DOI: 10.1203/00006450‐199010000‐00012.
 406.Taga G, Asakawa K, Maki A, Konishi Y, Koizumi H. Brain imaging in awake infants by near‐infrared optical topography. Proc Natl Acad Sci U S A 100: 10722‐10727, 2003. DOI: 10.1073/pnas.1932552100.
 407.Taguchi H, Heistad DD, Kitazono T, Faraci FM. ATP‐sensitive K+ channels mediate dilatation of cerebral arterioles during hypoxia. Circ Res 74: 1005‐1008, 1994.
 408.Takahashi T, Shirane R, Sato S, Yoshimoto T. Developmental changes of cerebral blood flow and oxygen metabolism in children. Am J Neuroradiol 20: 917‐922, 1999.
 409.Tekes A, Poretti A, Scheurkogel MM, Huisman TA, Howlett JA, Alqahtani E, Lee JH, Parkinson C, Shapiro K, Chung SE, Jennings JM, Gilmore MM, Hogue CW, Martin LJ, Koehler RC, Northington FJ, Lee JK. Apparent diffusion coefficient scalars correlate with near‐infrared spectroscopy markers of cerebrovascular autoregulation in neonates cooled for perinatal hypoxic‐ischemic injury. AJNR Am J Neuroradiol 36: 188‐193, 2015. DOI: 10.3174/ajnr.A4083.
 410.Teng GQ, Nauli SM, Brayden JE, Pearce WJ. Maturation alters the contribution of potassium channels to resting and 5HT‐induced tone in small cerebral arteries of the sheep. Brain Res Dev Brain Res 133: 81‐91, 2002.
 411.Ting P, Yamaguchi S, Bacher JD, Killens RH, Myers RE. Hypoxic‐ischemic cerebral necrosis in midgestational sheep fetuses: Physiopathologic correlations. Exp Neurol 80: 227‐245, 1983.
 412.Tomimoto H, Nishimura M, Suenaga T, Nakamura S, Akiguchi I, Wakita H, Kimura J, Mayer B. Distribution of nitric oxide synthase in the human cerebral blood vessels and brain tissues. J Cereb Blood Flow Metab 14: 930‐938, 1994. DOI: 10.1038/jcbfm.1994.124.
 413.Tomiyama Y, Brian JE Jr, Todd MM. Cerebral blood flow during hemodilution and hypoxia in rats: Role of ATP‐sensitive potassium channels. Stroke 30: 1942‐1947, 1999.
 414.Tong S, Parfenova H, Shibata M, Zuckerman S, Armstead WM, Leffler CW. Pituitary adenylate cyclase‐activating polypeptide dilates cerebral arterioles of newborn pigs. Proc Soc Exp Biol Med 203: 343‐347, 1993.
 415.Tortora D, Mattei PA, Navarra R, Panara V, Salomone R, Rossi A, Detre JA, Caulo M. Prematurity and brain perfusion: Arterial spin labeling MRI. Neuroimage Clin 15: 401‐407, 2017. DOI: 10.1016/j.nicl.2017.05.023.
 416.Tuor UI, Grewal D. Autoregulation of cerebral blood flow: Influence of local brain development and postnatal age. Am J Physiol Heart Circ Physiol 267: H2220‐H2228, 1994.
 417.Tuor UI, Kurpita G, Simone C. Correlation of local changes in cerebral blood flow, capillary density, and cytochrome oxidase during development. J Comp Neurol 342: 439‐448, 1994.
 418.Tuor UI, Simone C, Bascaramurty S. Local blood‐brain barrier in the newborn rabbit: Postnatal changes in à‐aminoisobutyric acid transfer within medulla, cortex, and selected brain areas. J Neurochem 59: 999‐1007, 1992.
 419.Tweed WA, Cote J, Pash M, Lou H. Arterial oxygenation determines autoregulation of cerebral blood flow in the fetal lamb. Pediatr Res 17: 246‐249, 1983. DOI: 10.1203/00006450‐198304000‐00002.
 420.Tweed WA, Cote J, Wade JG, Gregory G, Mills A. Preservation of fetal brain blood flow relative to other organs during hypovolemic hypotension. Pediatr Res 16: 137‐140, 1982.
