Comprehensive Physiology Wiley Online Library

Regulation of Cerebral Blood Flow: Response to Cytochrome P450 Lipid Metabolites

Full Article on Wiley Online Library



ABSTRACT

There have been numerous reviews related to the cerebral circulation. Most of these reviews are similar in many ways. In the present review, we thought it important to provide an overview of function with specific attention to details of cerebral arterial control related to brain homeostasis, maintenance of neuronal energy demands, and a unique perspective related to the role of astrocytes. A coming review in this series will discuss cerebral vascular development and unique properties of the neonatal circulation and developing brain, thus, many aspects of development are missing here. Similarly, a review of the response of the brain and cerebral circulation to heat stress has recently appeared in this series (8). By trying to make this review unique, some obvious topics were not discussed in lieu of others, which are from recent and provocative research such as endothelium‐derived hyperpolarizing factor, circadian regulation of proteins effecting cerebral blood flow, and unique properties of the neurovascular unit. © 2018 American Physiological Society. Compr Physiol 8:801‐821, 2018.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Schematic representation of glucose metabolism. Glucose enters cells trough glucose transporters (GLUTs) and is phosphorylated by hexokinase (HK) to produce glucose‐6‐phosphate (glucose‐6P). Glucose‐6P can be processed into three main metabolic pathways. First, it can be metabolized through glycolysis (i), giving rise to two molecules of pyruvate and producing adenosine phosphate (ATP) and nicotinamide adenine dinucleotide (NADH). Pyruvate can then enter mitochondria, where it is metabolized through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (Ox Phos), producing ATP and CO2 while consuming oxygen. Pyruvate can otherwise be reduced to lactate‐by‐lactate dehydrogenase (LDH). This lactate can be released in the extracellular space through monocarboxylate transporters (MCTs). The complete oxidation of glucose produces larger amounts of energy in the form of ATP in the mitochondria (30‐34 ATP) compared to glycolysis (2 ATP). Alternatively, glucose‐6P can be processed through the pentose phosphate pathway (PPP) (ii), leading to the production of reducing equivalent in the form of nicotinamide adenine dinucleotide phosphate (NADPH). Note that the PPP and glycolysis are linked at the level of glyceraldehyde‐3‐phosphate (GA3P) and fructose‐6‐phosphate (fructose‐6P). Finally, in astrocytes, glucose‐6P can also be used to store glucosyl units as glycogen (iii). Abbreviations are as follows: GPI, glucose‐6‐phosphate isomerase; PFK, phosphofructokinase‐1; Fructose‐1,6‐P2, fructose‐1,6‐bisphosphate; DHAP, dihydroxyacetone phosphate; TPI, triose phosphate isomerase; G6PDH, glucose‐6‐phosphate dehydrogenase; 6‐PGL, 6‐phosphoglucono‐δ‐lactone; 6‐PG, 6‐phosphogluconate; 6 PGDH, 6‐phosphogluconate dehydrogenase; ribulose‐5P, ribulose‐5‐phosphate; ribose‐5P, ribose‐5‐phosphate; xylulose‐5P, xylulose‐5‐phosphate; TK, transketolase; sedoheptulose‐7P, sedoheptulose‐7‐phosphate; TA, transaldolase; and erythrose‐4P, erythrose‐4‐phosphate. (Reproduced, with permission, from Belanger M, Allaman I, and Magistretti PJ, 2011, 12.)
Figure 2. Figure 2. Dependence of brain activity on blood flow in response to initiation of different task performance as shown by functional MRI signals.
Figure 3. Figure 3. Schematic representation of a neurovascular unit with astrocytes being the central processor of neuronal signals as depicted in both panels A and panel B.
Figure 4. Figure 4. (A) Immunostaining depiction of components of the neurovascular unit (NVU). The astrocytes (stained with rhodamine labeled GFAP) shown in red. The neurons are stained with fluorescein tagged NSE shown in green and the blood vessels are stained with PECAM shown in blue. Note the location of the foot processes around the vasculature. (B) Histochemical localization of β‐galactosidase expression in rat brain following lateral ventricular infusion of Ad5/CMV‐β‐galactosidase (magnification × 1000). Note staining of astrocytes and astrocytic foot processes surrounding blood vessel emulating the exploded section of the immunostained brain slice.
Figure 5. Figure 5. Image taken using intravital microscopy via a two‐photon microscope system through a surgically implanted chronic cranial window in mice depicting projections of astrocyte foot processes contacting or touching cerebral microvessels shown using FITC‐dextran, green.
Figure 6. Figure 6. An increase in intraluminal pressure from 10 to 100 mmHg induces depolarization of arterial muscle cell via reduction in outward K+ current through Ca2+‐activated K+ channel (KCa) and K+ efflux leading to regenerative electrical spike activity stimulated by an increase in inward current carried by voltage‐gated L‐type Ca2+ channel causing arterial myocyte constriction.
Figure 7. Figure 7. Resting potential (Em = mV, ordinate) as a function of external K+ concentration [K]o (Log scale) for vascular smooth muscle cells of cat basilar artery. The data are represented as mean ± SD for 10 to 16 cells in seven different preparations.
Figure 8. Figure 8. Changes in morphology of astrocytes and cerebral capillary endothelial cells when cocultured. (A) Formation of capillary‐like structures (arrow) double‐labeled with acetylated LDL labelled with 1,1′‐dioctadecyl‐1,3,3,3′,3′‐tetramethyl‐indocarbocyanine perchlorate (DiI‐Ac‐LDL; red) and glial fibrillary acidic protein (GFAP; green) in the coculture of astrocytes and cerebral capillary endothelial cells. (B) Formation of capillary‐like structures (arrow) triple labeled by platelet endothelial cell adhesion molecule‐1 (PECAM‐1; green), GFAP (red), and 4,6‐diamidino‐2‐phenylindole (DAPI; blue) in the coculture of astrocytes and cerebral capillary endothelial cells. Note the interaction between astrocytes and endothelial tubes (arrowheads in A and B). (C) Double immunolabeling of blood vessels and astrocytes with PECAM‐1 (green) and GFAP (red) in a section from normal rat cortex showing astrocytes from foot processes that impinge on vessels (arrows). (D) Cells (*) forming tube identified by DAPI staining (E) in coculture exhibit punctate staining of connexin‐43 on their cell‐cell borders (arrow). Arrow points to area shown in the inset at higher magnification. (F) cells (*) outside tubes in the same coculture as in D show moderate to light cytoplasmic staining of connexin‐43. Bar in A = 25 μm for A and B, bar = 20 μm in C, bar in F = 10 μm for D to F, and bar in inset of D = 5 μm. (Reproduced, with permission, from Zhang C and Harder DR, 2002, 237.)
Figure 9. Figure 9. Rhythmic CYP epoxygenase expression and activity in endothelial cells. (A) RT‐PCR was used to measure the rhythmic mRNA expression of Cyp2c11 in rat brain microvascular endothelial cells. (B) Formation of 11,12‐EET from AA metabolism by CYP 2C11 epoxygenase in cultured endothelial cells was measured by LC/MS analysis. Cosinor analysis was used to test for 12 and 24‐h period rhythms. Phase Φ and P values are indicated for each graph.
Figure 10. Figure 10. Schematic diagram of possible signaling mechanisms in astrocytes and vascular smooth muscle that result in cerebrovascular vasodilation during hypoxemia. ADO, adenosine; ARA, arachidonic acid; DAG, diacylglycerol; GLT1, primary astrocytic glutamate transporter; Glu, glutamate; IP3, inositol triphosphate; mGluR, metabotropic glutamate receptor; PLA2, phospholipase A2; PLC, phospholipase C; CYP, cytochrome P‐450; EET, epoxyeicosatrienoic acids; TRPV4, transient receptor potential vanilloid 4; KCa, calcium‐activated K+; KIR, inwardly rectifying K+ channel; 20‐HETE, 20‐hydroxyeicosatetraenoic acid. Dashed line indicates negative effect. (Reproduced, with permission, from Liu X et al., 2015, 144.)


Figure 1. Schematic representation of glucose metabolism. Glucose enters cells trough glucose transporters (GLUTs) and is phosphorylated by hexokinase (HK) to produce glucose‐6‐phosphate (glucose‐6P). Glucose‐6P can be processed into three main metabolic pathways. First, it can be metabolized through glycolysis (i), giving rise to two molecules of pyruvate and producing adenosine phosphate (ATP) and nicotinamide adenine dinucleotide (NADH). Pyruvate can then enter mitochondria, where it is metabolized through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (Ox Phos), producing ATP and CO2 while consuming oxygen. Pyruvate can otherwise be reduced to lactate‐by‐lactate dehydrogenase (LDH). This lactate can be released in the extracellular space through monocarboxylate transporters (MCTs). The complete oxidation of glucose produces larger amounts of energy in the form of ATP in the mitochondria (30‐34 ATP) compared to glycolysis (2 ATP). Alternatively, glucose‐6P can be processed through the pentose phosphate pathway (PPP) (ii), leading to the production of reducing equivalent in the form of nicotinamide adenine dinucleotide phosphate (NADPH). Note that the PPP and glycolysis are linked at the level of glyceraldehyde‐3‐phosphate (GA3P) and fructose‐6‐phosphate (fructose‐6P). Finally, in astrocytes, glucose‐6P can also be used to store glucosyl units as glycogen (iii). Abbreviations are as follows: GPI, glucose‐6‐phosphate isomerase; PFK, phosphofructokinase‐1; Fructose‐1,6‐P2, fructose‐1,6‐bisphosphate; DHAP, dihydroxyacetone phosphate; TPI, triose phosphate isomerase; G6PDH, glucose‐6‐phosphate dehydrogenase; 6‐PGL, 6‐phosphoglucono‐δ‐lactone; 6‐PG, 6‐phosphogluconate; 6 PGDH, 6‐phosphogluconate dehydrogenase; ribulose‐5P, ribulose‐5‐phosphate; ribose‐5P, ribose‐5‐phosphate; xylulose‐5P, xylulose‐5‐phosphate; TK, transketolase; sedoheptulose‐7P, sedoheptulose‐7‐phosphate; TA, transaldolase; and erythrose‐4P, erythrose‐4‐phosphate. (Reproduced, with permission, from Belanger M, Allaman I, and Magistretti PJ, 2011, 12.)


