Comprehensive Physiology Wiley Online Library

Neurogenic Control of the Vascular System: Focus on Cerebral Circulation

Full Article on Wiley Online Library



Abstract

The sections in this article are:

1 Neurogenic Regulation of Vascular Homeostasis—Problem Analysis
2 Brain Stem Control of The Circulation
2.1 Nucleus of Solitary Tract
2.2 Fastigial Nucleus
2.3 Locus Coeruleus
3 Central Noradrenergic Nerves and Stroke
4 Vascular Neuroeffector System
4.1 Neuromuscular Relationship
4.2 Intraneuronal Distribution of the Transmitter
4.3 Release of Transmitter and Effector Activation
5 Neurohistological Methods for Studies of Vascular Innervation
5.1 Fluorescence Histochemistry
5.2 Immunohistochemistry
5.3 Cholinesterase Histochemistry
6 Autonomic Innervation of Blood Vessels
6.1 Adrenergic Nerves
6.2 Cholinergic Nerves
6.3 P‐Type Nerves: Nonadrenergic, Noncholinergic
7 Sympathetic Control of Cerebrovascular Bed
8 Cerebrovascular Sympathetic Nerves and Vasospasm After Subarachnoid Hemorrhage
9 Blood‐Brain Barrier
9.1 Morphological Barrier
9.2 Enzymatic Barrier
10 Experimental Opening of Blood‐Brain Barrier
10.1 Effects of Circulating Neurotransmitters
11 Vasodilatation Mediated by Autonomic Receptors in Vessel Wall
11.1 β‐Adrenoceptors
11.2 Dopamine Receptors
11.3 Serotonergic Mechanisms
11.4 Histamine Receptors
11.5 Cholinergic Mechanisms
12 Neurogenic Vasodilatation
12.1 Ganglionic Inhibition by Dopamine
12.2 Peripheral Inhibition of Adrenergic Neurotransmission
12.3 Neuropeptide‐Induced Vasodilatation
13 Summary
Figure 1. Figure 1.

Fluorescence photomicrograph of whole mount of rat mesentery showing perivascular sympathetic nerves demonstrated by Falck‐Hillarp formaldehyde technique. Very dense plexus of green‐fluorescent, noradrenergic fibers supplies a small artery, whereas corresponding vein has less well‐developed innervation. Nerve terminals are seen to accompany small vessels of arteriolar caliber. × 155.

From Falck 99
Figure 2. Figure 2.

Arrangement of vascular neuroeffector apparatus. Postganglionic autonomic nerves ramify into small bundles forming a primary plexus, which is located in loose adventitia. Bundles consist of axons with smooth appearance characteristic of preterminal portion of nerve fiber. Bundles give rise to beaded (varicosed) fibers forming terminal effector plexus, located on surface of media layer. A few terminals may sometimes extend, but only for a short distance, into outermost layer of the media. Thus only smooth muscle cells located in adventi‐tia‐media junction receive direct autonomic innervation.

From Verity 279
Figure 3. Figure 3.

Location of pressor (crosshatching) and depressor (horizontal lines) centers in brain stem of cat. A‐C: cross sections through medulla at levels indicated by dotted lines to D. D: semidiagrammatic projection of pressor and depressor regions onto dorsal surface of brain stem, viewed with cerebellar peduncles cut across and cerebellum removed. AT, auditory tubercle; BC, brachium conjunctivum; BP, brachium pontis; C1, 1st cervical nerve; CN, cuneate nucleus; FG, facial genu; GN, gracile n.; IC, inferior colliculus: IO, inferior olivary n.; LN, lateral reticular n.; RB, restiform body; SO, superior olivary n.; SPV, spinal trigeminal tract; TB, trapezoid body; TC, tuberculum cinereum; TS, tractus solitarius; V‐VII and X, cranial nerves; and I‐III, levels of transection.

From Alexander 5
Figure 4. Figure 4.

Representation of some cerebral circulatory studies in which either stimulation or lesions of brain stem have been examined. To allow comparison, studies have been transferred to midsagittal section of human brain. Sections I and II, positions of ascending noradrenergic system and ascending reticular activating system, respectively. Section III, filled areas show lesioned sites that abolish cerebral CO2 reactivity in dogs. Crosshatching in this and remaining sections show lesions where no effect was noted. Sections IV and V, brain stem transections and pontine cold lesions, respectively, that abolish CO2 reactivity in cats. Sections VI‐VIII, studies in rhesus monkeys and baboons that demonstrate brain stem sites where stimulation results in increased cerebral blood flow and metabolism.

From Edvinsson and MacKenzie 81
Figure 5. Figure 5.

Cardiovascular reflex arc involving nucleus of solitary tract (NST). 1: Afferent cardiovascular neuron, from periphery to NST (cell bodies in nodose ganglion). 2: Central cardiovascular neuron (cell bodies in NST); a: axons to efferent cardiovascular neurons; b: axons to medullary centers; and c: axons running back from medullary centers to efferent neurons. 3: Efferent cardiovascular neuron, running from medulla or spinal cord to periphery; a: preganglionic cells in dorsal vagal nucleus and intermediolateral nucleus; b: postganglionic cells in peripheral autonomic ganglia. Filled circles with open centers, reflex arc neurons; open circle with filled center, modulatory neuron; solid lines, central axons; dashed lines, peripheral nerve connections.

From Palkovits 227
Figure 6. Figure 6.

a: Electron micrograph of innervated capillary in cat anterior hypothalamus, lined by endothelium (E) with junctional complexes (J) at cellular margins. Endothelial nucleus is not in plane of section. Pericyte cell nucleus (Pn) appears at top left. Arrows, pericyte cytoplasmic processes. Axon terminal is in outlined area. Vertical bar, 1.0 μm. b: Outlined area in a at higher magnification, showing axon terminal (Ax) surrounded by basement membrane and containing synaptic vesicles. Major contact in this section is between terminal and pericyte, although a small portion of axon abuts directly on endothelium. Pp, pericyte process; A, astrocyte process. Vertical bar, 0.5 μm. c: Axon terminal in direct apposition to endothelium of a different capillary. Vertical bar, 0.5 μm. d: Portion of large capillary (∼9.3 μm luminal diam) of heart ventricle from mouse treated with 5‐hydroxydopamine. Adrenergic terminals contain small, dense‐core vesicles (arrows), 1 of which is closely apposed both to a pericyte (PP) and to endothelium. No Schwann cell covering is present. Small circular profile between fibroblast (F) and axon terminal is intervaricose axon. Vertical bar, 0.5 μm.

From Forbes et al. 103 and Rennels and Nelson 238
Figure 7. Figure 7.

Fluorescence photomicrograph showing myosin immunoreactivity in wall of rat brain capillaries. In separate tests the same structures were shown to contain immunoreactive actin. Strong myosin reaction is also seen in smooth muscle media layer of arterioles (A). × 250.

From Owman et al. 221
Figure 8. Figure 8.

A: regenerative sprouting of catecholaminergic nerve fibers 7 wk after damage of central neurons by electrolytic lesion in rat mesencephalon. Entire thickness of wall of cross‐sectioned small vessel close to the lesion at base of the brain is invaded by abnormally distributed, catecholaminergic fibers. B: small artery at base of the brain of normal rat showing characteristic distribution of sympathetic nerve fibers in adventitia, superimposed on smooth muscle media layer. Nonspecific autofluorescence of ruffled inner elastic membrane is clearly visible. × 180.

From Katzman et al. 153
Figure 9. Figure 9.

After release of norepinephrine (NE) from sympathetic nerve terminals in the adventitia‐media junction of vessel, receptors of the directly innervated smooth muscle cells in outermost layer of media are activated first. Deeper muscle cell layers are activated by diffusion of transmitter substance, which reaches receptors in decreasing concentrations, and by propagation between adjacent smooth muscle cells via low‐resistance connections. Part of released transmitter is removed from effector region through neuronal uptake and part of it is metabolized extraneurally.

From Ljung 184
Figure 10. Figure 10.

A: Falck‐Hillarp formaldehyde technique for fluorescence‐histochemical visualization of monoamine transmitters. Proposed reaction sequence between catecholamines and formaldehyde to form products visible in fluorescence microscope. Amines are condensed with formaldehyde to 6,7‐dihydroxy‐1,2,3,4‐tetrahydroisoquinoline (I) and 4,6,7‐trihydroxy‐1,2,3,4‐tetrahydroisoquinoline (IV). Formation of fluorescent products (fluorophores) can proceed in 2 ways: either through autoxidation to 6,7‐dihydroxy‐3,4‐dihydroisoquinolines (R‐H) or by a 2nd, acid‐catalyzed reaction with formaldehyde to yield 2‐methyl‐6,7‐dihydroxy‐3,4‐dihydroisoquinolinium compounds (R‐CH3). At neutral pH fluorophores are in their tautomeric quinoidal forms (III and VI); they are converted to their nonquinoidal forms (II and V) after brief treatment with HCl. On prolonged treatment with HCl, the fluorophores with a 4‐hydroxy group (V) are transformed into fully aromatic compounds (VII). B: histochemical visualization of acetylcholinesterase (AChE), which is primarily localized in cholinergic nerves. Because the substrate used, acetylthiocholine (AThCh+), is hydrolyzed both by AChE and the nonspecific enzyme butyrylcholinesterase (BuChE), BuChE must be efficiently inhibited in order to show cholinergic nerves selectively. Tissue sections are incubated with AThCh+ in a medium saturated with copper thiocholine in order to effect its precipitation as rapidly as that formed from the enzymatic liberation of thiocholine. After rinsing, sections are immersed in yellow ammonium sulfide solution to convert precipitated copper sulfide. Sites of enzymatic activity are then visualized as dark‐brown sulfide deposits.

A from Björklund et al. 38; B from Koelle 159
Figure 11. Figure 11.

Indirect immunofluorescence technique (top) of Coons et al. 57 and peroxidase‐antiperoxidase (PAP) technique (bottom) of Sternberger 263 are the most commonly used methods for immunohistochemistry. In indirect immunofluorescence technique, sections are exposed to specific peptide antiserum in appropriate dilution for various lengths of time (usually 3 h) at room temperature. After thorough rinsing in phosphate buffer, they are incubated in fluorescein‐labeled antirabbit (or anti‐guinea pig) immunoglobulin G (IgG) for 30 min at room temperature. Sections are mounted in phosphate‐buffered glycerin after another rinsing in phosphate buffer and are examined with fluorescence microscope equipped with filters selected to give peak excitation at 490 nm. In the PAP procedure, sections are exposed to peptide antiserum overnight at 4°C. After rinsing in phosphate buffer, sections are incubated in unlabeled goat (or swine or sheep) antirabbit or anti‐guinea pig IgG for 30 min at room temperature, followed by incubation for 30 min with PAP complex [i.e., peroxidase coupled to rabbit (or guinea pig) antibody raised against horseradish peroxidase]. This is followed by incubation with a solution of 0.05% diaminobenzidine hydrochloride and 0.01% hydrogen peroxide in Tris buffer (pH 7.6) for 1 h. Sections are then examined under a light microscope.

Figure 12. Figure 12.

Anterior cerebral arteries obtained from human fetuses at 21 wk of gestation. Vessels were cut open and mounted flat on microscope slides. A: fluorescence photomicrograph after using Falck‐Hillarp formaldehyde technique to demonstrate adrenergic nerves. Vessel is supplied by dense plexus of sympathetic fibers. × 100. B: AChE‐containing nerves visualized by incubation with AThCh+ after inhibition of pseudocholinesterase with Mipafox. Artery receives equally dense supply of cholinergic fibers. × 100.

From Edvinsson et al. 91
Figure 13. Figure 13.