 421.Tzeng YC, Ainslie PN. Blood pressure regulation IX: Cerebral autoregulation under blood pressure challenges. Eur J Appl Physiol 114: 545‐559, 2014. DOI: 10.1007/s00421‐013‐2667‐y.
 422.Van Bel F, Sola A, Roman C, Rudolph AM. Role of nitric oxide in the regulation of the cerebral circulation in the lamb fetus during normoxemia and hypoxemia. Biol Neonate 68: 200‐210, 1995.
 423.Van Bel F, Sola A, Roman C, Rudolph AM. Perinatal regulation of the cerebral circulation: Role of nitric oxide and prostaglandins. Pediatr Res 42: 299‐304, 1997.
 424.Vandekar SN, Shou H, Satterthwaite TD, Shinohara RT, Merikangas AK, Roalf DR, Ruparel K, Rosen A, Gennatas ED, Elliott MA, Davatzikos C, Gur RC, Gur RE, Detre JA. Sex differences in estimated brain metabolism in relation to body growth through adolescence. J Cereb Blood Flow Metab 39: 524‐535, 2019. DOI: 10.1177/0271678X17737692.
 425.Vannucci SJ, Clark RR, Koehler‐Stec E, Li K, Smith CB, Davies P, Maher F, Simpson IA. Glucose transporter expression in brain: Relationship to cerebral glucose utilization. Dev Neurosci 20: 369‐379, 1998. DOI: 10.1159/000017333.
 426.Vannucci SJ, Maher F, Koehler E, Simpson IA. Altered expression of GLUT‐1 and GLUT‐3 glucose transporters in neurohypophysis of water‐deprived or diabetic rats. Am J Phys 267: E605‐E611, 1994. DOI: 10.1152/ajpendo.1994.267.4.E605.
 427.Vannucci SJ, Seaman LB, Brucklacher RM, Vannucci RC. Glucose transport in developing rat brain: Glucose transporter proteins, rate constants and cerebral glucose utilization. Mol Cell Biochem 140: 177‐184, 1994.
 428.Vannucci SJ, Simpson IA. Developmental switch in brain nutrient transporter expression in the rat. Am J Physiol Endocrinol Metab 285: E1127‐E1134, 2003. DOI: 10.1152/ajpendo.00187.2003.
 429.Vavilala MS, Kincaid MS, Muangman SL, Suz P, Rozet I, Lam AM. Gender differences in cerebral blood flow velocity and autoregulation between the anterior and posterior circulations in healthy children. Pediatr Res 58: 574‐578, 2005. DOI: 10.1203/01.PDR.0000179405.30737.0F.
 430.Vavilala MS, Lee LA, Boddu K, Visco E, Newell DW, Zimmerman JJ, Lam AM. Cerebral autoregulation in pediatric traumatic brain injury. Pediatr Crit Care Med 5: 257‐263, 2004.
 431.Venteicher A, Armstead WM. Vasopressin contributes to dynorphin modulation of hypoxic cerebrovasodilation. Am J Physiol Heart Circ Physiol 275: H2072‐H2079, 1998. DOI: 10.1152/ajpheart.1998.275.6.H2072.
 432.Vesoulis ZA, Liao SM, Trivedi SB, Ters NE, Mathur AM. A novel method for assessing cerebral autoregulation in preterm infants using transfer function analysis. Pediatr Res 79: 453‐459, 2016. DOI: 10.1038/pr.2015.238.
 433.Virgintino D, Errede M, Girolamo F, Capobianco C, Robertson D, Vimercati A, Serio G, Di Benedetto A, Yonekawa Y, Frei K, Roncali L. Fetal blood‐brain barrier P‐glycoprotein contributes to brain protection during human development. J Neuropathol Exp Neurol 67: 50‐61, 2008. DOI: 10.1097/nen.0b013e31815f65d9.
 434.Virgintino D, Errede M, Robertson D, Capobianco C, Girolamo F, Vimercati A, Bertossi M, Roncali L. Immunolocalization of tight junction proteins in the adult and developing human brain. Histochem Cell Biol 122: 51‐59, 2004. DOI: 10.1007/s00418‐004‐0665‐1.