Figure 2. Dependence of brain activity on blood flow in response to initiation of different task performance as shown by functional MRI signals.


Figure 3. Schematic representation of a neurovascular unit with astrocytes being the central processor of neuronal signals as depicted in both panels A and panel B.


Figure 4. (A) Immunostaining depiction of components of the neurovascular unit (NVU). The astrocytes (stained with rhodamine labeled GFAP) shown in red. The neurons are stained with fluorescein tagged NSE shown in green and the blood vessels are stained with PECAM shown in blue. Note the location of the foot processes around the vasculature. (B) Histochemical localization of β‐galactosidase expression in rat brain following lateral ventricular infusion of Ad5/CMV‐β‐galactosidase (magnification × 1000). Note staining of astrocytes and astrocytic foot processes surrounding blood vessel emulating the exploded section of the immunostained brain slice.


Figure 5. Image taken using intravital microscopy via a two‐photon microscope system through a surgically implanted chronic cranial window in mice depicting projections of astrocyte foot processes contacting or touching cerebral microvessels shown using FITC‐dextran, green.


Figure 6. An increase in intraluminal pressure from 10 to 100 mmHg induces depolarization of arterial muscle cell via reduction in outward K+ current through Ca2+‐activated K+ channel (KCa) and K+ efflux leading to regenerative electrical spike activity stimulated by an increase in inward current carried by voltage‐gated L‐type Ca2+ channel causing arterial myocyte constriction.


Figure 7. Resting potential (Em = mV, ordinate) as a function of external K+ concentration [K]o (Log scale) for vascular smooth muscle cells of cat basilar artery. The data are represented as mean ± SD for 10 to 16 cells in seven different preparations.


Figure 8. Changes in morphology of astrocytes and cerebral capillary endothelial cells when cocultured. (A) Formation of capillary‐like structures (arrow) double‐labeled with acetylated LDL labelled with 1,1′‐dioctadecyl‐1,3,3,3′,3′‐tetramethyl‐indocarbocyanine perchlorate (DiI‐Ac‐LDL; red) and glial fibrillary acidic protein (GFAP; green) in the coculture of astrocytes and cerebral capillary endothelial cells. (B) Formation of capillary‐like structures (arrow) triple labeled by platelet endothelial cell adhesion molecule‐1 (PECAM‐1; green), GFAP (red), and 4,6‐diamidino‐2‐phenylindole (DAPI; blue) in the coculture of astrocytes and cerebral capillary endothelial cells. Note the interaction between astrocytes and endothelial tubes (arrowheads in A and B). (C) Double immunolabeling of blood vessels and astrocytes with PECAM‐1 (green) and GFAP (red) in a section from normal rat cortex showing astrocytes from foot processes that impinge on vessels (arrows). (D) Cells (*) forming tube identified by DAPI staining (E) in coculture exhibit punctate staining of connexin‐43 on their cell‐cell borders (arrow). Arrow points to area shown in the inset at higher magnification. (F) cells (*) outside tubes in the same coculture as in D show moderate to light cytoplasmic staining of connexin‐43. Bar in A = 25 μm for A and B, bar = 20 μm in C, bar in F = 10 μm for D to F, and bar in inset of D = 5 μm. (Reproduced, with permission, from Zhang C and Harder DR, 2002, 237.)


Figure 9. Rhythmic CYP epoxygenase expression and activity in endothelial cells. (A) RT‐PCR was used to measure the rhythmic mRNA expression of Cyp2c11 in rat brain microvascular endothelial cells. (B) Formation of 11,12‐EET from AA metabolism by CYP 2C11 epoxygenase in cultured endothelial cells was measured by LC/MS analysis. Cosinor analysis was used to test for 12 and 24‐h period rhythms. Phase Φ and P values are indicated for each graph.