Cat's posterior cerebral artery, cut open longitudinally and mounted flat on microscope slide. Vessel was first treated with formaldehyde gas under mild conditions for visualization of norepinephrine (a) and was subsequently incubated for staining of AChE (b). There is close correspondence between the adrenergic and cholinergic nerve plexuses. × 235. c: Electron micrograph of adrenergic nerve varicosity (A) containing electron‐dense synaptic vesicles, located in close apposition (membrane‐to‐membrane distance, 20 nm) to cholinergic varicosity (Ch). Both terminals are located at innervation distance (∼100 nm) from membrane of smooth muscle cell (M). × 73,500.

From Owman 214
Figure 14. Figure 14.

Interaction between adrenergic and cholinergic mechanisms in brain circulation. Cerebral blood flow (CBF) was measured in caudate n. (CN) by heat clearance, using a thermistor heated a few tenths of a degree above cerebral temperature (TEMP) and measured by a contralateral reference thermistor. Temperature difference is maintained at constant level despite blood flow changes, with heating current required varying in direct proportion to alterations in flow. Left part of figure shows effects on CN blood flow of postganglionic sympathetic nerve stimulation (St; 6 V, 2‐ms duration, and 20‐Hz freq) or infusion of 2.0 μg·kg−1·min−1 of norepinephrine [N‐Ep (NE)] before or during intracarotid carbachol (CARB) infusion (2.5 μg·kg−1·min−1). Administration of atropine (At; 0.5 mg/kg) inhibited CARB‐induced increase in CN blood flow but did not affect antagonistic action of CARB during sympathetic nerve stimulation. Also, NE response was unchanged. There was no significant alteration in any other parameters recorded, except for small rise in systemic blood pressure (BP) during NE infusion. Slight rebound in flow after cessation of sympathetic nerve stimulation was potentiated by CARB. Diagram to right summarizes measurements in CN of 13 unanesthetized rabbits. Open bars, mean increases (± SE) in CN blood flow during intracarotid infusion with 2.5 or 1.3 μg·kg−1·min−1 of CARB. Stippled bars, mean reduction (± SE) in CN blood flow during postganglionic cervical sympathetic nerve stimulation (CS), either alone or during intracarotid infusion of 2.5 μg·kg−1·min−1 of CARB. , partial pressure of CO2 in arterial blood; , partial pressure of O2 in arterial blood.

From Aubineau et al. 15
Figure 15. Figure 15.

Schematic drawing of ultrastructure of different types of autonomic nerve endings. Left: cholinergic; center: noradrenergic; right: “P type.” Cholinergic nerve profile contains vesicles, most of which are small and electron lucent, whereas vesicles of adrenergic nerve profile are predominantly small and electron dense. The P‐type nerve profile is characterized by relative abundance of fairly large granules of varying electron density; they are sometimes referred to as large opaque vesicles.

From Håkanson et al. 122
Figure 16. Figure 16.

Schematic representation of how synthesis, storage, release, and inactivation of ATP is assumed to take place in a purinergic autonomic nerve terminal.

From Burnstock 45
Figure 17. Figure 17.

Vasoactive intestinal polypeptide (VIP) immunoreactivity in anterior cerebral arteries of cat. A: picture is composed of 2 adjacent cross sections and shows many VIPergic nerves enclosing vessel. × 200. B: oblique section through vessel wall. Several delicate VIPergic nerve fibers with beaded appearance run in wall and also enclose adjacent small arteriole in bottom right corner. × 450. C: cross section of vessel, demonstrating that VIPergic nerves run on surface of smooth muscle layer in a characteristic manner. × 450. Internal elastic membrane is autofluorescent in all 3 pictures.

From Larsson et al. 168
Figure 18. Figure 18.

Sympathetic nerve fibers visualized with Falck‐Hillarp formaldehyde histofluorescence technique. A: section of cat's brain, showing intracerebral arteriole branching into brain parenchyma from pial arteries visible in top lefthand margin of picture. Resistance vessel is supplied with a well‐developed plexus of sympathetic nerve terminals. × 250. B: whole mount of cat pial vein, cut open and mounted flat on microscope slide. Capacitance vessel receives substantial number of sympathetic fibers forming a network in wall. × 100.

Figure 19. Figure 19.

Superior cervical ganglion of rat showing green‐fluorescent, noradrenergic nerve cell bodies as demonstrated by Falck‐Hillarp formaldehyde technique. Cell nucleus appears dark. Note that ganglion also contains nonadrenergic cells. × 400.

Figure 20. Figure 20.

Fluorometric determinations of NE content in rabbit pial vessels at various times after pre‐ or postganglionic sympathectomy. Values indistinguishable from 0 are reached within 2 days after ganglionectomy. Decentralization leads to slightly elevated tissue NE level, due to reduced activity in postganglionic neuron devoid of its preganglionic input. Values are means ± SE, n = 4 determinations (10 at time 0), with tissues from 3 animals being pooled for each determination. Student's t test: *, 0.01 < P < 0.05; ***, P < 0.001.

From Edvinsson et al. 89
Figure 21. Figure 21.

Fluorescence microscopic estimation of sympathetic innervation (formaldehyde reaction) of intracerebral resistance vessels in baboons. In each region, number of arterioles accompanied by sympathetic fibers was related to total number of arterioles in 6‐μm‐thick sections.

From Edvinsson and Owman 88
Figure 22. Figure 22.

Continuous measurement of blood flow in caudate nucleus (CN) and lateral geniculate body (LGB) in unanesthetized rabbits, with thermoclearance technique (cf. Fig. 14). After unilateral transection of sympathetic trunk below superior cervical ganglion, there was an immediate increase in flow, followed by prompt reduction during electrical stimulation (30 Hz) of distal nerve stump. In reference to flow seen during inhalation of 7% CO2, effect of sympathetic denervation or stimulation was always greater in CN, which is better innervated by sympathetic perivascular nerves, than in LGB (cf. Fig. 21). ECoG, electrocorticogram; BP, systemic blood pressure; and , arterial gas tensions, measured by mass spectrometry; and TEMP, contralateral brain temperature.

From Sercombe et al. 253
Figure 23. Figure 23.

Schematic representation of modulatory effects of cranial sympathetics on cerebral blood flow autoregulation. A, lower limit of autoregulation during hemorrhage, which is accompanied by marked sympathoadrenal activation. B, upper limit of autoregulation during hypertension. C, lower limit of autoregulation can be moved toward lower blood pressure levels, provided perivascular sympathetic innervation of brain vessels is inactivated. D, upper limit of autoregulation can be moved toward higher blood pressure levels during enhanced activation of cerebrovascular sympathetic nerves.

Figure 24. Figure 24.

Possible pathogenesis of stroke after sympathetic denervation in stroke‐prone, spontaneously hypertensive rats.

From Heistad et al. 135
Figure 25. Figure 25.

Effect of sympathomimetic and sympathetic stimulation on cerebral blood volume (CBV) in mice. A: tyramine (which acts by uptake into adrenergic nerves, followed by displacement of transmitter) reduced CBV in intact, but not to any significant degree in sympathectomized, animals. B: bilateral electrical stimulation of sympathetic trunk in the neck significantly lowered CBV in comparison with untreated or sham‐operated animals, with the effect being blocked by injection of the α‐receptor antagonist phenoxybenzamine (PBZ; 2 mg/kg, iv). Values are means ± SE; number of animals in parentheses.

From Edvinsson et al. 84
Figure 26. Figure 26.

Sympathetic denervation leads to a variety of changes in cerebral hemodynamics, as illustrated by measurements of CBV in mice with a non‐diffusible tracer ([131I]‐labeled serum albumin), depending on type of denervation and postoperative time lapse. After postganglionic denervation (bilateral excision of superior cervical ganglia), CBV decreases during transmitter leakage, then increases when the degenerating vasoconstrictor nerves lose their transmitter, and finally normalizes when denervation supersensitivity ensues. After preganglionic denervation (decentralization of superior cervical ganglia), CBV is enhanced because of loss of vascular tone (cf. Fig. 20).

From Edvinsson et al. 84
Figure 27. Figure 27.

Circadian rhythm of CBV in mice subjected to 12‐h light/12‐h dark periods. Values are means ± SE; number of animals in parentheses.

From Edvinsson et al. 82
Figure 28. Figure 28.

Both the choroid plexus, responsible for cerebrospinal fluid (CSF) production, and the cerebrovascular bed, where most of blood volume is held in capacitance vessels, are innervated by sympathetic nerves. CSF dynamics and cerebral blood volume (CBV) are major factors in regulation of intracranial pressure. Postganglionic sympathetic denervation (SyX) of rabbits leads to approximately equal elevations in CSF production and CBV compared with unoperated controls (C), and to a marked increase in intracranial pressure. Values are means ± SE; number of animals in parentheses; differences between mean values are indicated according to Student's t test.

From Owman and Lindvall 226
Figure 29. Figure 29.

Whole mount preparations of longitudinally opened rabbit anterior cerebral arteries, processed according to Falck‐Hillarp formaldehyde technique for demonstration of perivascular adrenergic nerves. A: vessel from untreated control animal showing dense sympathetic nerve plexus; B: artery from animal treated with subarachnoid blood injection 3 days before sacrifice. Both the number of nerve terminals and their fluorescence intensity are markedly reduced (cf. Fig. 30). × 300.

From Edvinsson et al. 71
Figure 30. Figure 30.

Experimental subarachnoid hemorrhage in rabbit, achieved by injection of 1–2 ml of autologous blood into chiasmatic and basal cisterns. Blood deposited in subarachnoid space results in reduced in vitro uptake of [3H]NE into perivascular adrenergic nerves, concomitant with reduction in number and intensity of nerves with formaldehyde‐induced fluorescence (cf. Fig. 29), reflecting decrease in concentration of NE in blood vessels. Those changes are followed by “vasospasm,” as measured on arteriograms in terms of basiliar artery caliber. All parameters show similar pattern of normalization in course of 3 wk after maximum changes.

Adapted from Edvinsson et al. 71
Figure 31. Figure 31.

Cisternal blood injections in rabbits, used in an attempt to mimic subarachnoid hemorrhage, within 3 days results in 5‐fold increase in in vitro reactivity of basilar artery to NE (○‐ ‐ ‐○) in comparison with vessels from control animals (•–•). Values are means ± SE; n = 10.

From Svendgaard et al. 267
Figure 32. Figure 32.

Schematic representation of brain capillary. Endothelial cells are connected by continuous belts of tight junctions at several levels of opposing membranes, thereby restricting intercellular diffusion. Together with paucity of transendothelial pinocytosis in brain vessels, morphological blood‐brain barrier efficiently impedes entry of, e.g., transmitter amines and peptides from circulation into brain.

From Rapoport 235
Figure 33. Figure 33.

Fluorescence‐microscopic demonstration of enzymatic blood‐brain barrier. Animal received L‐dihydroxyphenylalanine, which is taken up into capillary endothelial cells and pericytes by facilitated transport system for amino acids. Within cells, the amino acid is decarboxylated to dopamine, which is trapped locally because the degrading enzyme, monoamine oxidase (also present within endothelial cells and pericytes), has been inhibited. Highest fluorescence intensity is in nuclear region, where cell is most voluminous. × 225.

From Owman et al. 220
Figure 34. Figure 34.

Incidence and degree of blood‐brain barrier lesions, as evidenced by extravasation of Evans blue‐albumin (EBA) complex or [14C]inulin in right hemisphere of rat brain after hypertensive insult (rapid blood injection into right carotid artery), hypercapnia or papaverine, or combined treatment. Vasodilatation by hypercapnia or papaverine significantly enhanced leakage induced by hypertensive insult (Student's t test: P < 0.001). Values are means ± SE.

Figure 35. Figure 35.