 435.Virgintino D, Robertson D, Benagiano V, Errede M, Bertossi M, Ambrosi G, Roncali L. Immunogold cytochemistry of the blood‐brain barrier glucose transporter GLUT1 and endogenous albumin in the developing human brain. Brain Res Dev Brain Res 123: 95‐101, 2000.
 436.Wagerle LC, Busija DW. Cholinergic mechanisms in the cerebral circulation of the newborn piglet: Effect of inhibitors of arachidonic acid metabolism. Circ Res 64: 1030‐1036, 1989.
 437.Wagerle LC, Degiulio PA. Indomethacin‐sensitive CO2 reactivity of cerebral arterioles is restored by vasodilator prostaglandin. Am J Physiol Heart Circ Physiol 266: H1332‐H1338, 1994. DOI: 10.1152/ajpheart.1994.266.4.H1332.
 438.Wagerle LC, Delivoria‐Papadopoulos M. Alpha‐adrenergic receptor subtypes in the cerebral circulation of newborn piglets. Am J Physiol Heart Circ Physiol 252: R1092‐R1098, 1987. DOI: 10.1152/ajpregu.1987.252.6.R1092.
 439.Wagerle LC, Heffernan TM, Sacks LM, Delivoria‐Papadopoulos M. Sympathetic effect on cerebral blood flow regulation in hypoxic newborn lambs. Am J Physiol Heart Circ Physiol 245: H487‐H494, 1983. DOI: 10.1152/ajpheart.1983.245.3.H487.
 440.Wagerle LC, Kumar SP, Delivoria‐Papadopoulos M. Effect of sympathetic nerve stimulation on cerebral blood flow in newborn piglets. Pediatr Res 20: 131‐135, 1986. DOI: 10.1203/00006450‐198602000‐00007.
 441.Wagerle LC, Kurth CD, Busija DW. Cholinergic reactivity of cerebral arteries in the developing fetal and newborn lamb. J Dev Physiol 17: 51‐54, 1992.
 442.Wagerle LC, Kurth CD, Roth RA. Sympathetic reactivity of cerebral arteries in developing fetal lamb and adult sheep. Am J Physiol Heart Circ Physiol 258: H1432‐H1438, 1990. DOI: 10.1152/ajpheart.1990.258.5.H1432.
 443.Wagerle LC, Mishra OP. Mechanism of CO2 response in cerebral arteries of the newborn pig: Role of phospholipase, cyclooxygenase, and lipoxygenase pathways. Circ Res 62: 1019‐1026, 1988.
 444.Wagerle LC, Moliken W, Russo P. Nitric oxide and beta‐adrenergic mechanisms modify contractile responses to norepinephrine in ovine fetal and newborn cerebral arteries. Pediatr Res 38: 237‐242, 1995. DOI: 10.1203/00006450‐199508000‐00017.
 445.Wagner KR, Ting P, Westfall MV, Yamaguchi S, Bacher JD, Myers RE. Brain metabolic correlates of hypoxic‐ischemic cerebral necrosis in mid‐gestational sheep fetuses: Significance of hypotension. J Cereb Blood Flow Metab 6: 425‐434, 1986. DOI: 10.1038/jcbfm.1986.75.
 446.Wang Q, Kjaer T, Jorgensen MB, Paulson OB, Lassen NA, Diemer NH, Lou HC. Nitric oxide does not act as a mediator coupling cerebral blood flow to neural activity following somatosensory stimuli in rats. Neurol Res 15: 33‐36, 1993.
 447.Wang R, Wang Z, Wu L. Carbon monoxide‐induced vasorelaxation and the underlying mechanisms. Br J Pharmacol 121: 927‐934, 1997.
 448.Wang R, Wu L. The chemical modification of KCa channels by carbon monoxide in vascular smooth muscle cells. J Biol Chem 272: 8222‐8226, 1997.
 449.Wang R, Wu L, Wang Z. The direct effect of carbon monoxide on KCa channels in vascular smooth muscle cells. Pflugers Arch 434: 285‐291, 1997.