Figure 10. Schematic diagram of possible signaling mechanisms in astrocytes and vascular smooth muscle that result in cerebrovascular vasodilation during hypoxemia. ADO, adenosine; ARA, arachidonic acid; DAG, diacylglycerol; GLT1, primary astrocytic glutamate transporter; Glu, glutamate; IP3, inositol triphosphate; mGluR, metabotropic glutamate receptor; PLA2, phospholipase A2; PLC, phospholipase C; CYP, cytochrome P‐450; EET, epoxyeicosatrienoic acids; TRPV4, transient receptor potential vanilloid 4; KCa, calcium‐activated K+; KIR, inwardly rectifying K+ channel; 20‐HETE, 20‐hydroxyeicosatetraenoic acid. Dashed line indicates negative effect. (Reproduced, with permission, from Liu X et al., 2015, 144.)
References
 1.Abbott NJ. Astrocyte‐endothelial interactions and blood‐brain barrier permeability. J Anat 200(2): 629‐638, 2002.
 2.Ainslie PN, Murrell C, Peebles K, Swart M, Skinner MA, Williams MJ, Taylor RD. Early morning impairment in cerebral autoregulation and cerebrovascular CO2 reactivity in healthy humans: Relation to endothelial function. Exp Physiol 92(2): 769‐777, 2007.
 3.Alkayed NJ, Birks EK, Narayanan J, Petrie KA, Kohler‐Cabot AE, Harder DR. Role of P‐450 arachidonic acid epoxygenase in the response of cerebral blood flow to glutamate in rats. Stroke 28(2): 1066‐1072, 1997.
 4.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(2): 971‐979, 1996.
 5.Amruthesh SC, Falck JR, Ellis EF. Brain synthesis and cerebrovascular action of epoxygenase metabolites of arachidonic acid. J Neurochem 58(2): 503‐510, 1992.
 6.Andreotti F, Davies GJ, Hackett DR, Khan MI, De Bart AC, Aber VR, Maseri A, Kluft C. Major circadian fluctuations in fibrinolytic factors and possible relevance to time of onset of myocardial infarction, sudden cardiac death and stroke. Am J Cardiol 62(2): 635‐637, 1988.
 7.Bachetti T, Comini L, Curello S, Bastianon D, Palmieri M, Bresciani G, Callea F, Ferrari R. Co‐expression and modulation of neuronal and endothelial nitric oxide synthase in human endothelial cells. J Mol Cell Cardiol 37(2): 939‐945, 2004.
 8.Bain AR, Nybo L, Ainslie PN. Cerebral vascular control and metabolism in heat stress. Compr Physiol 5(2): 1345‐1380, 2015.
 9.Ballabh P, Braun A, Nedergaard M. The blood‐brain barrier: An overview: Structure, regulation, and clinical implications. Neurobiol Dis 16(2): 1‐13, 2004.
 10.Barry DI. Cerebral blood flow in hypertension. J Cardiovasc Pharmacol 7(Suppl 2): S94‐S98, 1985.
 11.Bayliss WM. On the local reactions of the arterial wall to changes of internal pressure. J Physiol 28(2): 220‐231, 1902.
 12.Belanger M, Allaman I, Magistretti PJ. Brain energy metabolism: Focus on astrocyte‐neuron metabolic cooperation. Cell Metab 14(2): 724‐738, 2011.
 13.Bergers G, Song S. The role of pericytes in blood‐vessel formation and maintenance. Neuro Oncol 7(2): 452‐464, 2005.
 14.Bogatcheva NV, Sergeeva MG, Dudek SM, Verin AD. Arachidonic acid cascade in endothelial pathobiology. Microvasc Res 69(2): 107‐127, 2005.
 15.Boison D. Adenosine dysfunction in epilepsy. Glia 60(2): 1234‐1243, 2012.
 16.Borsook D, Maleki N, Burstein R. Migraine. Academic Press, Elsevier Science: London, 2015, p. 708.
 17.Brickman AM, Guzman VA, Gonzalez‐Castellon M, Razlighi Q, Gu Y, Narkhede A, Janicki S, Ichise M, Stern Y, Manly JJ, Schupf N, Marshall RS. Cerebral autoregulation, beta amyloid, and white matter hyperintensities are interrelated. Neurosci Lett 592: 54‐58, 2015.
 18.Bylund J, Zhang C, Harder DR. Identification of a novel cytochrome P450, CYP4X1, with unique localization specific to the brain. Biochem Biophys Res Commun 296(2): 677‐684, 2002.
 19.Campbell WB, Gebremedhin D, Pratt PF, Harder DR. Identification of epoxyeicosatrienoic acids as endothelium‐derived hyperpolarizing factors. Circ Res 78(2): 415‐423, 1996.
 20.Capdevila JH, Dishman E, Karara A, Falck JR. Cytochrome P450 arachidonic acid epoxygenase: Stereochemical characterization of epoxyeicosatrienoic acids. Methods Enzymol 206: 441‐453, 1991.
 21.Capettini LS, Cortes SF, Gomes MA, Silva GA, Pesquero JL, Lopes MJ, Teixeira MM, Lemos VS. Neuronal nitric oxide synthase‐derived hydrogen peroxide is a major endothelium‐dependent relaxing factor. Am J Physiol Heart Circ Physiol 295(2): H2503‐H2511, 2008.
 22.Carrier GO, Fuchs LC, Winecoff AP, Giulumian AD, White RE. Nitrovasodilators relax mesenteric microvessels by cGMP‐induced stimulation of Ca‐activated K channels. Am J Physiol 273(1 Pt 2): H76‐H84, 1997.
 23.Carver KA, Lourim D, Tryba AK, Harder DR. Rhythmic expression of cytochrome P450 epoxygenases CYP4x1 and CYP2c11 in the rat brain and vasculature. Am J Physiol Cell Physiol 307(2): C989‐C998, 2014.
 24.Cauli B, Hamel E. Revisiting the role of neurons in neurovascular coupling. Front Neuroenergetics 2: 9, 2010.
 25.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(2): 8940‐8949, 2004.
 26.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(2): 8940‐8949, 2004.
 27.Chaturvedi S, Adams HP, Jr, Woolson RF. Circadian variation in ischemic stroke subtypes. Stroke 30(2): 1792‐1795, 1999.
 28.Chen H, Chan DC. Mitochondrial dynamics‐–fusion, fission, movement, and mitophagy–‐in neurodegenerative diseases. Hum Mol Genet 18(R2): R169‐R1676, 2009.
 29.Chen H, Chomyn A, Chan DC. Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem 280(2): 26185‐26192, 2005.
 30.Cooke‐Ariel H. Circadian variations in cardiovascular function and their relation to the occurrence and timing of cardiac events. Am J Health Syst Pharm 55(Suppl 3): S5‐S11, 1998.
 31.Dalkara T, Alarcon‐Martinez L. Cerebral microvascular pericytes and neurogliovascular signaling in health and disease. Brain Res 1623: 3‐17, 2015.
 32.de Eulate RG, Goni I, Galiano A, Vidorreta M, Recio M, Riverol M, Zubieta JL, Fernandez‐Seara MA. Reduced cerebral blood flow in mild cognitive impairment assessed using phase‐contrast MRI. J Alzheimers Dis 58: 585‐595, 2017.
 33.Dehouck MP, Meresse S, Delorme P, Fruchart JC, Cecchelli R. An easier, reproducible, and mass‐production method to study the blood‐brain barrier in vitro. J Neurochem 54(2): 1798‐1801, 1990.
 34.Detmer SA, Chan DC. Functions and dysfunctions of mitochondrial dynamics. Nat Rev Mol Cell Biol 8(2): 870‐879, 2007.
 35.Devonshire IM, Papadakis NG, Port M, Berwick J, Kennerley AJ, Mayhew JE, Overton PG. Neurovascular coupling is brain region‐dependent. Neuroimage 59(2): 1997‐2006, 2012.
 36.Dey P, Velazquez‐Villegas LA, Faria M, Turner A, Jonsson P, Webb P, Williams C, Gustafsson JA, Strom AM. Estrogen receptor beta2 induces hypoxia signature of gene expression by stabilizing HIF‐1alpha in prostate cancer. PLoS One 10(2): e0128239, 2015.
 37.Di Marco LY, Venneri A, Farkas E, Evans PC, Marzo A, Frangi AF. Vascular dysfunction in the pathogenesis of Alzheimer's disease–‐a review of endothelium‐mediated mechanisms and ensuing vicious circles. Neurobiol Dis 82: 593‐606, 2015.
 38.Dong DL, Yue P, Yang BF, Wang WH. Hydrogen peroxide stimulates the Ca(2+)‐activated big‐conductance K channels (BK) through cGMP signaling pathway in cultured human endothelial cells. Cell Physiol Biochem 22(1‐4): 119‐126, 2008.
 39.Dora KA, Garland CJ, Kwan HY, Yao X. Endothelial cell protein kinase G inhibits release of EDHF through a PKG‐sensitive cation channel. Am J Physiol Heart Circ Physiol 280(2): H1272‐H1277, 2001.
 40.Dowell JA, Johnson JA, Li L. Identification of astrocyte secreted proteins with a combination of shotgun proteomics and bioinformatics. J Proteome Res 8(2): 4135‐4143, 2009.
 41.Dumpich M, Mannherz HG, Theiss C. VEGF signaling regulates cofilin and the Arp2/3‐complex within the axonal growth cone. Curr Neurovasc Res 12(2): 293‐307, 2015.
 42.Earley S, Straub SV, Brayden JE. Protein kinase C regulates vascular myogenic tone through activation of TRPM4. Am J Physiol Heart Circ Physiol 292(2): H2613‐H2622, 2007.
 43.Elliot WJ. Cyclic and circadian variations in cardiovascular events. Am J Hypertens 14(9 Pt 2): 291S‐295S, 2001.
 44.Ellis EF, Police RJ, Yancey L, McKinney JS, Amruthesh SC. Dilation of cerebral arterioles by cytochrome P‐450 metabolites of arachidonic acid. Am J Physiol 259(4 Pt 2): H1171‐H1177, 1990.
 45.Elshenawy OH, Shoieb SM, Mohamed A, El‐Kadi AO. Clinical implications of 20‐hydroxyeicosatetraenoic acid in the kidney, liver, lung and brain: An emerging therapeutic target. Pharmaceutics 9(1): 9, 2017.
 46.El‐Sikhry HE, Alsaleh N, Dakarapu R, Falck JR, Seubert JM. Novel roles of epoxyeicosanoids in regulating cardiac mitochondria. PLoS One 11(2): e0160380, 2016.
 47.Elwood PC, Beswick A, Pickering J, McCarron P, O'Brien JR, Renaud SR, Flower RJ. Platelet tests in the prediction of myocardial infarction and ischaemic stroke: Evidence from the caerphilly prospective study. Br J Haematol 113(2): 514‐520, 2001.
 48.El‐Yazbi AF, Abd‐Elrahman KS, Moreno‐Dominguez A. PKC‐mediated cerebral vasoconstriction: Role of myosin light chain phosphorylation versus actin cytoskeleton reorganization. Biochem Pharmacol 95(2): 263‐278, 2015.