Regional cerebral blood flow (rCBF), measured by [14C]ethanol method in untreated control rats (C), as well as after infusion of catecholamines [NE, epinephrine (E), and isoproterenol (Iso)], alone at rate of 1 mg·kg−1·min−1, or after pretreatment of animals with competitive antagonists [phentolamine (Phent), propranolol (Prop)]. Values are means ± SE; n = 10 controls, 4–5 treated. Examples from frontal cortex (marked reduction in flow after NE and E, no effect by Iso; effects blocked or slightly reversed by Phent) and thalamus (little or no reduction by NE and E, enhanced flow after α‐receptor blockade by Phent; increased flow by Iso, counteracted by Prop).

From Edvinsson et al. 79
Figure 36. Figure 36.

rCBF in rats, measured by [14C]ethanol method, exemplified by cortex (open bars) and caudate nucleus (stippled bars). Values are means ± SE; number of animals indicated. In comparison with untreated controls, hypercapnia induced expected flow increase, whereas NE (1 μg·kg−1·min−1, iv) reduced rCBF (cf. Fig. 35). When the same dose of NE was infused after opening of morphological blood‐brain barrier by hypertonic shock (2 M urea injected into internal carotid), flow was significantly elevated secondary to NE‐induced increase in brain metabolism. Comparison between mean values (Student's t test): *, 0.01 < P < 0.05; **, 0.001 < P < 0.01; ***, P < 0.001.

Adapted from Edvinsson et al. 79
Figure 37. Figure 37.

Parasympathomimetic effects on brain circulation (CBF) of rats studied with venous outflow method. A: with intact blood‐brain barrier, intracarotid infusion of carbacholine induced small and short‐lasting changes in flow via direct effects on cholinergic receptors of vessel walls. B: after hypertensive opening of blood‐brain barrier with intracarotid bolus of autologous blood, biphasic response was more pronounced and long lasting. Augmented effect was due to added parasympathomimetic stimulation of cerebral metabolic rate of O2, which was enhanced by ∼20% when carbacholine was allowed to enter brain. MABP, mean arterial blood pressure; ICP, intracranial pressure.

From Owman et al. 222
Figure 38. Figure 38.

Composite picture of representative curves comparing effects of histamine on feline intra‐ and extracranial arteries in vitro. Contractile responses were obtained with histamine H2 receptors blocked. Dilatation effects were recorded during blockade of H1 receptors at an active tonic contraction of 250 dyns by 3 × 10−6 M serotonin. Histamine contracted extracranial arteries more efficiently than intracranial arteries. Dilatory response was slightly more pronounced intracranially. Contractile effect on intracranial vessels was, in contrast to other responses, nonspecific in the sense that the specific histamine antagonist tested failed to produce a parallel shift of dose‐response curve toward higher agonist concentrations. ED50, effective dose, 50%; EAM, maximum response.

From Owman et al. 219
Figure 39. Figure 39.

Experiments on 3 different strips of canine saphenous vein showing that concentrations of 3 vasoactive amines, which do not cause changes in tension of unstimulated preparations or during contractions with exogenous NE, do cause pronounced relaxation during sympathetic nerve stimulation (top graphs). Relaxation is due to decrease in evoked release of adrenergic neurotransmitter (bottom graphs), measured in terms of tritium overflow after preincubation of vessel preparation in presence of [3H]NE. ES, electrical stimulation.

From McGrath and Vanhoutte 198
Figure 40. Figure 40.

Schematic illustration of events mediated by substance P (SP) occurring during peripheral activation of small‐diameter sensory nerves. Intradermal injection of SP, for example, releases histamine (H), which acts on sensory nerves to produce an itching sensation as well as release of SP from perivascular nerves containing it, resulting in antidromic vasodilatation. BV, blood vessel; M, mast cell; filled circles, mast cell granules; and P1 and P2, primary sensory neurons 1 and 2.

From Hägermark et al. 121
Figure 41. Figure 41.

Increase in caliber of cat's pial arteries (ranging in size from 40 to 180 μm) and veins, expressed as percent change in size from preinjection values, during administration of various concentrations of VIP into perivascular space. Values are means ± SE. Significant changes (Student's t test) in caliber in presence of VIP in comparison with injection of vehicle (mock CSF): *, 0.01 < P < 0.02; **, 0.001 < P < 0.01; and ***, P < 0.001.

From Edvinsson et al. 72
Figure 42. Figure 42.

Time course of changes in CBF and cerebral O2 consumption in baboons during intracarotid infusion of VIP (10−11 mol/min for 20 min). Open circles, with intact blood‐brain barrier, the 2 parameters are not significantly altered. Filled circles, significant and parallel increase in both flow and metabolism after opening of barrier by intracarotid bolus injection of hypertonic urea at time 0. Values are means ± SE and are expressed as percent changes from their respective levels prior to administration of peptide. Significant differences from base line values (Student's t test): *, 0.01 < P < 0.05.

From Edvinsson et al. 72
Figure 43. Figure 43.

Vasodilatory effects of morphine (A) and Leu‐enkephalin (B) on isolated feline middle cerebral artery. Vessel had been given tonic contraction with prostaglandin F (2.5 × 10−6 M) beforehand. Dose‐response curves are shifted in parallel to right, indicating competitive inhibition, in presence of 2 concentrations of naloxone.

From Hanko et al. 123


Figure 1.

Fluorescence photomicrograph of whole mount of rat mesentery showing perivascular sympathetic nerves demonstrated by Falck‐Hillarp formaldehyde technique. Very dense plexus of green‐fluorescent, noradrenergic fibers supplies a small artery, whereas corresponding vein has less well‐developed innervation. Nerve terminals are seen to accompany small vessels of arteriolar caliber. × 155.

From Falck 99


Figure 2.

Arrangement of vascular neuroeffector apparatus. Postganglionic autonomic nerves ramify into small bundles forming a primary plexus, which is located in loose adventitia. Bundles consist of axons with smooth appearance characteristic of preterminal portion of nerve fiber. Bundles give rise to beaded (varicosed) fibers forming terminal effector plexus, located on surface of media layer. A few terminals may sometimes extend, but only for a short distance, into outermost layer of the media. Thus only smooth muscle cells located in adventi‐tia‐media junction receive direct autonomic innervation.

From Verity 279


Figure 3.

Location of pressor (crosshatching) and depressor (horizontal lines) centers in brain stem of cat. A‐C: cross sections through medulla at levels indicated by dotted lines to D. D: semidiagrammatic projection of pressor and depressor regions onto dorsal surface of brain stem, viewed with cerebellar peduncles cut across and cerebellum removed. AT, auditory tubercle; BC, brachium conjunctivum; BP, brachium pontis; C1, 1st cervical nerve; CN, cuneate nucleus; FG, facial genu; GN, gracile n.; IC, inferior colliculus: IO, inferior olivary n.; LN, lateral reticular n.; RB, restiform body; SO, superior olivary n.; SPV, spinal trigeminal tract; TB, trapezoid body; TC, tuberculum cinereum; TS, tractus solitarius; V‐VII and X, cranial nerves; and I‐III, levels of transection.

From Alexander 5


Figure 4.

Representation of some cerebral circulatory studies in which either stimulation or lesions of brain stem have been examined. To allow comparison, studies have been transferred to midsagittal section of human brain. Sections I and II, positions of ascending noradrenergic system and ascending reticular activating system, respectively. Section III, filled areas show lesioned sites that abolish cerebral CO2 reactivity in dogs. Crosshatching in this and remaining sections show lesions where no effect was noted. Sections IV and V, brain stem transections and pontine cold lesions, respectively, that abolish CO2 reactivity in cats. Sections VI‐VIII, studies in rhesus monkeys and baboons that demonstrate brain stem sites where stimulation results in increased cerebral blood flow and metabolism.

From Edvinsson and MacKenzie 81


Figure 5.

Cardiovascular reflex arc involving nucleus of solitary tract (NST). 1: Afferent cardiovascular neuron, from periphery to NST (cell bodies in nodose ganglion). 2: Central cardiovascular neuron (cell bodies in NST); a: axons to efferent cardiovascular neurons; b: axons to medullary centers; and c: axons running back from medullary centers to efferent neurons. 3: Efferent cardiovascular neuron, running from medulla or spinal cord to periphery; a: preganglionic cells in dorsal vagal nucleus and intermediolateral nucleus; b: postganglionic cells in peripheral autonomic ganglia. Filled circles with open centers, reflex arc neurons; open circle with filled center, modulatory neuron; solid lines, central axons; dashed lines, peripheral nerve connections.

From Palkovits 227


Figure 6.

a: Electron micrograph of innervated capillary in cat anterior hypothalamus, lined by endothelium (E) with junctional complexes (J) at cellular margins. Endothelial nucleus is not in plane of section. Pericyte cell nucleus (Pn) appears at top left. Arrows, pericyte cytoplasmic processes. Axon terminal is in outlined area. Vertical bar, 1.0 μm. b: Outlined area in a at higher magnification, showing axon terminal (Ax) surrounded by basement membrane and containing synaptic vesicles. Major contact in this section is between terminal and pericyte, although a small portion of axon abuts directly on endothelium. Pp, pericyte process; A, astrocyte process. Vertical bar, 0.5 μm. c: Axon terminal in direct apposition to endothelium of a different capillary. Vertical bar, 0.5 μm. d: Portion of large capillary (∼9.3 μm luminal diam) of heart ventricle from mouse treated with 5‐hydroxydopamine. Adrenergic terminals contain small, dense‐core vesicles (arrows), 1 of which is closely apposed both to a pericyte (PP) and to endothelium. No Schwann cell covering is present. Small circular profile between fibroblast (F) and axon terminal is intervaricose axon. Vertical bar, 0.5 μm.

From Forbes et al. 103 and Rennels and Nelson 238


Figure 7.

Fluorescence photomicrograph showing myosin immunoreactivity in wall of rat brain capillaries. In separate tests the same structures were shown to contain immunoreactive actin. Strong myosin reaction is also seen in smooth muscle media layer of arterioles (A). × 250.

From Owman et al. 221


Figure 8.

A: regenerative sprouting of catecholaminergic nerve fibers 7 wk after damage of central neurons by electrolytic lesion in rat mesencephalon. Entire thickness of wall of cross‐sectioned small vessel close to the lesion at base of the brain is invaded by abnormally distributed, catecholaminergic fibers. B: small artery at base of the brain of normal rat showing characteristic distribution of sympathetic nerve fibers in adventitia, superimposed on smooth muscle media layer. Nonspecific autofluorescence of ruffled inner elastic membrane is clearly visible. × 180.

From Katzman et al. 153


Figure 9.

After release of norepinephrine (NE) from sympathetic nerve terminals in the adventitia‐media junction of vessel, receptors of the directly innervated smooth muscle cells in outermost layer of media are activated first. Deeper muscle cell layers are activated by diffusion of transmitter substance, which reaches receptors in decreasing concentrations, and by propagation between adjacent smooth muscle cells via low‐resistance connections. Part of released transmitter is removed from effector region through neuronal uptake and part of it is metabolized extraneurally.

From Ljung 184


Figure 10.

A: Falck‐Hillarp formaldehyde technique for fluorescence‐histochemical visualization of monoamine transmitters. Proposed reaction sequence between catecholamines and formaldehyde to form products visible in fluorescence microscope. Amines are condensed with formaldehyde to 6,7‐dihydroxy‐1,2,3,4‐tetrahydroisoquinoline (I) and 4,6,7‐trihydroxy‐1,2,3,4‐tetrahydroisoquinoline (IV). Formation of fluorescent products (fluorophores) can proceed in 2 ways: either through autoxidation to 6,7‐dihydroxy‐3,4‐dihydroisoquinolines (R‐H) or by a 2nd, acid‐catalyzed reaction with formaldehyde to yield 2‐methyl‐6,7‐dihydroxy‐3,4‐dihydroisoquinolinium compounds (R‐CH3). At neutral pH fluorophores are in their tautomeric quinoidal forms (III and VI); they are converted to their nonquinoidal forms (II and V) after brief treatment with HCl. On prolonged treatment with HCl, the fluorophores with a 4‐hydroxy group (V) are transformed into fully aromatic compounds (VII). B: histochemical visualization of acetylcholinesterase (AChE), which is primarily localized in cholinergic nerves. Because the substrate used, acetylthiocholine (AThCh+), is hydrolyzed both by AChE and the nonspecific enzyme butyrylcholinesterase (BuChE), BuChE must be efficiently inhibited in order to show cholinergic nerves selectively. Tissue sections are incubated with AThCh+ in a medium saturated with copper thiocholine in order to effect its precipitation as rapidly as that formed from the enzymatic liberation of thiocholine. After rinsing, sections are immersed in yellow ammonium sulfide solution to convert precipitated copper sulfide. Sites of enzymatic activity are then visualized as dark‐brown sulfide deposits.