 450.Wei HS, Kang H, Rasheed ID, Zhou S, Lou N, Gershteyn A, McConnell ED, Wang Y, Richardson KE, Palmer AF, Xu C, Wan J, Nedergaard M. Erythrocytes are oxygen‐sensing regulators of the cerebral microcirculation. Neuron 91: 851‐862, 2016. DOI: 10.1016/j.neuron.2016.07.016.
 451.White CR, Hamade MW, Siami K, Chang MM, Mangalwadi A, Frangos JA, Pearce WJ. Maturation enhances fluid shear‐induced activation of eNOS in perfused ovine carotid arteries. Am J Physiol Heart Circ Physiol 289: H2220‐H2227, 2005. DOI: 10.1152/ajpheart.01013.2004.
 452.White CR, Hao X, Pearce WJ. Maturational differences in soluble guanylate cyclase activity in ovine carotid and cerebral arteries. Pediatr Res 47: 369‐375, 2000.
 453.White RP, Hindley C, Bloomfield PM, Cunningham VJ, Vallance P, Brooks DJ, Markus HS. The effect of the nitric oxide synthase inhibitor L‐NMMA on basal CBF and vasoneuronal coupling in man: A PET study. J Cereb Blood Flow Metab 19: 673‐678, 1999. DOI: 10.1097/00004647‐199906000‐00011.
 454.Wilderman MJ, Armstead WM. Relationship between nitric oxide and opioids in hypoxia‐induced pial artery vasodilation. Am J Physiol 270: H869‐H874, 1996. DOI: 10.1152/ajpheart.1996.270.3.H869.
 455.Wilderman MJ, Armstead WM. Role of neuronal NO synthase in relationship between NO and opioids in hypoxia‐induced pial artery dilation. Am J Physiol Heart Circ Physiol 273: H1807‐H1815, 1997.
 456.Wilderman MJ, Armstead WM. Role of endothelial nitric oxide synthase in hypoxia‐induced pial artery dilation. J Cereb Blood Flow Metab 18: 531‐538, 1998. DOI: 10.1097/00004647‐199805000‐00008.
 457.Williams JM, Hull AD, Pearce WJ. Maturational modulation of endothelium‐dependent vasodilatation in ovine cerebral arteries. Am J Physiol Regul Integr Comp Physiol 288: R149‐R157, 2005. DOI: 10.1152/ajpregu.00427.2004.
 458.Williams JM, Pearce WJ. Age‐dependent modulation of endothelium‐dependent vasodilatation by chronic hypoxia in ovine cranial arteries. J Appl Physiol (1985) 100: 225‐232, 2006. DOI: 10.1152/japplphysiol.00221.2005.
 459.Willis AP, Leffler CW. NO and prostanoids: Age dependence of hypercapnia and histamine‐induced dilations of pig pial arterioles. Am J Physiol Heart Circ Physiol 277: H299‐H307, 1999.
 460.Willis AP, Leffler CW. Endothelial NO and prostanoid involvement in newborn and juvenile pig pial arteriolar vasomotor responses. Am J Physiol Heart Circ Physiol 281: H2366‐H2377, 2001. DOI: 10.1152/ajpheart.2001.281.6.H2366.
 461.Winestone JS, Bonner C, Leffler CW. Carbon monoxide as an attenuator of vasoconstriction in piglet cerebral arterioles. Exp Biol Med (Maywood) 228: 46‐50, 2003.
 462.Winquist RJ, Webb RC, Bohr DF. Relaxation to transmural nerve stimulation and exogenously added norepinephrine in porcine cerebral vessels. A study utilizing cerebrovascular intrinsic tone. Circ Res 51: 769‐776, 1982.
 463.Wu L, Cao K, Lu Y, Wang R. Different mechanisms underlying the stimulation of KCa channels by nitric oxide and carbon monoxide. J Clin Invest 110: 691‐700, 2002.
 464.Xi Q, Tcheranova D, Basuroy S, Parfenova H, Jaggar JH, Leffler CW. Glutamate‐induced calcium signals stimulate CO production in piglet astrocytes. Am J Physiol Heart Circ Physiol 301: H428‐H433, 2011.
 465.Xi Q, Tcheranova D, Parfenova H, Horowitz B, Leffler CW, Jaggar JH. Carbon monoxide activates KCa channels in newborn arteriole smooth muscle cells by increasing apparent Ca2+ sensitivity of à‐subunits. Am J Physiol Heart Circ Physiol 286: H610‐H618, 2004.