 49.Escobar‐Henriques M, Anton F. Mechanistic perspective of mitochondrial fusion: Tubulation vs. fragmentation. Biochim Biophys Acta 1833(2): 162‐175, 2013.
 50.Etsuda H, Takase B, Uehata A, Kusano H, Hamabe A, Kuhara R, Akima T, Matsushima Y, Arakawa K, Satomura K, Kurita A, Ohsuzu F. Morning attenuation of endothelium‐dependent, flow‐mediated dilation in healthy young men: Possible connection to morning peak of cardiac events? Clin Cardiol 22(2): 417‐421, 1999.
 51.Ezan P, Andre P, Cisternino S, Saubamea B, Boulay AC, Doutremer S, Thomas MA, Quenech'du N, Giaume C, Cohen‐Salmon M. Deletion of astroglial connexins weakens the blood‐brain barrier. J Cereb Blood Flow Metab 32(2): 1457‐1467, 2012.
 52.Falck B, Nielsen KC, Owman C. Adrenergic innervation of the pial circulation. Scand J Clin Lab Invest Suppl 102(VI): B, 1968.
 53.Faraci FM, Baumbach GL, Heistad DD. Myogenic mechanisms in the cerebral circulation. J Hypertens Suppl 7(2): S61‐S64; discussion S65, 1989.
 54.Faraci FM, Mayhan WG, Werber AH, Heistad DD. Cerebral circulation: Effects of sympathetic nerves and protective mechanisms during hypertension. Circ Res 61(5 Pt 2): II102‐II106, 1987.
 55.Feletou M, Vanhoutte PM. Endothelium‐derived hyperpolarizing factor: Where are we now? Arterioscler Thromb Vasc Biol 26(2): 1215‐1225, 2006.
 56.Ferrara N. The role of vascular endothelial growth factor in pathological angiogenesis. Breast Cancer Res Treat 36(2): 127‐137, 1995.
 57.Ferrara N, Heinsohn H, Walder CE, Bunting S, Thomas GR. The regulation of blood vessel growth by vascular endothelial growth factor. Ann N Y Acad Sci 752: 246‐256, 1995.
 58.Figueroa XF, Duling BR. Gap junctions in the control of vascular function. Antioxid Redox Signal 11(2): 251‐266, 2009.
 59.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(2): 1397‐1403, 2006.
 60.Filosa JA, Iddings JA. Astrocyte regulation of cerebral vascular tone. Am J Physiol Heart Circ Physiol 305(2): H609‐H619, 2013.
 61.Filosa JA, Morrison HW, Iddings JA, Du W, Kim KJ. Beyond neurovascular coupling, role of astrocytes in the regulation of vascular tone. Neuroscience 323: 96‐109, 2016.
 62.Filosa JA, Morrison HW, Iddings JA, Du W, Kim KJ. Beyond neurovascular coupling, role of astrocytes in the regulation of vascular tone. Neuroscience 323: 96‐109, 2016.
 63.Fisslthaler B, Michaelis UR, Randriamboavonjy V, Busse R, Fleming I. Cytochrome P450 epoxygenases and vascular tone: Novel role for HMG‐CoA reductase inhibitors in the regulation of CYP 2C expression. Biochim Biophys Acta 1619(2): 332‐339, 2003.
 64.Fleming I. Cytochrome p450 and vascular homeostasis. Circ Res 89(2): 753‐762, 2001.
 65.Fleming I, Michaelis UR, Bredenkotter D, Fisslthaler B, Dehghani F, Brandes RP, Busse R. Endothelium‐derived hyperpolarizing factor synthase (Cytochrome P450 2C9) is a functionally significant source of reactive oxygen species in coronary arteries. Circ Res 88(2): 44‐51, 2001.
 66.Fogelholm RR, Turjanmaa VM, Nuutila MT, Murros KE, Sarna S. Diurnal blood pressure variations and onset of subarachnoid haemorrhage: A population‐based study. J Hypertens 13(2): 495‐498, 1995.
 67.Fox PT, Raichle ME. Focal physiological uncoupling of cerebral blood flow and oxidative metabolism during somatosensory stimulation in human subjects. Proc Natl Acad Sci U S A 83(2): 1140‐1144, 1986.
 68.Fox PT, Raichle ME, Mintun MA, Dence C. Nonoxidative glucose consumption during focal physiologic neural activity. Science 241(2): 462‐464, 1988.
 69.Furchgott RF, Vanhoutte PM. Endothelium‐derived relaxing and contracting factors. FASEB J 3(2): 2007‐2018, 1989.
 70.Gebremedhin D, Bonnet P, Greene AS, England SK, Rusch NJ, Lombard JH, Harder DR. Hypoxia increases the activity of Ca(2+)‐sensitive K+ channels in cat cerebral arterial muscle cell membranes. Pflugers Arch 428(5‐6): 621‐630, 1994.
 71.Gebremedhin D, Gopalakrishnan S, Harder DR. Endogenous events modulating myogenic regulation of cerebrovascular function. Curr Vasc Pharmacol 12(2): 810‐817, 2014.
 72.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 263(2 Pt 2): H519‐H525, 1992.
 73.Gebremedhin D, Weinberger B, Lourim D, Harder DR. Adenosine can mediate its actions through generation of reactive oxygen species. J Cereb Blood Flow Metab 30(2): 1777‐1790, 2010.
 74.Gebremedhin D, Yamaura K, Harder DR. Role of 20‐HETE in the hypoxia‐induced activation of Ca2+‐activated K+ channel currents in rat cerebral arterial muscle cells. Am J Physiol Heart Circ Physiol 294(2): H107‐H120, 2008.
 75.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(2): 1678‐1687, 2003.
 76.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.
 77.Gegg ME, Beltran B, Salas‐Pino S, Bolanos JP, Clark JB, Moncada S, Heales SJ. Differential effect of nitric oxide on glutathione metabolism and mitochondrial function in astrocytes and neurones: Implications for neuroprotection/neurodegeneration? J Neurochem 86(2): 228‐237, 2003.
 78.Gerhardt H, Betsholtz C. Endothelial‐pericyte interactions in angiogenesis. Cell Tissue Res 314(2): 15‐23, 2003.
 79.Girouard H, Iadecola C. Neurovascular coupling in the normal brain and in hypertension, stroke, and Alzheimer disease. J Appl Physiol (1985) 100(2): 328‐335, 2006.
 80.Girouard H, Wang G, Gallo EF, Anrather J, Zhou P, Pickel VM, Iadecola C. NMDA receptor activation increases free radical production through nitric oxide and NOX2. J Neurosci 29(2): 2545‐2552, 2009.
 81.Gjedde A, Marrett S, Vafaee M. Oxidative and nonoxidative metabolism of excited neurons and astrocytes. J Cereb Blood Flow Metab 22(2): 1‐14, 2002.
 82.Gomes CV, Kaster MP, Tome AR, Agostinho PM, Cunha RA. Adenosine receptors and brain diseases: Neuroprotection and neurodegeneration. Biochim Biophys Acta 1808(2): 1380‐1399, 2011.
 83.Gonzales AL, Earley S. Regulation of cerebral artery smooth muscle membrane potential by Ca(2)(+)‐activated cation channels. Microcirculation 20(2): 337‐347, 2013.
 84.Guengerich FP, Munro AW. Unusual cytochrome p450 enzymes and reactions. J Biol Chem 288(2): 17065‐17073, 2013.
 85.Halliday MR, Rege SV, Ma Q, Zhao Z, Miller CA, Winkler EA, Zlokovic BV. Accelerated pericyte degeneration and blood‐brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer's disease. J Cereb Blood Flow Metab 36(2): 216‐227, 2016.
 86.Halpern W, Osol G. Influence of transmural pressure of myogenic responses of isolated cerebral arteries of the rat. Ann Biomed Eng 13(3‐4): 287‐293, 1985.
 87.Hamel E. Perivascular nerves and the regulation of cerebrovascular tone. J Appl Physiol (1985) 100(2): 1059‐1064, 2006.
 88.Hamel E. Cholinergic modulation of the cortical microvascular bed. Prog Brain Res 145: 171‐178, 2004.
 89.Hamner JW, Tan CO. Relative contributions of sympathetic, cholinergic, and myogenic mechanisms to cerebral autoregulation. Stroke 45(2): 1771‐1777, 2014.
 90.Harder DR. Comparison of electrical properties of middle cerebral and mesenteric artery in cat. Am J Physiol 239(2): C23‐C26, 1980.
 91.Harder DR. Pressure‐dependent membrane depolarization in cat middle cerebral artery. Circ Res 55(2): 197‐202, 1984.
 92.Harder DR. A cellular mechanism for myogenic regulation of cat cerebral arteries. Ann Biomed Eng 13(3‐4): 335‐339, 1985.
 93.Harder DR, Alkayed NJ, Lange AR, Gebremedhin D, Roman RJ. Functional hyperemia in the brain: Hypothesis for astrocyte‐derived vasodilator metabolites. Stroke 29(2): 229‐234, 1998.
 94.Harder DR, Roman RJ, Gebremedhin D. Molecular mechanisms controlling nutritive blood flow: Role of cytochrome P450 enzymes. Acta Physiol Scand 168(2): 543‐549, 2000.
 95.Harder DR, Zhang C, Gebremedhin D. Astrocytes function in matching blood flow to metabolic activity. News Physiol Sci 17: 27‐31, 2002.
 96.Harper SL, Bohlen HG. Microvascular adaptation in the cerebral cortex of adult spontaneously hypertensive rats. Hypertension 6(2): 408‐419, 1984.
 97.Hawkins VE, Takakura AC, Trinh A, Malheiros‐Lima MR, Cleary CM, Wenker IC, Dubreuil T, Rodriguez EM, Nelson MT, Moreira TS, Mulkey DK. Purinergic regulation of vascular tone in the retrotrapezoid nucleus is specialized to support the drive to breathe. Elife 6: pii: e25232, 2017.
 98.Hayakawa K, Esposito E, Wang X, Terasaki Y, Liu Y, Xing C, Ji X, Lo EH. Transfer of mitochondria from astrocytes to neurons after stroke. Nature 535(2): 551‐555, 2016.
 99.Hayakawa T, McGarrigle CA, Coen RF, Soraghan CJ, Foran T, Lawlor BA, Kenny RA. Orthostatic blood pressure behavior in people with mild cognitive impairment predicts conversion to dementia. J Am Geriatr Soc 63(2): 1868‐1873, 2015.
 100.Hayashi Y, Nomura M, Yamagishi S, Harada S, Yamashita J, Yamamoto H. Induction of various blood‐brain barrier properties in non‐neural endothelial cells by close apposition to co‐cultured astrocytes. Glia 19(2): 13‐26, 1997.
 101.Hein TW, Xu W, Ren Y, Kuo L. Cellular signalling pathways mediating dilation of porcine pial arterioles to adenosine A(2)A receptor activation. Cardiovasc Res 99(2): 156‐163, 2013.