A from Björklund et al. 38; B from Koelle 159


Figure 11.

Indirect immunofluorescence technique (top) of Coons et al. 57 and peroxidase‐antiperoxidase (PAP) technique (bottom) of Sternberger 263 are the most commonly used methods for immunohistochemistry. In indirect immunofluorescence technique, sections are exposed to specific peptide antiserum in appropriate dilution for various lengths of time (usually 3 h) at room temperature. After thorough rinsing in phosphate buffer, they are incubated in fluorescein‐labeled antirabbit (or anti‐guinea pig) immunoglobulin G (IgG) for 30 min at room temperature. Sections are mounted in phosphate‐buffered glycerin after another rinsing in phosphate buffer and are examined with fluorescence microscope equipped with filters selected to give peak excitation at 490 nm. In the PAP procedure, sections are exposed to peptide antiserum overnight at 4°C. After rinsing in phosphate buffer, sections are incubated in unlabeled goat (or swine or sheep) antirabbit or anti‐guinea pig IgG for 30 min at room temperature, followed by incubation for 30 min with PAP complex [i.e., peroxidase coupled to rabbit (or guinea pig) antibody raised against horseradish peroxidase]. This is followed by incubation with a solution of 0.05% diaminobenzidine hydrochloride and 0.01% hydrogen peroxide in Tris buffer (pH 7.6) for 1 h. Sections are then examined under a light microscope.



Figure 12.

Anterior cerebral arteries obtained from human fetuses at 21 wk of gestation. Vessels were cut open and mounted flat on microscope slides. A: fluorescence photomicrograph after using Falck‐Hillarp formaldehyde technique to demonstrate adrenergic nerves. Vessel is supplied by dense plexus of sympathetic fibers. × 100. B: AChE‐containing nerves visualized by incubation with AThCh+ after inhibition of pseudocholinesterase with Mipafox. Artery receives equally dense supply of cholinergic fibers. × 100.

From Edvinsson et al. 91


Figure 13.

Cat's posterior cerebral artery, cut open longitudinally and mounted flat on microscope slide. Vessel was first treated with formaldehyde gas under mild conditions for visualization of norepinephrine (a) and was subsequently incubated for staining of AChE (b). There is close correspondence between the adrenergic and cholinergic nerve plexuses. × 235. c: Electron micrograph of adrenergic nerve varicosity (A) containing electron‐dense synaptic vesicles, located in close apposition (membrane‐to‐membrane distance, 20 nm) to cholinergic varicosity (Ch). Both terminals are located at innervation distance (∼100 nm) from membrane of smooth muscle cell (M). × 73,500.

From Owman 214


Figure 14.

Interaction between adrenergic and cholinergic mechanisms in brain circulation. Cerebral blood flow (CBF) was measured in caudate n. (CN) by heat clearance, using a thermistor heated a few tenths of a degree above cerebral temperature (TEMP) and measured by a contralateral reference thermistor. Temperature difference is maintained at constant level despite blood flow changes, with heating current required varying in direct proportion to alterations in flow. Left part of figure shows effects on CN blood flow of postganglionic sympathetic nerve stimulation (St; 6 V, 2‐ms duration, and 20‐Hz freq) or infusion of 2.0 μg·kg−1·min−1 of norepinephrine [N‐Ep (NE)] before or during intracarotid carbachol (CARB) infusion (2.5 μg·kg−1·min−1). Administration of atropine (At; 0.5 mg/kg) inhibited CARB‐induced increase in CN blood flow but did not affect antagonistic action of CARB during sympathetic nerve stimulation. Also, NE response was unchanged. There was no significant alteration in any other parameters recorded, except for small rise in systemic blood pressure (BP) during NE infusion. Slight rebound in flow after cessation of sympathetic nerve stimulation was potentiated by CARB. Diagram to right summarizes measurements in CN of 13 unanesthetized rabbits. Open bars, mean increases (± SE) in CN blood flow during intracarotid infusion with 2.5 or 1.3 μg·kg−1·min−1 of CARB. Stippled bars, mean reduction (± SE) in CN blood flow during postganglionic cervical sympathetic nerve stimulation (CS), either alone or during intracarotid infusion of 2.5 μg·kg−1·min−1 of CARB. , partial pressure of CO2 in arterial blood; , partial pressure of O2 in arterial blood.

From Aubineau et al. 15


Figure 15.

Schematic drawing of ultrastructure of different types of autonomic nerve endings. Left: cholinergic; center: noradrenergic; right: “P type.” Cholinergic nerve profile contains vesicles, most of which are small and electron lucent, whereas vesicles of adrenergic nerve profile are predominantly small and electron dense. The P‐type nerve profile is characterized by relative abundance of fairly large granules of varying electron density; they are sometimes referred to as large opaque vesicles.

From Håkanson et al. 122


Figure 16.

Schematic representation of how synthesis, storage, release, and inactivation of ATP is assumed to take place in a purinergic autonomic nerve terminal.

From Burnstock 45


Figure 17.

Vasoactive intestinal polypeptide (VIP) immunoreactivity in anterior cerebral arteries of cat. A: picture is composed of 2 adjacent cross sections and shows many VIPergic nerves enclosing vessel. × 200. B: oblique section through vessel wall. Several delicate VIPergic nerve fibers with beaded appearance run in wall and also enclose adjacent small arteriole in bottom right corner. × 450. C: cross section of vessel, demonstrating that VIPergic nerves run on surface of smooth muscle layer in a characteristic manner. × 450. Internal elastic membrane is autofluorescent in all 3 pictures.

From Larsson et al. 168


Figure 18.

Sympathetic nerve fibers visualized with Falck‐Hillarp formaldehyde histofluorescence technique. A: section of cat's brain, showing intracerebral arteriole branching into brain parenchyma from pial arteries visible in top lefthand margin of picture. Resistance vessel is supplied with a well‐developed plexus of sympathetic nerve terminals. × 250. B: whole mount of cat pial vein, cut open and mounted flat on microscope slide. Capacitance vessel receives substantial number of sympathetic fibers forming a network in wall. × 100.



Figure 19.

Superior cervical ganglion of rat showing green‐fluorescent, noradrenergic nerve cell bodies as demonstrated by Falck‐Hillarp formaldehyde technique. Cell nucleus appears dark. Note that ganglion also contains nonadrenergic cells. × 400.



Figure 20.

Fluorometric determinations of NE content in rabbit pial vessels at various times after pre‐ or postganglionic sympathectomy. Values indistinguishable from 0 are reached within 2 days after ganglionectomy. Decentralization leads to slightly elevated tissue NE level, due to reduced activity in postganglionic neuron devoid of its preganglionic input. Values are means ± SE, n = 4 determinations (10 at time 0), with tissues from 3 animals being pooled for each determination. Student's t test: *, 0.01 < P < 0.05; ***, P < 0.001.

From Edvinsson et al. 89


Figure 21.

Fluorescence microscopic estimation of sympathetic innervation (formaldehyde reaction) of intracerebral resistance vessels in baboons. In each region, number of arterioles accompanied by sympathetic fibers was related to total number of arterioles in 6‐μm‐thick sections.

From Edvinsson and Owman 88


Figure 22.

Continuous measurement of blood flow in caudate nucleus (CN) and lateral geniculate body (LGB) in unanesthetized rabbits, with thermoclearance technique (cf. Fig. 14). After unilateral transection of sympathetic trunk below superior cervical ganglion, there was an immediate increase in flow, followed by prompt reduction during electrical stimulation (30 Hz) of distal nerve stump. In reference to flow seen during inhalation of 7% CO2, effect of sympathetic denervation or stimulation was always greater in CN, which is better innervated by sympathetic perivascular nerves, than in LGB (cf. Fig. 21). ECoG, electrocorticogram; BP, systemic blood pressure; and , arterial gas tensions, measured by mass spectrometry; and TEMP, contralateral brain temperature.

From Sercombe et al. 253


Figure 23.

Schematic representation of modulatory effects of cranial sympathetics on cerebral blood flow autoregulation. A, lower limit of autoregulation during hemorrhage, which is accompanied by marked sympathoadrenal activation. B, upper limit of autoregulation during hypertension. C, lower limit of autoregulation can be moved toward lower blood pressure levels, provided perivascular sympathetic innervation of brain vessels is inactivated. D, upper limit of autoregulation can be moved toward higher blood pressure levels during enhanced activation of cerebrovascular sympathetic nerves.



Figure 24.

Possible pathogenesis of stroke after sympathetic denervation in stroke‐prone, spontaneously hypertensive rats.

From Heistad et al. 135


Figure 25.

Effect of sympathomimetic and sympathetic stimulation on cerebral blood volume (CBV) in mice. A: tyramine (which acts by uptake into adrenergic nerves, followed by displacement of transmitter) reduced CBV in intact, but not to any significant degree in sympathectomized, animals. B: bilateral electrical stimulation of sympathetic trunk in the neck significantly lowered CBV in comparison with untreated or sham‐operated animals, with the effect being blocked by injection of the α‐receptor antagonist phenoxybenzamine (PBZ; 2 mg/kg, iv). Values are means ± SE; number of animals in parentheses.

From Edvinsson et al. 84


Figure 26.

Sympathetic denervation leads to a variety of changes in cerebral hemodynamics, as illustrated by measurements of CBV in mice with a non‐diffusible tracer ([131I]‐labeled serum albumin), depending on type of denervation and postoperative time lapse. After postganglionic denervation (bilateral excision of superior cervical ganglia), CBV decreases during transmitter leakage, then increases when the degenerating vasoconstrictor nerves lose their transmitter, and finally normalizes when denervation supersensitivity ensues. After preganglionic denervation (decentralization of superior cervical ganglia), CBV is enhanced because of loss of vascular tone (cf. Fig. 20).

From Edvinsson et al. 84


Figure 27.

Circadian rhythm of CBV in mice subjected to 12‐h light/12‐h dark periods. Values are means ± SE; number of animals in parentheses.

From Edvinsson et al. 82


Figure 28.

Both the choroid plexus, responsible for cerebrospinal fluid (CSF) production, and the cerebrovascular bed, where most of blood volume is held in capacitance vessels, are innervated by sympathetic nerves. CSF dynamics and cerebral blood volume (CBV) are major factors in regulation of intracranial pressure. Postganglionic sympathetic denervation (SyX) of rabbits leads to approximately equal elevations in CSF production and CBV compared with unoperated controls (C), and to a marked increase in intracranial pressure. Values are means ± SE; number of animals in parentheses; differences between mean values are indicated according to Student's t test.

From Owman and Lindvall 226


Figure 29.

Whole mount preparations of longitudinally opened rabbit anterior cerebral arteries, processed according to Falck‐Hillarp formaldehyde technique for demonstration of perivascular adrenergic nerves. A: vessel from untreated control animal showing dense sympathetic nerve plexus; B: artery from animal treated with subarachnoid blood injection 3 days before sacrifice. Both the number of nerve terminals and their fluorescence intensity are markedly reduced (cf. Fig. 30). × 300.