 466.Xi Q, Umstot E, Zhao G, Narayanan D, Leffler CW, Jaggar JH. Glutamate regulates Ca2+ signals in smooth muscle cells of newborn piglet brain slice arterioles through astrocyte‐ and heme oxygenase‐dependent mechanisms. Am J Physiol Heart Circ Physiol 298: H562‐H569, 2010.
 467.Yamada H, Sadato N, Konishi Y, Kimura K, Tanaka M, Yonekura Y, Ishii Y. A rapid brain metabolic change in infants detected by fMRI. Neuroreport 8: 3775‐3778, 1997.
 468.Yamada H, Sadato N, Konishi Y, Muramoto S, Kimura K, Tanaka M, Yonekura Y, Ishii Y, Itoh H. A milestone for normal development of the infantile brain detected by functional MRI. Neurology 55: 218‐223, 2000.
 469.Yamaura K, Gebremedhin D, Zhang C, Narayanan J, Hoefert K, Jacobs ER, Koehler RC, Harder DR. Contribution of epoxyeicosatrienoic acids to the hypoxia‐induced activation of Ca2+‐activated K+ channel current in cultured rat hippocampal astrocytes. Neuroscience 143: 703‐716, 2006.
 470.Yan XX, Garey LJ. Morphological diversity of nitric oxide synthesising neurons in mammalian cerebral cortex. J Hirnforsch 38: 165‐172, 1997.
 471.Yan XX, Garey LJ, Jen LS. Prenatal development of NADPH‐diaphorase‐reactive neurons in human frontal cortex. Cereb Cortex 6: 737‐745, 1996.
 472.Yan XX, Jen LS, Garey LJ. NADPH‐diaphorase‐positive neurons in primate cerebral cortex colocalize with GABA and calcium‐binding proteins. Cereb Cortex 6: 524‐529, 1996.
 473.Younkin DP, Reivich M, Jaggi J, Obrist W, Delivoria‐Papadopoulos M. Noninvasive method of estimating human newborn regional cerebral blood flow. J Cereb Blood Flow Metab 2: 415‐420, 1982. DOI: 10.1038/jcbfm.1982.47.
 474.Younkin DP, Reivich M, Jaggi JL, Obrist WD, Delivoria‐Papadopoulos M. The effect of hematocrit and systolic blood pressure on cerebral blood flow in newborn infants. J Cereb Blood Flow Metab 7: 295‐299, 1987. DOI: 10.1038/jcbfm.1987.66.
 475.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. DOI: 10.1203/00006450‐199809000‐00004.
 476.Zehendner CM, Tsohataridis S, Luhmann HJ, Yang JW. Developmental switch in neurovascular coupling in the immature rodent barrel cortex. PLoS One 8: e80749, 2013. DOI: 10.1371/journal.pone.0080749.
 477.Zeiler FA, Donnelly J, Menon DK, Smielewski P, Zweifel C, Brady K, Czosnyka M. Continuous autoregulatory indices derived from multi‐modal monitoring: Each one is not like the other. J Neurotrauma 34: 3070‐3080, 2017. DOI: 10.1089/neu.2017.5129.
 478.Zhang Y, Leffler CW. Compensatory role of NO in cerebral circulation of piglets chronically treated with indomethacin. Am J Physiol Regul Integr Comp Physiol 282: R400‐R410, 2002.
 479.Zimmermann A, Leffler CW, Tcheranova D, Fedinec AL, Parfenova H. Cerebroprotective effects of the CO‐releasing molecule CORM‐A1 against seizure‐induced neonatal vascular injury. Am J Physiol Heart Circ Physiol 293: H2501‐H2507, 2007.
 480.Zuckerman SL, Armstead WM, Hsu P, Shibata M, Leffler CW. Age dependence of cerebrovascular response mechanisms in domestic pigs. Am J Physiol Heart Circ Physiol 271: H535‐H540, 1996.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Raymond C. Koehler. Regulation of the Cerebral Circulation During Development. Compr Physiol 2021, 11: 2371-2432. doi: 10.1002/cphy.c200028