 102.Heistad DD, Marcus ML. Evidence that neural mechanisms do not have important effects on cerebral blood flow. Circ Res 42(2): 295‐302, 1978.
 103.Heistad DD, Marcus ML, Abboud FM. Effects of sympathetic nerve stimulation on cerebral blood flow. Acta Neurol Scand Suppl 64: 306‐307, 1977.
 104.Heistad DD, Marcus ML, Gross PM. Effects of sympathetic nerves on cerebral vessels in dog, cat, and monkey. Am J Physiol 235(2): H544‐H552, 1978.
 105.Hellstrom M, Kalen M, Lindahl P, Abramsson A, Betsholtz C. Role of PDGF‐B and PDGFR‐beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development 126(2): 3047‐3055, 1999.
 106.Hermsmeyer K. Excitation of vascular muscles by norepinephrine. Ann Biomed Eng 11(2): 567‐577, 1983.
 107.Hertz L. Intercellular metabolic compartmentation in the brain: past, present and future. Neurochem Int 45(2‐3): 285‐296, 2004.
 108.Hertz L. Astrocytic energy metabolism and glutamate formation–‐relevance for 13C‐NMR spectroscopy and importance of cytosolic/mitochondrial trafficking. Magn Reson Imaging 29(2): 1319‐1329, 2011.
 109.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(2): C1068‐C1078, 2010.
 110.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(2): 95‐110, 2015.
 111.Himmels P, Paredes I, Adler H, Karakatsani A, Luck R, Marti HH, Ermakova O, Rempel E, Stoeckli ET, Ruiz de Almodovar C. Motor neurons control blood vessel patterning in the developing spinal cord. Nat Commun 8: 14583, 2017.
 112.Hong KW, Shin HK, Kim CD, Lee WS, Rhim BY. Restoration of vasodilation and CBF autoregulation by genistein in rat pial artery after brain injury. Am J Physiol Heart Circ Physiol 281(2): H308‐H315, 2001.
 113.Hotta H, Uchida S, Kagitani F, Maruyama N. Control of cerebral cortical blood flow by stimulation of basal forebrain cholinergic areas in mice. J Physiol Sci 61(2): 201‐209, 2011.
 114.Iadecola C. The neurovascular unit coming of age: A journey through neurovascular coupling in health and disease. Neuron 96(2): 17‐42, 2017.
 115.Iliff JJ, Close LN, Selden NR, Alkayed NJ. A novel role for P450 eicosanoids in the neurogenic control of cerebral blood flow in the rat. Exp Physiol 92(2): 653‐658, 2007.
 116.Iliff JJ, Fairbanks SL, Balkowiec A, Alkayed NJ. Epoxyeicosatrienoic acids are endogenous regulators of vasoactive neuropeptide release from trigeminal ganglion neurons. J Neurochem 115(2): 1530‐1542, 2010.
 117.Iliff JJ, Wang R, Zeldin DC, Alkayed NJ. Epoxyeicosanoids as mediators of neurogenic vasodilation in cerebral vessels. Am J Physiol Heart Circ Physiol 296(2): H1352‐H1363, 2009.
 118.Itoh T, Kuriyama H, Suzuki H. Differences and similarities in the noradrenaline‐ and caffeine‐induced mechanical responses in the rabbit mesenteric artery. J Physiol 337: 609‐629, 1983.
 119.Iwayama T, Furness JB, Burnstock G. Dual adrenergic and cholinergic innervation of the cerebral arteries of the rat. An ultrastructural study. Circ Res 26(2): 635‐646, 1970.
 120.Jensen LD, Nakamura M, Brautigam L, Li X, Liu Y, Samani NJ, Cao Y. VEGF‐B‐Neuropilin‐1 signaling is spatiotemporally indispensable for vascular and neuronal development in zebrafish. Proc Natl Acad Sci U S A 112(2): E5944‐E5953, 2015.
 121.Jeric M, Vukojevic K, Vuica A, Filipovic N. Diabetes mellitus influences the expression of NPY and VEGF in neurons of rat trigeminal ganglion. Neuropeptides 62: 57‐64, 2017.
 122.Jha MK, Seo M, Kim JH, Kim BG, Cho JY, Suk K. The secretome signature of reactive glial cells and its pathological implications. Biochim Biophys Acta 1834(2): 2418‐2428, 2013.
 123.Kacem K, Lacombe P, Seylaz J, Bonvento G. Structural organization of the perivascular astrocyte endfeet and their relationship with the endothelial glucose transporter: A confocal microscopy study. Glia 23(2): 1‐10, 1998.
 124.Kario K, Yano Y, Matsuo T, Hoshide S, Asada Y, Shimada K. Morning blood pressure surge, morning platelet aggregation, and silent cerebral infarction in older Japanese hypertensive patients. J Hypertens 29(2): 2433‐2439, 2011.
 125.Khakh BS, McCarthy KD. Astrocyte calcium signaling: From observations to functions and the challenges therein. Cold Spring Harb Perspect Biol 7(2): a020404, 2015.
 126.Khakh BS, Sofroniew MV. Diversity of astrocyte functions and phenotypes in neural circuits. Nat Neurosci 18(2): 942‐952, 2015.
 127.Kitazono T, Faraci FM, Taguchi H, Heistad DD. Role of potassium channels in cerebral blood vessels. Stroke 26(2): 1713‐1723, 1995.
 128.Knot HJ, Zimmermann PA, Nelson MT. Extracellular K(+)‐induced hyperpolarizations and dilatations of rat coronary and cerebral arteries involve inward rectifier K(+) channels. J Physiol 492(Pt 2): 419‐430, 1996.
 129.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(2): 221‐231, 2008.
 130.Koehler RC, Roman RJ, Harder DR. Astrocytes and the regulation of cerebral blood flow. Trends Neurosci 32(2): 160‐169, 2009.
 131.Lagaud G, Gaudreault N, Moore ED, Van Breemen C, Laher I. Pressure‐dependent myogenic constriction of cerebral arteries occurs independently of voltage‐dependent activation. Am J Physiol Heart Circ Physiol 283(2): H2187‐H2195, 2002.
 132.Lam AM, Artru AA. Autoregulation of cerebral blood flow in response to adenosine‐induced hypotension in dogs. J Neurosurg Anesthesiol 4(2): 120‐127, 1992.
 133.Laterra J, Goldstein GW. Astroglial‐induced in vitro angiogenesis: Requirements for RNA and protein synthesis. J Neurochem 57(2): 1231‐1239, 1991.
 134.Laterra J, Guerin C, Goldstein GW. Astrocytes induce neural microvascular endothelial cells to form capillary‐like structures in vitro. J Cell Physiol 144(2): 204‐215, 1990.
 135.Lecrux C, Kocharyan A, Sandoe CH, Tong XK, Hamel E. Pyramidal cells and cytochrome P450 epoxygenase products in the neurovascular coupling response to basal forebrain cholinergic input. J Cereb Blood Flow Metab 32(2): 896‐906, 2012.
 136.Leffler CW, Fedinec AL. Newborn piglet cerebral microvascular responses to epoxyeicosatrienoic acids. Am J Physiol 273(1 Pt 2): H333‐H338, 1997.
 137.Legros F, Lombes A, Frachon P, Rojo M. Mitochondrial fusion in human cells is efficient, requires the inner membrane potential, and is mediated by mitofusins. Mol Biol Cell 13(2): 4343‐4354, 2002.
 138.Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443(2): 787‐795, 2006.
 139.Lindblom P, Gerhardt H, Liebner S, Abramsson A, Enge M, Hellstrom M, Backstrom G, Fredriksson S, Landegren U, Nystrom HC, Bergstrom G, Dejana E, Ostman A, Lindahl P, Betsholtz C. Endothelial PDGF‐B retention is required for proper investment of pericytes in the microvessel wall. Genes Dev 17(2): 1835‐1840, 2003.
 140.Liu D, Chan SL, de Souza‐Pinto NC, Slevin JR, Wersto RP, Zhan M, Mustafa K, de Cabo R, Mattson MP. Mitochondrial UCP4 mediates an adaptive shift in energy metabolism and increases the resistance of neurons to metabolic and oxidative stress. Neuromolecular Med 8(2): 389‐414, 2006.
 141.Liu M, Alkayed NJ. Hypoxic preconditioning and tolerance via hypoxia inducible factor (HIF) 1alpha‐linked induction of P450 2C11 epoxygenase in astrocytes. J Cereb Blood Flow Metab 25(2): 939‐948, 2005.
 142.Liu M, Zhu Q, Wu J, Yu X, Hu M, Xie X, Yang Z, Yang J, Feng YQ, Yue J. Glutamate affects the production of epoxyeicosanoids within the brain: The up‐regulation of brain CYP2J through the MAPK‐CREB signaling pathway. Toxicology 381: 31‐38, 2017.
 143.Liu X, Davis CM, Alkayed NJ. P450 eicosanoids and reactive oxygen species interplay in brain injury and neuroprotection. Antioxid Redox Signal 2017 [Epub ahead of print].
 144.Liu X, Gebremedhin D, Harder DR, Koehler RC. Contribution of epoxyeicosatrienoic acids to the cerebral blood flow response to hypoxemia. J Appl Physiol (1985) 119(2): 1202‐1209, 2015.
 145.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(2): H373‐H381, 2011.
 146.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(2): H373‐H381, 2011.
 147.Liu Y, Wan Y, Fang Y, Yao E, Xu S, Ning Q, Zhang G, Wang W, Huang X, Xie M. Epoxyeicosanoid signaling provides multi‐target protective effects on neurovascular unit in rats after focal ischemia. J Mol Neurosci 58(2): 254‐265, 2016.
 148.Longden TA, Dabertrand F, Koide M, Gonzales AL, Tykocki NR, Brayden JE, Hill‐Eubanks D, Nelson MT. Capillary K+‐sensing initiates retrograde hyperpolarization to increase local cerebral blood flow. Nat Neurosci 20(2): 717‐726, 2017.
 149.Ly JD, Grubb DR, Lawen A. The mitochondrial membrane potential (deltapsi(m)) in apoptosis; an update. Apoptosis 8(2): 115‐128, 2003.
 150.Ma Y, Zhang P, Li J, Lu J, Ge J, Zhao Z, Ma X, Wan S, Yao X, Shen B. Epoxyeicosatrienoic acids act through TRPV4‐TRPC1‐KCa1.1 complex to induce smooth muscle membrane hyperpolarization and relaxation in human internal mammary arteries. Biochim Biophys Acta 1852(2): 552‐559, 2015.
 151.Ma Y, Zhao P, Zhu J, Yan C, Li L, Zhang H, Zhang M, Gao X, Fan X. Naoxintong protects primary neurons from oxygen‐glucose deprivation/reoxygenation induced injury through PI3K‐Akt signaling pathway. Evid Based Complement Alternat Med 2016: 5815946, 2016.