From Edvinsson et al. 71


Figure 30.

Experimental subarachnoid hemorrhage in rabbit, achieved by injection of 1–2 ml of autologous blood into chiasmatic and basal cisterns. Blood deposited in subarachnoid space results in reduced in vitro uptake of [3H]NE into perivascular adrenergic nerves, concomitant with reduction in number and intensity of nerves with formaldehyde‐induced fluorescence (cf. Fig. 29), reflecting decrease in concentration of NE in blood vessels. Those changes are followed by “vasospasm,” as measured on arteriograms in terms of basiliar artery caliber. All parameters show similar pattern of normalization in course of 3 wk after maximum changes.

Adapted from Edvinsson et al. 71


Figure 31.

Cisternal blood injections in rabbits, used in an attempt to mimic subarachnoid hemorrhage, within 3 days results in 5‐fold increase in in vitro reactivity of basilar artery to NE (○‐ ‐ ‐○) in comparison with vessels from control animals (•–•). Values are means ± SE; n = 10.

From Svendgaard et al. 267


Figure 32.

Schematic representation of brain capillary. Endothelial cells are connected by continuous belts of tight junctions at several levels of opposing membranes, thereby restricting intercellular diffusion. Together with paucity of transendothelial pinocytosis in brain vessels, morphological blood‐brain barrier efficiently impedes entry of, e.g., transmitter amines and peptides from circulation into brain.

From Rapoport 235


Figure 33.

Fluorescence‐microscopic demonstration of enzymatic blood‐brain barrier. Animal received L‐dihydroxyphenylalanine, which is taken up into capillary endothelial cells and pericytes by facilitated transport system for amino acids. Within cells, the amino acid is decarboxylated to dopamine, which is trapped locally because the degrading enzyme, monoamine oxidase (also present within endothelial cells and pericytes), has been inhibited. Highest fluorescence intensity is in nuclear region, where cell is most voluminous. × 225.

From Owman et al. 220


Figure 34.

Incidence and degree of blood‐brain barrier lesions, as evidenced by extravasation of Evans blue‐albumin (EBA) complex or [14C]inulin in right hemisphere of rat brain after hypertensive insult (rapid blood injection into right carotid artery), hypercapnia or papaverine, or combined treatment. Vasodilatation by hypercapnia or papaverine significantly enhanced leakage induced by hypertensive insult (Student's t test: P < 0.001). Values are means ± SE.



Figure 35.

Regional cerebral blood flow (rCBF), measured by [14C]ethanol method in untreated control rats (C), as well as after infusion of catecholamines [NE, epinephrine (E), and isoproterenol (Iso)], alone at rate of 1 mg·kg−1·min−1, or after pretreatment of animals with competitive antagonists [phentolamine (Phent), propranolol (Prop)]. Values are means ± SE; n = 10 controls, 4–5 treated. Examples from frontal cortex (marked reduction in flow after NE and E, no effect by Iso; effects blocked or slightly reversed by Phent) and thalamus (little or no reduction by NE and E, enhanced flow after α‐receptor blockade by Phent; increased flow by Iso, counteracted by Prop).

From Edvinsson et al. 79


Figure 36.

rCBF in rats, measured by [14C]ethanol method, exemplified by cortex (open bars) and caudate nucleus (stippled bars). Values are means ± SE; number of animals indicated. In comparison with untreated controls, hypercapnia induced expected flow increase, whereas NE (1 μg·kg−1·min−1, iv) reduced rCBF (cf. Fig. 35). When the same dose of NE was infused after opening of morphological blood‐brain barrier by hypertonic shock (2 M urea injected into internal carotid), flow was significantly elevated secondary to NE‐induced increase in brain metabolism. Comparison between mean values (Student's t test): *, 0.01 < P < 0.05; **, 0.001 < P < 0.01; ***, P < 0.001.

Adapted from Edvinsson et al. 79


Figure 37.

Parasympathomimetic effects on brain circulation (CBF) of rats studied with venous outflow method. A: with intact blood‐brain barrier, intracarotid infusion of carbacholine induced small and short‐lasting changes in flow via direct effects on cholinergic receptors of vessel walls. B: after hypertensive opening of blood‐brain barrier with intracarotid bolus of autologous blood, biphasic response was more pronounced and long lasting. Augmented effect was due to added parasympathomimetic stimulation of cerebral metabolic rate of O2, which was enhanced by ∼20% when carbacholine was allowed to enter brain. MABP, mean arterial blood pressure; ICP, intracranial pressure.

From Owman et al. 222


Figure 38.

Composite picture of representative curves comparing effects of histamine on feline intra‐ and extracranial arteries in vitro. Contractile responses were obtained with histamine H2 receptors blocked. Dilatation effects were recorded during blockade of H1 receptors at an active tonic contraction of 250 dyns by 3 × 10−6 M serotonin. Histamine contracted extracranial arteries more efficiently than intracranial arteries. Dilatory response was slightly more pronounced intracranially. Contractile effect on intracranial vessels was, in contrast to other responses, nonspecific in the sense that the specific histamine antagonist tested failed to produce a parallel shift of dose‐response curve toward higher agonist concentrations. ED50, effective dose, 50%; EAM, maximum response.

From Owman et al. 219


Figure 39.

Experiments on 3 different strips of canine saphenous vein showing that concentrations of 3 vasoactive amines, which do not cause changes in tension of unstimulated preparations or during contractions with exogenous NE, do cause pronounced relaxation during sympathetic nerve stimulation (top graphs). Relaxation is due to decrease in evoked release of adrenergic neurotransmitter (bottom graphs), measured in terms of tritium overflow after preincubation of vessel preparation in presence of [3H]NE. ES, electrical stimulation.

From McGrath and Vanhoutte 198


Figure 40.

Schematic illustration of events mediated by substance P (SP) occurring during peripheral activation of small‐diameter sensory nerves. Intradermal injection of SP, for example, releases histamine (H), which acts on sensory nerves to produce an itching sensation as well as release of SP from perivascular nerves containing it, resulting in antidromic vasodilatation. BV, blood vessel; M, mast cell; filled circles, mast cell granules; and P1 and P2, primary sensory neurons 1 and 2.

From Hägermark et al. 121


Figure 41.

Increase in caliber of cat's pial arteries (ranging in size from 40 to 180 μm) and veins, expressed as percent change in size from preinjection values, during administration of various concentrations of VIP into perivascular space. Values are means ± SE. Significant changes (Student's t test) in caliber in presence of VIP in comparison with injection of vehicle (mock CSF): *, 0.01 < P < 0.02; **, 0.001 < P < 0.01; and ***, P < 0.001.

From Edvinsson et al. 72


Figure 42.

Time course of changes in CBF and cerebral O2 consumption in baboons during intracarotid infusion of VIP (10−11 mol/min for 20 min). Open circles, with intact blood‐brain barrier, the 2 parameters are not significantly altered. Filled circles, significant and parallel increase in both flow and metabolism after opening of barrier by intracarotid bolus injection of hypertonic urea at time 0. Values are means ± SE and are expressed as percent changes from their respective levels prior to administration of peptide. Significant differences from base line values (Student's t test): *, 0.01 < P < 0.05.

From Edvinsson et al. 72


Figure 43.

Vasodilatory effects of morphine (A) and Leu‐enkephalin (B) on isolated feline middle cerebral artery. Vessel had been given tonic contraction with prostaglandin F (2.5 × 10−6 M) beforehand. Dose‐response curves are shifted in parallel to right, indicating competitive inhibition, in presence of 2 concentrations of naloxone.

From Hanko et al. 123
References
 1. Abrahams, V. C., and S. M. Hilton. Active muscle vasodilatation and its relation to the “flight and fight reactions” in the conscious animal (Abstract). J. Physiol. London 140: 16P–17P, 1958
 2. Abrahams, V. C., S. M. Hilton, and A. Zbrozyna. Active muscle vasodilatation elicited by mesencephalic stimulation. Its relation to the defense reaction (Abstract). J. Physiol. London 148: 32P–33P, 1959
 3. Achari, N. K., and C. B. Downman. Autonomic effector responses to stimulation of nucleus fastigius. J. Physiol. London 210: 637–650, 1970
 4. Ahlquist, R. P. A study of the adrenotropic receptors. Am. J. Physiol. 153: 586–600, 1948
 5. Alexander, R. S. Tonic and reflex functions of medullary sympathetic cardiovascular centers. J. Neurophysiol. 9: 205–217, 1946
 6. Allen, G. S., L. H. A. Gold, S. N. Chou, and L. A. French. Cerebral arterial spasm. 3. In vivo intracisternal production of spasm by serotonin and blood and its reversal by phenoxybenzamine. J. Neurosurg. 40: 451–458, 1974
 7. Allen, G. S., C. J. Gross, L. M. Henderson, and S. N. Chou. Cerebral arterial spasm. 4. In vitro effects of temperature, serotonin analogues, large nonphysiological concentrations of serotonin, and extracellular calcium and magnesium on serotonin‐induced contractions of the canine basilar artery. J. Neurosurg. 44: 585–593, 1976
 8. Allen, G. S., L. M. Henderson, S. N. Chou, and L. A. French. Cerebral arterial spasm. 1. In vitro contractile activity of vasoactive agents on canine basilar and middle cerebral arteries. J. Neurosurg. 40: 433–441, 1974
 9. Allen, G. S., L. M. Henderson, S. N. Chou, and L. A. French. Cerebral arterial spasm. 2. In vitro contractile activity of serotonin in human serum and CSF on the canine basilar artery, and its blockade by methylsergide and phenoxybenzamine. J. Neurosurg. 40: 442–450, 1974
 10. Andén, N. E., A. Rubenson, K. Fuxe, and T. Hökfelt. Evidence for dopamine receptor stimulation by apomorphine. J. Pharm. Pharmacol. 19: 627–629, 1967
 11. Anderson, W. D., and W. G. Kubicek. Effects of betahistine HCl, nicotinic acid, and histamine on basilar blood flow in anesthetized dogs. Stroke 2: 409–415, 1971
 12. Andersson, R. G. G., and S. R. M. Johansson. Beta‐adrenoceptors and the cyclic AMP system. Acta Pharmacol. Toxicol. 44, Suppl. 2: 21–27, 1979
 13. Ariëns, E. J., A. J. Beld, J. F. Rodrigues de Miranda, and A. M. Simonis. The pharmacon‐receptor‐effector concept. In: The Receptors. General Principles and Procedures, edited by R. D. O'Brien. New York: Plenum, 1979, vol. 1, p. 33–91.
 14. Armal, D. G., and H. M. Sinnamon. The locus coeruleus: neurobiology of a central noradrenergic nucleus. Prog. Neurobiol. 9: 147–196, 1977
 15. Aubineau, P., R. Sercombe, L. Edvinsson, and C. Owman. Cholinergic influence on the cerebrovascular bed mediated through actions on the vascular smooth musculature and on the perivascular sympathetic nerves. In: Neurogenic Control of the Brain Circulation, edited by C. Owman and L. Edvinsson. Oxford, UK: Pergamon, 1977, p. 331–342. (Wenner‐Gren Center Int. Symp. Ser., vol. 30.)
 16. Auer, L. M., W. Kuschinsky, B. B. Johansson, and L. Edvinsson. Sympatho‐adrenergic influence on pial veins and arteries in cat. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 291–300. (Dev. Neurosci. Ser., vol. 14.)
 17. Barja, F., and R. Mathison. Adrenergic and peptidergic (substance P and vasoactive intestinal polypeptide) innervation of the rat portal vein. Blood Vessels 19: 263–272, 1982
 18. Barja, F., R. Mathison, and H. Huggel. Substance P‐containing nerve fibres in large peripheral blood vessels of the rat. Cell Tissue Res. 229: 411–422, 1983
 19. Bates, D., R. M. Weinshilboum, R. J. Campbell, and T. Sundt, Jr. The effect of lesions in the locus coeruleus on the physiological responses of the cerebral blood vessels in cats. Brain Res. 136: 431–433, 1977
 20. Baumgarten, H. G., A.‐F. Holstein, and C. Owman. Auerbach's plexus of mammals and man: electron microscopic identification of three different types of neuronal processes in myenteric ganglia of the large intestine from rhesus monkeys, guinea‐pigs and man. Z. Zellforsch. Mikrosk. Anat. 106: 376–397, 1970
 21. Beaudet, A., and L. Descarries. The monoamine innervation of rat cerebral cortex: synaptic and nonsynaptic axon terminals. Neuroscience 3: 851–860, 1978
 22. Bell, C. Selective cholinergic denervation of the uterine artery in the guinea‐pig. Experientia 30: 257–258, 1974
 23. Bell, C., and G. Burnstock. Cholinergic vasomotor neuroeffector junctions. In: Physiology and Pharmacology of Vascular Neuroeffector Systems, edited by J. A. Bevan, R. F. Furchgott, R. A. Maxwell, and A. P. Somlyo. Basel: Karger, 1971, p. 37–46.
 24. Bennett, M. R. Autonomic Neuromuscular Transmission. Cambridge, UK: Cambridge Univ. Press, 1972 (Monogr. Physiol. Soc, vol. 30.)
 25. Bernard, M. C. Leçons sur la physiologie et la pathologie du système nerveux. Paris: Ballière, 1858, vol. 1, p. 376–396.
 26. Bertler, Å., B. Falck, C. Owman, and E. Rosengren. The localization of monoaminergic blood‐brain barrier mechanisms. Pharmacol. Rev. 18: 369–385, 1966
 27. Bevan, A. T., A. J. Honour, and F. H. Stott. Direct arterial pressure recording in unrestricted man. Clin. Sci. 36: 329–344, 1969
 28. Bevan, J. A., R. D. Bevan, and S. P. Duckles. Adrenergic regulation of vascular smooth muscle. Handbook of Physiology. The Cardiovascular System. Vascular Smooth Muscle, edited by D. F. Bohr, A. P. Somlyo, and H. V. Sparks, Jr. Bethesda, MD: Am. Physiol. Soc., 1980, sect. 2, vol. II, chapt. 18, p. 515–566.
 29. Bevan, J. A., G. M. Buga, A. Snowden, and S. I. Said. Is the neurovasodilator mechanism to cerebral and extracerebral arteries the same? In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 421–430. (Dev. Neurosci. Ser., vol. 14.)
 30. Bevan, J. A., D. W. Hosmer, B. Ljung, B. L. Pegram, and C. Su. Innervation patterns and neurogenic response of rabbit veins. Blood Vessels 11: 172–182, 1974
 31. Bevan, J. A., and R. E. Purdy. Variations in adrenergic innervation and contractile responses of the rabbit saphenous artery. Circ. Res. 32: 746–751, 1973
 32. Bevan, J. A., and C. Su. Sympathetic mechanisms in blood vessels: nerve and muscle relationships. Annu. Rev. Pharmacol. 13: 269–285, 1973
 33. Bevan, R. D. Effect of sympathetic denervation on smooth muscle cell proliferation in the growing rabbit ear artery. Circ. Res. 37: 15–19, 1975
 34. Bevan, R. D., and H. Tsuru. Functional and structural changes in the rabbit ear artery after sympathetic denervation. Circ. Res. 49: 478–485, 1981
 35. Biber, B., J. Fara, and O. Lundgren. Intestinal vasodilatation in response to transmural electrical field stimulation. Acta Physiol. Scand. 87: 277–282, 1973
 36. Bill, A., and J. Linder. Sympathetic control of cerebral blood flow in acute arterial hypertension. Acta Physiol. Scand. 96: 114–121, 1976
 37. Björklund, A., B. Falck, and O. Lindvall. Microspectrofluorometric analysis of cellular monoamines after formaldehyde or glyoxylic acid condensation. In: Methods in Brain Research, edited by P. B. Bradley. London: Wiley, 1975, p. 249–294.
 38. Björklund, A., B. Falck, and C. Owman. Fluorescence microscopic and microspectrofluorometric techniques for the cellular localization and characterization of biogenic amines. In: Methods in Investigative and Diagnostic Endocrinology. The Thyroid and Biogenic Amines, edited by J. E. Rall and I. J. Kopin. Amsterdam: North‐Holland, 1972, vol. 1, p. 318–368.
 39. Black, J. W., W. A. M. Duncan, C. J. Durant, C. R. Ganellin, and E. M. Parsons. Definition and antagonism of histamine H2‐receptors. Nature London 236: 385–390, 1972
 40. Bloom, S. R., and J. M. Polak. Peptidergic versus purinergic (Letter to the editor). Lancet 1: 93, 1978
 41. Brody, M. J. Histaminergic and cholinergic vasodilator systems. In: Mechanism of Vasodilatation, edited by P. M. Vanhoutte and I. Leusen. Basel: Karger, 1978, p. 266–277.
 42. Brody, M. J., and R. A. Shaffer. Distribution of vasodilator nerves in the canine hindlimb. Am. J. Physiol. 218: 470–474, 1970
 43. Buckner, C. K., and P. N. Patil. Steric aspects of adrenergic drugs. XVI. Beta adrenergic receptors of guinea‐pig atria and trachea. J. Pharmacol. Exp. Ther. 176: 634–649, 1971
 44. Bülbring, E., and H. H. Burn. The sympathetic dilator fibres in the muscles of cat and dog. J. Physiol. London 83: 483–501, 1935
 45. Burnstock, G. Purinergic nerves. Pharmacol. Rev. 24: 509–580, 1972
 46. Burnstock, G. Purinergic transmission. In: Handbook of Psychopharmacology. Synaptic Modulators, edited by L. L. Iversen, S. D. Iversen, and S. H. Snyder. New York: Plenum, 1975, vol. 5, p. 131–194.
 47. Burnstock, G. Putative neurotransmitters: adenosine triphosphate. Neurosci. Res. Program Bull. 17: 406–414, 1979
 48. Burnstock, G., and C. Bell. Peripheral autonomic transmission. In: The Peripheral Nervous System, edited by J. I. Hubbard. New York: Plenum, 1974, p. 277–327.
 49. Burnstock, G., and M. Costa. Adrenergic Neurons. Their Organization, Function and Development in the Peripheral Nervous System. London: Chapman & Hall, 1975
 50. Burnstock, G., T. Hökfelt, M. D. Gershon, L. I. Iversen, H. W. Kosterlitz, and J. H. Szurszewski. Non‐adrenergic, non‐cholinergic autonomic neurotransmission mechanisms. Neurosci. Res. Program Bull. 17: 377–519, 1979
 51. Burnstock, G., and J. H. Szurszewski. Non‐adrenergic, non‐cholinergic autonomic nerves: physiological roles. Neurosci. Res. Program Bull. 17: 396–405, 1979
 52. Busija, D. W., D. D. Heistad, and M. L. Marcus. Effects of sympathetic nerves on cerebral vessels during acute, moderate increases in arterial pressure in dogs and cats. Circ. Res. 46: 696–702, 1980
 53. Carlsson, C., M. Hägerdal, A. E. Kaasik, and B. K. Siesjö. A catecholamine‐mediated increase in the cerebral oxygen uptake during immobilisation stress in rats. Brain Res. 119: 227–231, 1977
 54. Carraway, R., and S. E. Leeman. The isolation of a new hypotensive peptide, neurotensin, from bovine hypothalami. J. Biol. Chem. 248: 6854–6861, 1973
 55. Chang, M. M., S. E. Leeman, and H. H. Niall. Amino‐acid sequence of substance P. Nature London New Biol. 232: 86–87, 1971
 56. Cohen, M. L., and B. A. Berkowitz. Differences between the effects of dopamine and apomorphine on rat aortic strips. Eur. J. Pharmacol. 34: 49–58, 1975
 57. Coons, A. H., H. J. Creech, R. N. Jones, and E. Berliner. The demonstration of pneumococcal antigen in tissues by the use of fluorescent antibody. J. Immunol. 45: 159–170, 1942
 58. Dahlgren, N., O. Lindvall, T. Sakabe, U. Stenevi, and B. K. Siesjö. Cerebral blood flow and oxygen consumption in the rat brain after lesions of the noradrenergic locus coeruleus system. Brain Res. 209: 11–23, 1981
 59. Dahlström, A. The Intraneuronal Distribution of Noradrenaline and the Transport and Life‐Span of Amine Storage Granules in the Sympathetic Adrenergic Neuron. A Histochemical and Biochemical Study. Stockholm: Haeggström, 1966
 60. Dahlström, A., and K. Fuxe. The adrenergic innervation of the nasal mucosa of certain mammals. Acta Oto‐Laryngol. 59: 65–72, 1965
 61. Daly, I. de B., and C. Hebb. Pulmonary and Bronchial Vascular Systems. London: Arnold, 1966