 152.Mackenzie F, Ruhrberg C. Diverse roles for VEGF‐A in the nervous system. Development 139(2): 1371‐1380, 2012.
 153.MacVicar BA, Newman EA. Astrocyte regulation of blood flow in the brain. Cold Spring Harb Perspect Biol 7(5): pii: a020388, 2015.
 154.Magistretti PJ. Neuron‐glia metabolic coupling and plasticity. Exp Physiol 96(2): 407‐410, 2011.
 155.Magistretti PJ, Pellerin L. Cellular bases of brain energy metabolism and their relevance to functional brain imaging: Evidence for a prominent role of astrocytes. Cereb Cortex 6(2): 50‐61, 1996.
 156.Magistretti PJ, Pellerin L. Cellular mechanisms of brain energy metabolism. Relevance to functional brain imaging and to neurodegenerative disorders. Ann N Y Acad Sci 777: 380‐387, 1996.
 157.Mani N, Khaibullina A, Krum JM, Rosenstein JM. Astrocyte growth effects of vascular endothelial growth factor (VEGF) application to perinatal neocortical explants: Receptor mediation and signal transduction pathways. Exp Neurol 192(2): 394‐406, 2005.
 158.Mani N, Khaibullina A, Krum JM, Rosenstein JM. Vascular endothelial growth factor enhances migration of astroglial cells in subventricular zone neurosphere cultures. J Neurosci Res 88(2): 248‐257, 2010.
 159.Marler JR, Price TR, Clark GL, Muller JE, Robertson T, Mohr JP, Hier DB, Wolf PA, Caplan LR, Foulkes MA. Morning increase in onset of ischemic stroke. Stroke 20(2): 473‐476, 1989.
 160.Marowsky A, Burgener J, Falck JR, Fritschy JM, Arand M. Distribution of soluble and microsomal epoxide hydrolase in the mouse brain and its contribution to cerebral epoxyeicosatrienoic acid metabolism. Neuroscience 163(2): 646‐661, 2009.
 161.Maurya AK, Vinayak M. Quercetin Attenuates Cell Survival, Inflammation, and Angiogenesis via Modulation of AKT Signaling in Murine T‐Cell Lymphoma. Nutr Cancer 69(2): 470‐480, 2017.
 162.Mayhan WG. VEGF increases permeability of the blood‐brain barrier via a nitric oxide synthase/cGMP‐dependent pathway. Am J Physiol 276(5 Pt 1): C1148‐C1153, 1999.
 163.McNamara P, Seo SB, Rudic RD, Sehgal A, Chakravarti D, FitzGerald GA. Regulation of CLOCK and MOP4 by nuclear hormone receptors in the vasculature: A humoral mechanism to reset a peripheral clock. Cell 105(2): 877‐889, 2001.
 164.Medhora M, Narayanan J, Harder D. Dual regulation of the cerebral microvasculature by epoxyeicosatrienoic acids. Trends Cardiovasc Med 11(2): 38‐42, 2001.
 165.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(2): 1619‐1627, 2016.
 166.Munoz MF, Puebla M, Figueroa XF. Control of the neurovascular coupling by nitric oxide‐dependent regulation of astrocytic Ca(2+) signaling. Front Cell Neurosci 9: 59, 2015.
 167.Munzenmaier DH, Harder DR. Cerebral microvascular endothelial cell tube formation: Role of astrocytic epoxyeicosatrienoic acid release. Am J Physiol Heart Circ Physiol 278(2): H1163‐H1167, 2000.
 168.Murakami M, Kudo I. Phospholipase A2. J Biochem 131(2): 285‐292, 2002.
 169.Murfee WL, Skalak TC, Peirce SM. Differential arterial/venous expression of NG2 proteoglycan in perivascular cells along microvessels: Identifying a venule‐specific phenotype. Microcirculation 12(2): 151‐160, 2005.
 170.Nakamura Y, Ohya Y, Abe I, Fujishima M. Sodium‐potassium pump current in smooth muscle cells from mesenteric resistance arteries of the guinea‐pig. J Physiol 519(Pt 1): 203‐212, 1999.
 171.Navarrete M, Araque A. The Cajal school and the physiological role of astrocytes: A way of thinking. Front Neuroanat 8: 33, 2014.
 172.Nehls V, Drenckhahn D. The versatility of microvascular pericytes: From mesenchyme to smooth muscle? Histochemistry 99(2): 1‐12, 1993.
 173.Neild TO, Kotecha N. Relation between membrane potential and contractile force in smooth muscle of the rat tail artery during stimulation by norepinephrine, 5‐hydroxytryptamine, and potassium. Circ Res 60(2): 791‐795, 1987.
 174.Nelson MT, Brayden JE. Regulation of arterial tone by calcium‐dependent K+ channels and ATP‐sensitive K+ channels. Cardiovasc Drugs Ther 7(Suppl 3): 605‐610, 1993.
 175.Neuhaus J, Risau W, Wolburg H. Induction of blood‐brain barrier characteristics in bovine brain endothelial cells by rat astroglial cells in transfilter coculture. Ann N Y Acad Sci 633: 578‐580, 1991.
 176.Nielsen KC, Owman C. Adrenergic innervation of pial arteries related to the circle of Willis in the cat. Brain Res 6(2): 773‐776, 1967.
 177.Nielsen KC, Owman C, Sporrong B. Ultrastructure of the autonomic innervation apparatus in the main pial arteries of rats and cats. Brain Res 27(2): 25‐32, 1971.
 178.Nikolaeva S, Pradervand S, Centeno G, Zavadova V, Tokonami N, Maillard M, Bonny O, Firsov D. The circadian clock modulates renal sodium handling. J Am Soc Nephrol 23(2): 1019‐1026, 2012.
 179.Nithipatikom K, Grall AJ, Holmes BB, Harder DR, Falck JR, Campbell WB. Liquid chromatographic‐electrospray ionization‐mass spectrometric analysis of cytochrome P450 metabolites of arachidonic acid. Anal Biochem 298(2): 327‐336, 2001.
 180.Nizar K, Uhlirova H, Tian P, Saisan PA, Cheng Q, Reznichenko L, Weldy KL, Steed TC, Sridhar VB, MacDonald CL, Cui J, Gratiy SL, Sakadzic S, Boas DA, Beka TI, Einevoll GT, Chen J, Masliah E, Dale AM, Silva GA, Devor A. In vivo stimulus‐induced vasodilation occurs without IP3 receptor activation and may precede astrocytic calcium increase. J Neurosci 33(2): 8411‐8422, 2013.
 181.Nordlander MI. Inhibition of vascular myogenic tone and reactivity by calcium antagonists. J Hypertens Suppl 7(2): S141‐S145; discussion S146, 1989.
 182.Orrenius S, Gogvadze V, Zhivotovsky B. Mitochondrial oxidative stress: Implications for cell death. Annu Rev Pharmacol Toxicol 47: 143‐183, 2007.
 183.Pan ZG, Mao Y, Sun FY. VEGF enhances reconstruction of neurovascular units in the brain after injury. Sheng Li Xue Bao 69(2): 96‐108, 2017.
 184.Park WS, Han J, Earm YE. Physiological role of inward rectifier K(+) channels in vascular smooth muscle cells. Pflugers Arch 457(2): 137‐147, 2008.
 185.Paulson OB, Hasselbalch SG, Rostrup E, Knudsen GM, Pelligrino D. Cerebral blood flow response to functional activation. J Cereb Blood Flow Metab 30(2): 2‐14, 2010.
 186.Paulson OB, Waldemar G, Schmidt JF, Strandgaard S. Cerebral circulation under normal and pathologic conditions. Am J Cardiol 63(2): 2C‐5C, 1989.
 187.Pearce WJ. In cerebrovascular circadian rhythms, EETs keep the beat. Focus on “Rhythmic expression of cytochrome P450 epoxygenases CYP4x1 and CYP2c11 in the rat brain and vasculature.” Am J Physiol Cell Physiol 307(2): C986‐C988, 2014.
 188.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(2): H2029‐H2037, 2002.
 189.Phillis JW, Walter GA, O'Regan MH, Stair RE. Increases in cerebral cortical perfusate adenosine and inosine concentrations during hypoxia and ischemia. J Cereb Blood Flow Metab 7(2): 679‐686, 1987.
 190.Pombero A, Garcia‐Lopez R, Martinez S. Brain mesenchymal stem cells: physiology and pathological implications. Dev Growth Differ 58(2): 469‐480, 2016.
 191.Qu YY, Yuan MY, Liu Y, Xiao XJ, Zhu YL. The protective effect of epoxyeicosatrienoic acids on cerebral ischemia/reperfusion injury is associated with PI3K/Akt pathway and ATP‐sensitive potassium channels. Neurochem Res 40(2): 1‐14, 2015.
 192.Quayle JM, McCarron JG, Brayden JE, Nelson MT. Inward rectifier K+ currents in smooth muscle cells from rat resistance‐sized cerebral arteries. Am J Physiol 265(5 Pt 1): C1363‐C1370, 1993.
 193.Randriamboavonjy V, Busse R, Fleming I. 20‐HETE‐induced contraction of small coronary arteries depends on the activation of Rho‐kinase. Hypertension 41(3 Pt 2): 801‐806, 2003.
 194.Ribatti D, Nico B, Crivellato E. The role of pericytes in angiogenesis. Int J Dev Biol 55(2): 261‐268, 2011.
 195.Rosenblum WI. Neurogenic control of cerebral circulation. Stroke 2(2): 429‐439, 1971.
 196.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(2): 13463‐13474, 2015.
 197.Rosenstein JM, Mani N, Khaibullina A, Krum JM. Neurotrophic effects of vascular endothelial growth factor on organotypic cortical explants and primary cortical neurons. J Neurosci 23(2): 11036‐11044, 2003.
 198.Sandoo A, van Zanten JJ, Metsios GS, Carroll D, Kitas GD. The endothelium and its role in regulating vascular tone. Open Cardiovasc Med J 4: 302‐312, 2010.
 199.Sarkar P, Zaja I, Bienengraeber M, Rarick KR, Terashvili M, Canfield S, Falck JR, Harder DR. Epoxyeicosatrienoic acids pretreatment improves amyloid beta‐induced mitochondrial dysfunction in cultured rat hippocampal astrocytes. Am J Physiol Heart Circ Physiol 306(2): H475‐H484, 2014.
 200.Sato A, Sato Y, Uchida S. Activation of the intracerebral cholinergic nerve fibers originating in the basal forebrain increases regional cerebral blood flow in the rat's cortex and hippocampus. Neurosci Lett 361(1‐3): 90‐93, 2004.