 62. Davis, J. N., and A. Carlsson. The effect of hypoxia on monoamine synthesis, levels and metabolism in rat brain. J. Neurochem. 21: 783–790, 1973
 63. Devine, C. E., and F. O. Simpson. The fine structure of vascular sympathetic neuromuscular contacts in the rat. Am. J. Anat. 121: 153–173, 1967
 64. Doba, N., and D. J. Reis. Changes in regional blood flow and cardiodynamics evoked by electrical stimulation of the fastigial nucleus in the cat and their similarity to orthostatic reflexes. J. Physiol. London 227: 729–747, 1972
 65. Doba, N., and D. J. Reis. Localization within the lower brainstem of a receptive area mediating the pressor response to increased intracranial pressure (the Cushing response). Brain Res. 47: 487–491, 1972
 66. Duckles. S. P. Acetylcholine and vasoactive intestinal polypeptide: cerebrovascular neurotransmitters. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 441–446. (Dev. Neurosci. Ser., vol. 14.)
 67. Duckles, S. P., and J. A. Bevan. Pharmacological characterization of adrenergic receptors of a rabbit cerebral artery. J. Pharmacol. Exp. Ther. 197: 371–378, 1976
 68. Eccles, R. M., and B. Libet. Origin and blockade of the synaptic responses of curarised sympathetic ganglia. J. Physiol. London 157: 484–503, 1961
 69. Echlin, F. A. Current concepts in the etiology and treatment of vasospasm. Clin. Neurosurg. 15: 133–160, 1968
 70. Edvinsson, L., P. Aubineau, C. Owman, R. Sercombe, and J. Seylaz. Sympathetic innervation of cerebral arteries: prejunctional supersensitivity to norepinephrine after sympathectomy or cocaine treatment. Stroke 6: 525–530, 1975
 71. Edvinsson, L., N. Egund, C. Owman, C. Sahlin, and N.A. Svendgaard. Reduced noradrenaline uptake and retention in cerebrovascular nerves associated with angiographically visible vasoconstriction following experimental subarachnoid hemorrhage in rabbits. Brain Res. Bull. 9: 799–805, 1982
 72. Edvinsson, L., J. Fahrenkrug, J. Hanko, J. McCulloch, C. Owman, and R. Uddman. Vasoactive intestinal polypeptide: distribution and effects on cerebral blood flow and metabolism. In: Cerebral Microcirculation and Metabolism, edited by J. Cervós‐Navarro and E. Fritschka. New York: Raven, 1981, p. 147–155.
 73. Edvinsson, L., J. Fahrenkrug, J. Hanko, C. Owman, F. Sundler, and R. Uddman. VIP (vasoactive intestinal polypeptide)‐containing nerves of intracranial arteries in mammals. Cell Tissue Res. 208: 135–142, 1980
 74. Edvinsson, L., B. Falck, and C. Owman. Possibilities for a cholinergic action on smooth musculature and sympathetic axons in brain vessels mediated by muscarinic and nicotinic receptors. J. Pharmacol Exp. Ther. 200: 117–126, 1977
 75. Edvinsson, L., J. E. Hardebo, E. T. MacKenzie, and C. Owman. Effect of exogenous noradrenaline on local cerebral blood flow after osmotic opening of the blood‐brain barrier in the rat. J. Physiol. London 274: 149–156, 1978
 76. Edvinsson, L., J. E. Hardebo, J. McCulloch, and C. Owman. Effects of dopaminergic agonists and antagonists on isolated cerebral blood vessels. Acta Physiol. Scand. 104: 349–359, 1978
 77. Edvinsson, L., J. E. Hardebo, and C. Owman. Pharmacological analysis of 5‐hydroxytryptamine receptors in isolated intracranial and extracranial vessels of cat and man. Circ. Res. 42: 143–151, 1978
 78. Edvinsson, L., H. J. Huggel, K. C. Nielsen, C. Owman, and J. G. Peristiany. Histochemistry of autonomic nerves in wing vessels of Pteropus giganteus (flying‐fox), and the effects of denervation. Cell Tissue Res. 152: 1–11, 1974
 79. Edvinsson, L., P. Lacombe, C. Owman, A.‐M. Reynier‐Rebuffel, and J. Seylaz. Quantitative changes in regional cerebral blood flow of rats induced by alpha‐ or beta‐adrenergic stimulants. Acta Physiol. Scand. 107: 289–296, 1979
 80. Edvinsson, L., M. Lindvall, K. C. Nielsen, and C. Owman. Are brain vessels innervated also by central (non‐sympathetic) adrenergic neurones? Brain Res. 63: 496–499, 1973
 81. Edvinsson, L., and E. T. MacKenzie. Amine mechanisms in the cerebral circulation. Pharmacol. Rev. 28: 275–348, 1977
 82. Edvinsson, L., K. C. Nielsen, and C. Owman. Circadian rhythm in cerebral blood volume of mouse. Experientia 29: 432–433, 1974
 83. Edvinsson, L., K. C. Nielsen, C. Owman, and B. Sporrong. Cholinergic mechanisms in pial vessels. Histochemistry, electron microscopy and pharmacology. Z. Zellforsch. Mikrosk. Anat. 134: 311–325, 1972
 84. Edvinsson, L., K. C. Nielsen, C. Owman, and K. A. West. Sympathetic adrenergic influence on brain vessels as studied by changes in cerebral blood volume of mice. Eur. Neurol. 6: 193–202, 1971/72
 85. Edvinsson, L., and C. Owman. Pharmacological characterization of adrenergic alpha and beta receptors mediating the vasomotor responses of cerebral arteries in vitro. Circ. Res. 35: 835–849, 1974
 86. Edvinsson, L., and C. Owman. A pharmacological comparison of histamine receptors in isolated extracranial and intracranial arteries in vitro. Neurology 25: 271–276, 1975
 87. Edvinsson, L., and C. Owman. Characterization of the uptake of 3H‐noradrenaline into rabbit brain vessels and its relation to the degree of sympathetic nerve supply. In: Neurogenic Control of the Brain Circulation, edited by C. Owman and L. Edvinsson. Oxford, UK: Pergamon, 1977, p. 121–132. (Wenner‐Gren Cent. Int. Symp. Ser., vol. 30.)
 88. Edvinsson, L., and C. Owman. Sympathetic innervation and adrenergic receptors in intraparenchymal cerebral arterioles of baboon. Acta Physiol. Scand. Suppl. 452: 57–59, 1977
 89. Edvinsson, L., C. Owman, E. Rosengren, and K. A. West. Concentration of noradrenaline in pial vessels, choroid plexus, and iris during two weeks after sympathetic ganglionectomy or decentralisation. Acta Physiol. Scand. 85: 201–206, 1972
 90. Edvinsson, L., C. Owman, and B. K. Siesjö. Physiological role of cerebrovascular sympathetic nerves in the autoregulation of cerebral blood flow. Brain Res. 117: 519–523, 1976
 91. Edvinsson, L., C. Owman, and N.‐O. Sjöberg. Autonomic nerves, mast cells, and amine receptors in human brain vessels. A histochemical and pharmacological study. Brain Res. 115: 377–393, 1976
 92. Edvinsson, L., C. Owman, and K. A. West. Influence of sympathetic denervation on intracranial pressure. In: Intracranial Pressure II, edited by N. Lundberg, U. Pontén, and M. Brock. Berlin: Springer‐Verlag, 1975, p. 455–459.
 93. Ehinger, B. Cholinesterases in ocular and orbital tissues of some mammals. Acta Univ. Lund Sect. 2 2: 1–16, 1966
 94. Ehinger, B., B. Falck, and B. Sporrong. Adrenergic fibres to the heart and to peripheral vessels. Bibl. Anat. 8: 35–45, 1967
 95. Elfvin, L.‐G. The ultrastructure of the superior cervical sympathetic ganglion of the cat. II. The structure of the preganglionic end fibers and the synapses as studied by serial sections. J. Ultrastruct. Res. 8: 441–476, 1963
 96. Erspamer, V. Recent research in the field of 5‐hydroxytryptamine and related indolealkylamines. Prog. Drug Res. 3: 151–367, 1961
 97. Essen, C. von. Effects of dopamine on the cerebral blood flow in the dog. Acta Neurol. Scand. 50: 39–52, 1974
 98. Euler, U. S. von, and J. H. Gaddum. An unidentified depressor substance in certain tissue extracts. J. Physiol. London 72: 74–87, 1931
 99. Falck, B. Observations on the possibilities of the cellular localization of monoamines by a fluorescence method. Acta Physiol. Scand. Suppl. 197: 1–25, 1962
 100. Fisher, G. H., K. Folkers, B. Pernow, and C. Y. Bowers. Synthesis and some biological activities of the tyrosine‐8 analog of substance P. J. Med. Chem. 19: 325–328, 1976
 101. Fitch, W., E. T. MacKenzie, and A. M. Harper. Effects of decreasing arterial pressure on cerebral blood flow in the baboon: influence of the sympathetic nervous system. Circ. Res. 37: 550–557, 1975
 102. Folkow, B., and E. Neil. Circulation. London: Oxford Univ. Press, 1971
 103. Forbes, M. S., M. L. Rennels, and E. Nelson. Innervation of myocardial microcirculation: terminal autonomic axons associated with capillaries and postcapillary venules in mouse heart. Am. J. Anat. 149: 71–92, 1977
 104. Fraser, R. A. R., B. M. Stein, R. E. Barrett, and J. L. Pool. Noradrenergic mediation of experimental cerebrovascular spasm. Stroke 1: 356–362, 1970
 105. Furchgott, R. F. The classification of adrenoceptors (adrenergic receptors). An evaluation from the standpoint of receptor theory. In: Handbook of Experimental Pharmacology. Catecholamines, edited by H. Blaschko and E. Muscholl. Berlin: Springer‐Verlag, 1972, vol. 33, p. 283–335.
 106. Furchgott, R. F., J. V. Zawadzki, and P. D. Cherry. Role of endothelium in the vasodilator response to acetylcholine. In: Vasodilatation, edited by P. M. Vanhoutte and I. Leusen. New York: Raven, 1981, p. 49–66.
 107. Furness, J. B., and M. Costa. The nervous release and the action of substances which affect intestinal muscle through neither adrenoceptors nor cholinoceptors. Philos. Trans. R. Soc. London Ser. B 265: 123–133, 1973
 108. Fuxe, K., and G. Sedvall. The distribution of adrenergic nerve fibres to the blood vessels of skeletal muscle. Acta Physiol. Scand. 64: 75–86, 1965
 109. Garattini, S., and L. Valzelli. Serotonin. New York: Elsevier, 1965
 110. Gazelius, B., E. Brodin, L. Olgart, and P. Panopolous. Evidence that substance P is a mediator of antidromic vasodilatation using somatostatin as a release inhibitor. Acta Physiol. Scand. 113: 155–159, 1981
 111. Gillespie, J. S., and S. M. Kirpekar. The localisation of endogenous and infused noradrenaline in the spleen (Abstract). J. Physiol. London 179: 46P–47P, 1965
 112. Glover, W. E., P. R. Carroll, and N. Latt. Histamine receptors in human temporal and rabbit ear arteries. In: International Symposium on Histamine H2‐Receptor Antagonists, edited by C. J. Wood and M. A. Simkins. London: Dasprint, 1973, p. 169–174.
 113. Goldberg, L. I. Cardiovascular and renal actions of dopamine: potential clinical applications. Pharmacol. Rev. 24: 1–29, 1972
 114. Goldberg, L. I. The dopamine vascular receptor. Biochem. Pharmacol. 24: 651–653, 1975
 115. Goldberg, L. I., P. F. Sonneville, and J. L. McNay. An investigation of the structural requirements for dopamine‐like renal vasodilation: phenylethylamines and apomorphine. J. Pharmacol. Exp. Ther. 163: 188–197, 1968
 116. Greenway, C. V., and A. E. Lawson. The effects of adrenaline and noradrenaline on venous return and regional flows in the anaesthetised cat with special reference to intestinal blood flow. J. Physiol. London 186: 579–595, 1966
 117. Greenway, C. V., and A. E. Lawson. Beta‐adrenergic receptors in the hepatic arterial bed of the anesthetized cat. Can. J. Physiol. Pharmacol. 47: 415–419, 1969
 118. Gross, P. M. Histamine H1‐ and H2‐receptors are differentially and spatially distributed in cerebral vessels. J. Cereb. Blood Flow Metab. 1: 441–446, 1981
 119. Gross, P. M., D. D. Heistad, M. R. Strait, M. L. Marcus, and M. J. Brody. Cerebral vascular responses to physiological stimulation of sympathetic pathways in cats. Circ. Res. 44: 288–294, 1979
 120. Haddy, F. J. Serotonin and the vascular system. Angiology 11: 21–24, 1960
 121. Hägermark, O., T. Hökfelt, and B. Pernow. Flare and itch induced by substance P in human skin. J. Invest. Dermatol. 71: 233–235, 1978
 122. Häkanson, R., S. Leander, F. Sundler, and R. Uddman. P‐Type nerves: purinergic or peptidergic? In: Cellular Basis of Chemical Messengers in the Digestive System, edited by M. I. Grossman, M. A. B. Brazier, and J. Lechago. New York: Academic, 1981, p. 169–198.