 201.Schebesch KM, Herbst A, Bele S, Schodel P, Brawanski A, Stoerr EM, Lohmeier A, Kagerbauer SM, Martin J, Proescholdt M. Calcitonin‐gene related peptide and cerebral vasospasm. J Clin Neurosci 20(2): 584‐586, 2013.
 202.Schulz K, Sydekum E, Krueppel R, Engelbrecht CJ, Schlegel F, Schroter A, Rudin M, Helmchen F. Simultaneous BOLD fMRI and fiber‐optic calcium recording in rat neocortex. Nat Methods 9(2): 597‐602, 2012.
 203.Serres S, Raffard G, Franconi JM, Merle M. Close coupling between astrocytic and neuronal metabolisms to fulfill anaplerotic and energy needs in the rat brain. J Cereb Blood Flow Metab 28(2): 712‐724, 2008.
 204.Shaik JS, Ahmad M, Li W, Rose ME, Foley LM, Hitchens TK, Graham SH, Hwang SH, Hammock BD, Poloyac SM. Soluble epoxide hydrolase inhibitor trans‐4‐[4‐(3‐adamantan‐1‐yl‐ureido)‐cyclohexyloxy]‐benzoic acid is neuroprotective in rat model of ischemic stroke. Am J Physiol Heart Circ Physiol 305(2): H1605‐H1613, 2013.
 205.Shen SW, Duan CL, Chen XH, Wang YQ, Sun X, Zhang QW, Cui HR, Sun FY. Neurogenic effect of VEGF is related to increase of astrocytes transdifferentiation into new mature neurons in rat brains after stroke. Neuropharmacology 108: 451‐461, 2016.
 206.Shin HK, Hong KW. Importance of calcitonin gene‐related peptide, adenosine and reactive oxygen species in cerebral autoregulation under normal and diseased conditions. Clin Exp Pharmacol Physiol 31(1‐2): 1‐7, 2004.
 207.Shin HK, Shin YW, Hong KW. Role of adenosine A(2B) receptors in vasodilation of rat pial artery and cerebral blood flow autoregulation. Am J Physiol Heart Circ Physiol 278(2): H339‐H344, 2000.
 208.Siddiq I, Park E, Liu E, Spratt SK, Surosky R, Lee G, Ando D, Giedlin M, Hare GM, Fehlings MG, Baker AJ. Treatment of traumatic brain injury using zinc‐finger protein gene therapy targeting VEGF‐A. J Neurotrauma 29(2): 2647‐2659, 2012.
 209.Simard M, Arcuino G, Takano T, Liu QS, Nedergaard M. Signaling at the gliovascular interface. J Neurosci 23(2): 9254‐9262, 2003.
 210.Sloan HL, Austin VC, Blamire AM, Schnupp JW, Lowe AS, Allers KA, Matthews PM, Sibson NR. Regional differences in neurovascular coupling in rat brain as determined by fMRI and electrophysiology. Neuroimage 53(2): 399‐411, 2010.
 211.Spector AA. Arachidonic acid cytochrome P450 epoxygenase pathway. J Lipid Res 50(Suppl): S52‐S56, 2009.
 212.Stapor PC, Sweat RS, Dashti DC, Betancourt AM, Murfee WL. Pericyte dynamics during angiogenesis: New insights from new identities. J Vasc Res 51(2): 163‐174, 2014.
 213.Sukumaran SV, Singh TU, Parida S, Narasimha Reddy C, Thangamalai R, Kandasamy K, Singh V, Mishra SK. TRPV4 channel activation leads to endothelium‐dependent relaxation mediated by nitric oxide and endothelium‐derived hyperpolarizing factor in rat pulmonary artery. Pharmacol Res 78: 18‐27, 2013.
 214.Suzuki A, Stern SA, Bozdagi O, Huntley GW, Walker RH, Magistretti PJ, Alberini CM. Astrocyte‐neuron lactate transport is required for long‐term memory formation. Cell 144(2): 810‐823, 2011.
 215.Sweeney MD, Ayyadurai S, Zlokovic BV. Pericytes of the neurovascular unit: Key functions and signaling pathways. Nat Neurosci 19(2): 771‐783, 2016.
 216.Szade A, Grochot‐Przeczek A, Florczyk U, Jozkowicz A, Dulak J. Cellular and molecular mechanisms of inflammation‐induced angiogenesis. IUBMB Life 67(2): 145‐159, 2015.
 217.Terashvili M, Pratt PF, Gebremedhin D, Narayanan J, Harder DR. Reactive oxygen species cerebral autoregulation in health and disease. Pediatr Clin North Am 53(2): 1029‐37, xi, 2006.
 218.Theodosis DT, Poulain DA, Oliet SH. Activity‐dependent structural and functional plasticity of astrocyte‐neuron interactions. Physiol Rev 88(2): 983‐1008, 2008.
 219.Tian X, Brookes O, Battaglia G. Pericytes from mesenchymal stem cells as a model for the blood‐brain barrier. Sci Rep 7: 39676, 2017.
 220.Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh SE, Katz S, Las G, Alroy J, Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, Shirihai OS. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J 27(2): 433‐446, 2008.
 221.Uddman R, Edvinsson L. Neuropeptides in the cerebral circulation. Cerebrovasc Brain Metab Rev 1(2): 230‐252, 1989.
 222.Ukai H, Ueda HR. Systems biology of mammalian circadian clocks. Annu Rev Physiol 72: 579‐603, 2010.
 223.Vidal M, Maniglier M, Deboux C, Bachelin C, Zujovic V, Baron‐Van Evercooren A. Adult DRG stem/progenitor cells generate pericytes in the presence of central nervous system (CNS) developmental cues, and Schwann cells in response to CNS demyelination. Stem Cells 33(2): 2011‐2024, 2015.
 224.Wang L, Chen M, Yuan L, Xiang Y, Zheng R, Zhu S. 14,15‐EET promotes mitochondrial biogenesis and protects cortical neurons against oxygen/glucose deprivation‐induced apoptosis. Biochem Biophys Res Commun 450(2): 604‐609, 2014.
 225.Wang X, Lou N, Xu Q, Tian GF, Peng WG, Han X, Kang J, Takano T, Nedergaard M. Astrocytic Ca2+ signaling evoked by sensory stimulation in vivo. Nat Neurosci 9(2): 816‐823, 2006.
 226.Wang X, Su B, Lee HG, Li X, Perry G, Smith MA, Zhu X. Impaired balance of mitochondrial fission and fusion in Alzheimer's disease. J Neurosci 29(2): 9090‐9103, 2009.
 227.Wang YQ, Cui HR, Yang SZ, Sun HP, Qiu MH, Feng XY, Sun FY. VEGF enhance cortical newborn neurons and their neurite development in adult rat brain after cerebral ischemia. Neurochem Int 55(2): 629‐636, 2009.
 228.Ward MC, Cunningham AM. Developmental expression of vascular endothelial growth factor receptor 3 and vascular endothelial growth factor C in forebrain. Neuroscience 303: 544‐557, 2015.
 229.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(2): 851‐862, 2016.
 230.Willie CK, Tzeng YC, Fisher JA, Ainslie PN. Integrative regulation of human brain blood flow. J Physiol 592(2): 841‐859, 2014.
 231.Winn HR, Morii S, Berne RM. The role of adenosine in autoregulation of cerebral blood flow. Ann Biomed Eng 13(3‐4): 321‐328, 1985.
 232.Witthoft A, Filosa JA, Karniadakis GE. Potassium buffering in the neurovascular unit: Models and sensitivity analysis. Biophys J 105(2): 2046‐2054, 2013.
 233.Wolters FJ, Mattace‐Raso FU, Koudstaal PJ, Hofman A, Ikram MA, Heart Brain Connection Collaborative Research Group. Orthostatic Hypotension and the Long‐Term Risk of Dementia: A Population‐Based Study. PLoS Med 13(2): e1002143, 2016.
 234.Woo J, Han D, Park J, Kim SJ, Kim Y. In‐depth characterization of the secretome of mouse CNS cell lines by LC‐MS/MS without prefractionation. Proteomics 15(2): 3617‐3622, 2015.
 235.Yamamoto T, Nakahata Y, Soma H, Akashi M, Mamine T, Takumi T. Transcriptional oscillation of canonical clock genes in mouse peripheral tissues. BMC Mol Biol 5: 18, 2004.
 236.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(2): 703‐716, 2006.
 237.Zhang C, Harder DR. Cerebral capillary endothelial cell mitogenesis and morphogenesis induced by astrocytic epoxyeicosatrienoic acid. Stroke 33(2): 2957‐2964, 2002.
 238.Zhang K, Lu J, Mori T, Smith‐Powell L, Synold TW, Chen S, Wen W. Baicalin increases VEGF expression and angiogenesis by activating the ERR{alpha}/PGC‐1{alpha} pathway. Cardiovasc Res 89(2): 426‐435, 2011.
 239.Zhang N, Xing M, Wang Y, Tao H, Cheng Y. Repetitive transcranial magnetic stimulation enhances spatial learning and synaptic plasticity via the VEGF and BDNF‐NMDAR pathways in a rat model of vascular dementia. Neuroscience 311: 284‐291, 2015.
 240.Zhang P, Ma Y, Wang Y, Ma X, Huang Y, Li RA, Wan S, Yao X. Nitric oxide and protein kinase G act on TRPC1 to inhibit 11,12‐EET‐induced vascular relaxation. Cardiovasc Res 104(2): 138‐146, 2014.
 241.Zhang W, Otsuka T, Sugo N, Ardeshiri A, Alhadid YK, Iliff JJ, DeBarber AE, Koop DR, Alkayed NJ. Soluble epoxide hydrolase gene deletion is protective against experimental cerebral ischemia. Stroke 39(2): 2073‐2078, 2008.
 242.Zhang Y, Hong G, Lee KS, Hammock BD, Gebremedhin D, Harder DR, Koehler RC, Sapirstein A. Inhibition of soluble epoxide hydrolase augments astrocyte release of vascular endothelial growth factor and neuronal recovery after oxygen‐glucose deprivation. J Neurochem 140(2): 814‐825, 2017.
 243.Zheng W, Talley Watts L, Holstein DM, Wewer J, Lechleiter JD. P2Y1R‐initiated, IP3R‐dependent stimulation of astrocyte mitochondrial metabolism reduces and partially reverses ischemic neuronal damage in mouse. J Cereb Blood Flow Metab 33(2): 600‐611, 2013.
 244.Zheng X, Zinkevich NS, Gebremedhin D, Gauthier KM, Nishijima Y, Fang J, Wilcox DA, Campbell WB, Gutterman DD, Zhang DX. Arachidonic acid‐induced dilation in human coronary arterioles: Convergence of signaling mechanisms on endothelial TRPV4‐mediated Ca2+ entry. J Am Heart Assoc 2(2): e000080, 2013.
 245.Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann KA, Pozzan T, Carmignoto G. Neuron‐to‐astrocyte signaling is central to the dynamic control of brain microcirculation. Nat Neurosci 6(2): 43‐50, 2003.
 246.Zorzano A, Liesa M, Sebastian D, Segales J, Palacin M. Mitochondrial fusion proteins: Dual regulators of morphology and metabolism. Semin Cell Dev Biol 21(6): 566‐574, 2010.

Teaching Material

D. R. Harder, K. R. Rarick, D. Gebremedhin, S. S. Cohen. Regulation of Cerebral Blood Flow: Response to Cytochrome P450 Lipid Metabolites. Compr Physiol. 8: 2018, 801-821.

Didactic Synopsis

Major Teaching Points:

  1. The high level of neuronal activity requires tight regulation of the cerebral vasculature to provide constant blood flow to maintain adequate energy substrate and neuronal health.
  2. The myogenic autoregulation of cerebral blood flow is a critical homeostatic mechanism that protects against blood pressure fluctuations and maintains constant blood flow to supply oxygen and nutrients.
  3. The neurovascular unit comprising of astrocytes, neurons, and the cerebral arterioles functions to compensate neuronal metabolic demand by coupling neuronal activity to cerebral blood flow.
  4. Impaired autoregulation of cerebral blood flow results in passive reductions or increases in cerebral blood flow leading to ischemic vascular damage or perfusion injuries including edema and blood-brain barrier breakdown.
  5. Dysregulation of neurovascular coupling is known to occur in conditions such as stroke, diabetes, traumatic brain injury, and also in neurodegenerative disorders including dementia and Alzheimer's disease.

Didactic Legends

The figures—in a freely downloadable PowerPoint format—can be found on the Images tab along with the formal legends published in the article. The following legends to the same figures are written to be useful for teaching.

Figure 1. Teaching points: Glucose enters neuronal cells via specific transporters (GLUTs), where it is phosphorylated by hexokinase to produce glucose-6-phosphate which is then processed through an array of metabolic pathways; these pathways involve mitochondrial metabolism, the pentose phosphate pathway, and gluconeogenesis. Through these pathways glucose is nearly completely oxidized to carbon dioxide and water.

Figure 2. Teaching points: The dependence of brain activity on blood flow in response to cognitive tasks is depicted in these functional MRI signals in response to initiation of brain activity.

Figure 3. Teaching points: The functional cellular organization of the brain is such that neurons, through chemical signals to astrocytes, signal vascular smooth muscle to dilate, increasing blood flow, and, thereby, meeting metabolic demand. Such activity occurs in discrete cellular clustering of neurons, astrocytes, and microvessels forming the neurovascular unit (NVU).

Figure 4. Teaching points: Astrocytes are visualized by immunostaining with antiglial fibrillary acidic protein (red). These astrocytes are surrounded by neurons labeled using antineuron specific enolase (green). Neurons and astrocytes are surrounded by the cerebral microcirculation (anti-PECAM blue) completing the NVU. The importance of astrocytes to regulate the cerebral circulation and mediate neurovascular coupling is suggested by the astrocyte's central location within the NVU and the close interaction of foot processes impinging on the microcirculation.

Figure 5. Teaching points: Signaling molecules released by astrocytes which act to dilate cerebral arterioles will reduce vascular tone increasing blood flow in response to neuronal metabolic demand. The views depicted here come from the cortex; however, as astrocytes make up over 60% of the cell mass in the brain, such NVU-dependent regulation occurs throughout all brain regions.

Figure 6. Teaching points: This process can be observed in isolated cannulated, pressurized arteries exhibiting regenerative electrical spike activity, the frequency and rate of rise of which increases as transmural pressure increases. Not all investigators observe regenerative electrical activity, but do measure vascular muscle depolarization.

Figure 7. Teaching points: Arterial smooth muscle cells depolarize with an average slope of between 50 and 60 mV/decade changes in [K]o. A rise in [K]o from 4 to 12 mmol/L results in hyperpolarization in that the Na/K ATPase saturates at a [K]o of 12 mV exerting a hyperpolarizing influence of up to 10 mV; other reasons for the [K]o-mediated hyperpolarization are discussed in the text. This figure depicts a classic Em versus log [K]o curve as described in constant field theory.

Figure 8. Teaching points: These data confirm other's work and clearly demonstrate that metabolites of AA formed via CYP epoxygenase by astrocytes uncovers a novel mechanism in brain angiogenesis and support a key role of astrocytes in regulating blood flow to meet neuronal metabolic demand.

Figure 9. Teaching points: Conserved transcriptional elements in CYP gene promoter regions can underlie circadian changes in expression and activity of CYP enzymes controlling blood flow. These changes are in response to cellular clock genes which can be found in different cell types.

Figure 10. Teaching points: A hypothesis can be extended to formation of 20-HETE which is oxygen dependent (43). Pharmacological reduction in 20-HETE levels increase CBF, whereas, 20-HETE alone leads to reduced blood flow under hypoxic states.


Related Articles:

Impact of Metabolic Diseases on Cerebral Circulation: Structural and Functional Consequences
Cerebral Circulation

Contact Editor

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

* Required Field

How to Cite

David R. Harder, Kevin R. Rarick, Debebe Gebremedhin, Susan S. Cohen. Regulation of Cerebral Blood Flow: Response to Cytochrome P450 Lipid Metabolites. Compr Physiol 2018, 8: 801-821. doi: 10.1002/cphy.c170025