 123. Hanko, J., J. E. Hardebo, and C. Owman. Vasomotor effects of some neuropeptides on isolated pial arteries of the cat. In: Cerebral Microcirculation and Metabolism, edited by J. Cervós‐Navarro and E. Fritschka. New York: Raven, 1981, p. 157–161.
 124. Hardebo, J. E. A time study in rat on the opening and reclosure of the blood‐brain barrier after hypertensive or hypertonic insult. Exp. Neurol. 70: 155–166, 1980
 125. Hardebo, J. E. Vasodilatation augments the blood‐brain barrier lesions induced by an acute rise in intracarotid pressure. Blood Vessels 18: 9–15, 1981
 126. Hardebo, J. E., and L. Edvinsson. Adenine compounds: cerebrovascular effects in vitro with reference to their possible involvement in migraine. Stroke 10: 58–62, 1979
 127. Hardebo, J. E., L. Edvinsson, E. T. MacKenzie, and C. Owman. Regional brain uptake of noradrenaline following mechanical or osmotic opening of the blood‐brain barrier. Acta Physiol. Scand. 101: 342–350, 1977
 128. Hardebo, J. E., L. Edvinsson, E. T. MacKenzie, and C. Owman. Histofluorescence study on monoamine entry into the brain before and after opening of the blood‐brain barrier by various mechanisms. Acta Neuropathol. 47: 145–150, 1979
 129. Hardebo, J. E., P. C. Emson, B. Falck, C. Owman, and E. Rosengren. Enzymes related to monoamine metabolism in brain microvessels. J. Neurochem. 35: 1388–1393, 1980
 130. Hardebo, J. E., and C. Owman. Barrier mechanisms for neurotransmitter monoamines and their precursors at the blood‐brain interface. Ann. Neurol. 8: 1–11, 1979
 131. Hardebo, J. E. and C. Owman. Characterization of the in vitro uptake of monoamines into brain microvessels. Acta Physiol. Scand. 108: 223–229, 1980
 132. Hart, M., D. D. Heistad, and M. J. Brody. Effect of chronic hypertension and sympathetic denervation on wall/lumen ratio of cerebral arteries. Hypertension 2: 419–423, 1980
 133. Hartman, B. K., D. Zide, and S. Udenfriend. The use of dopamine‐β‐hydroxylase as a marker for the central noradrenergic nervous system in rat brain. Proc. Natl. Acad. Sci. USA 69: 2722–2726, 1972
 134. Heistad, D. D., and M. L. Marcus (editors). Cerebral Blood Flow: Effect of Nerves and Neurotransmitters. Amsterdam: Elsevier/North‐Holland, 1982 (Dev. Neurosci. Ser., vol. 14.)
 135. Heistad, D., M. Marcus, D. Busija, and S. Sadoshima. Protective effects of sympathetic nerves in the cerebral circulation. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 267–273. (Dev. Neurosci. Ser., vol. 14.)
 136. Hernández‐Pérez, M. J., M. E. Raichle, and H. L. Stone. The role of the peripheral sympathetic nervous system in cerebral blood flow autoregulation. Stroke 6: 284–292, 1975
 137. Heros, R. C., N. T. Zervas, and M. Negoro. Cerebral vasospasm. Surg. Neurol. 5: 354–362, 1976
 138. Higgins, C. B., R. W. Millard, E. Braunwald, and S. F. Vatner. Effects and mechanisms of action of dopamine on regional hemodynamics in the conscious dog. Am. J. Physiol. 225: 432–437, 1973
 139. Hillarp, N.‐Å. Structure of the synapse and the peripheral innervation apparatus of the autonomic nervous system. Acta Anat. Suppl. 4: 1–153, 1946
 140. Hökfelt, T., K. Fuxe, and M. Goldstein. Applications of immunohistochemistry to studies on monoamine cell systems with special reference to nervous tissues. Ann. NY Acad. Sci. 254: 407–432, 1975
 141. Hökfelt, T., O. Johansson, å. Ljungdahl, J. M. Lundberg, and M. Schultzberg. Peptidergic neurones. Nature London 284: 515–521, 1980
 142. Hökfelt, T., S. Vincent, C.‐J. Dalsgaard, L. Skirboll, O. Johansson, M. Schultzberg, J. M. Lundberg, S. Rosell, B. Pernow, and G. Jancso. Distribution of substance P in brain and periphery and its possible role as a co‐transmitter. In: Substance P in the Nervous System, edited by G. Lawrenson. London: Pitman, 1982, p. 84–106. (Ciba Found. Symp. 91.)
 143. Holman, M. E., and A. McLean. The innervation of sheep mesenteric veins. J. Physiol. London 190: 55–69, 1965
 144. Holmstedt, B. A modification of the thiocholine method for the determination of cholinesterase. I. Biochemical evaluation of selective inhibitors. Acta Physiol. Scand. 40: 322–330, 1957
 145. Holmstedt, B. A modification of the thiocholine method for the determination of cholinesterase. II. Histochemical application. Acta Physiol. Scand. 40: 331–337, 1957
 146. Hughes, J., T. W. Smith, H. W. Kosterlitz, L. A. Fothergill, B. A. Morgan, and H. R. Morris. Identification of two related pentapeptides from the brain with potent opiate agonist activity. Nature London 259: 577–579, 1975
 147. Humphrey, C. S., and J. E. Fischer. Peptidergic versus purinergic nerves (Letter to the editor). Lancet 1: 390, 1978
 148. Imbs, J.‐L., and J. Schwartz (editors). Advances in the Biosciences. Peripheral Dopaminergic Receptors. Oxford, UK: Pergamon, 1979, vol. 20.
 149. Iversen, L. L. The Uptake and Storage of Noradrenaline in Sympathetic Nerves. Cambridge, UK: Cambridge Univ. Press, 1967
 150. Järhult, J., P. Hellstrand, and F. Sundler. Immunohistochemical localization and vascular effects of vasoactive intestinal polypeptide in skeletal muscle of the cat. Cell Tissue Res. 207: 55–64, 1980
 151. Johansson, B., B. Ljung, T. Malmfors, and L. Olson. Prejunctional supersensitivity in the rat portal vein as related to its pattern of innervation. Acta Physiol. Scand. Suppl. 349: 5–16, 1970
 152. Kågström, E., L. Granholm, and S. Rehncrona. Metabolite changes in CSF after subarachnoid hemorrhage (Abstract). Excerpta Med. Int. Congr. Ser. 293: 438, 1973
 153. Katzman, R., A. Björklund, C. Owman, U. Stenevi, and K. A. West. Evidence for regenerative axon sprouting of central catecholamine neurons in the rat mesencephalon following electrolytic lesions. Brain Res. 25: 579–596, 1971
 154. Keatinge, W. R. Electrical and mechanical responses of arteries to stimulation of sympathetic nerves. J. Physiol. London 185: 701–715, 1966
 155. Kebabian, J. W., and P. Greengard. Dopamine‐sensitive adenyl cyclase: possible role in synaptic transmission. Science 174: 1346–1349, 1971
 156. Kebabian, J. W., G. L. Petzold, and P. Greengard. Dopamine‐sensitive adenylate cyclase in caudate nucleus of rat brain, and its similarity to the “dopamine receptor.” Proc. Natl. Acad. Sci. USA 69: 2145–2149, 1972
 157. Kendrick, E., B. Öberg, and G. Wennergren. Vasoconstrictor fibre discharge to skeletal muscle, kidney, intestine and skin at varying levels of arterial baroreceptor activity in the cat. Acta Physiol. Scand. 85: 464–476, 1972
 158. Knoll, J. Neuronal peptide (enkephalin) receptors in the ear artery of the rabbit. Eur. J. Pharmacol. 39: 403–407, 1976
 159. Koelle, G. B. Cytological distributions and physiological functions of cholinesterases. In: Handbook of Experimental Pharmacology. Cholinesterases and Anticholinesterase Agents, edited by G. B. Koelle. Berlin: Springer‐Verlag, 1963, vol. 15, p. 187–298.
 160. Korner, P. I. Central nervous control of autonomic cardiovascular function. In: Handbook of Physiology. The Cardiovascular System. The Heart, edited by R. M. Berne and N. Sperelakis. Bethesda, MD: Am. Physiol. Soc., 1979, sect. 2, vol. I, chapt. 20, p. 691–739.
 161. Kostreva, D. R. Functional brain mapping of cardiovascular reflexes using [14C]deoxyglucose in the dog. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 75–86. (Dev. Neurosci. Ser., vol. 14.)
 162. Kóvach, A. G. B., J. Hamar, I. Nyáry, P. Sándor, M. Reivich, E. Dóra, L. Gyulai, and A. Eke. Cerebral blood flow and metabolism in hemorrhagic shock in the baboon. In: Blood Flow and Metabolism in the Brain, edited by A. M. Harper, W. B. Jennett, J. D. Miller, and J. O. Rowan. Edinburgh: Churchill Livingstone, 1975, p. 2. 17–2.19.
 163. Kuschinsky, W., and M. Wahl. Local chemical and neurogenic regulation of cerebral vascular resistance. Physiol. Rev. 58: 656–689, 1978
 164. Lachenmayer, L. Adrenergic innervation of the umbilical vessels. Light‐ and fluorescence microscopic studies. Z. Zellforsch. Mikrosk. Anat. 120: 120–136, 1971
 165. Lai, F. M., and S. Spector. Studies on the monoamine oxidase and catechol‐O‐methyltransferase of the rat cerebral microvessels. Arch. Int. Pharmacodyn. Ther. 233: 227–234, 1978
 166. Lands, A. M., A. Arnold, J. P. McAulliff, F. P. Luduena, and T. G. Brown. Differentiation of receptor systems activated by sympathomimetic amines. Nature London 214: 597–598, 1967
 167. Larsson, B., T. Skärby, L. Edvinsson, J. E. Hardebo, and C. Owman. Possible involvement of microtubules in the vesicular transport through the blood‐brain barrier induced by high intravascular pressure. Neurosci. Lett. 17: 155–159, 1980
 168. Larsson, L.‐I., L. Edvinsson, J. Fahrenkrug, R. Håkanson, C. Owman, O. B. Schaffalitzky de Muckadell, and F. Sundler. Immunohistochemical localization of a vasodilatory polypeptide (VIP) in cerebrovascular nerves. Brain Res. 113: 400–404, 1976
 169. Lee, T. J.‐F. Morphopharmacological study of cerebral vasodilator and constrictor nerves. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 431–439. (Dev. Neurosci. Ser., vol. 14.)
 170. Leeman, S. E., and E. A. Mroz. Substance P. Life Sci. 15: 2033–2044, 1974
 171. Lembeck, F. Zur Frage der zentralen Übertragung afferenter Impulse. III. Mitteilung. Das Vorkommen und die Bedeutung der Substanz P in den dorsalen Wurzeln des Rückenmarks. Naunyn‐Schmiedebergs Arch. Pharmakol. Exp. Pathol. 219: 197–213, 1953
 172. Lembeck, F., and R. Gamse. Substance P in peripheral sensory processes. In: Substance P in the Nervous System, edited by G. Lawrenson. London: Pitman, 1982, p. 35–54. (Ciba Found. Symp. 91.)
 173. Lembeck, F., R. Gamse, and H. Juan. Substance P and sensory nerve endings. In: Substance P, edited by U. S. von Euler and B. Pernow. New York: Raven, 1977, p. 169–181.
 174. Liang, C. C. A possible sympathetic cholinergic mechanism in the renal reflex elicited by stimulation of abdominal viscera in the dog. Clin. Exp. Pharmacol. Physiol. 2: 103–117, 1975
 175. Libet, B. Generation of slow inhibitory and excitatory postsynaptic potentials. Federation Proc. 29: 1945–1956, 1970
 176. Libet, B., and C. Owman. Concomitant changes in formaldehyde‐induced fluorescence of dopamine interneurones and in slow inhibitory postsynaptic potentials of the rabbit superior cervical ganglion, induced by stimulation of the preganglionic nerve or by a muscarinic agent. J. Physiol. London 237: 635–662, 1974
 177. Libet, B., and T. Tosaka. Dopamine as a synaptic transmitter and modulator in sympathetic ganglia: a different mode of synaptic action. Proc. Natl. Acad. Sci. USA 67: 667–673, 1970
 178. Lindvall, M., J. Alumets, L. Edvinsson, J. Fahrenkrug, R. Håkanson, J. Hanko, C. Owman, O. B. Schaffalitzky de Muckadell, and F. Sundler. Peptidergic (VIP) nerves in the mammalian choroid plexus. Neurosci. Lett. 9: 77–82, 1978
 179. Lindvall, M., J. Cervós‐Navarro, L. Edvinsson, C. Owman, and U. Stenevi. Non‐sympathetic perivascular nerves in the brain: origin and mode of innervation studied by fluorescence and electron microscopy combined with stereotaxic lesions and sympathectomy. In: Blood Flow and Metabolism in the Brain, edited by A. M. Harper, W. B. Jennett, J. D. Miller, and J. O. Rowan. Edinburgh: Churchill Livingstone, 1975, p. 1. 7–1.9.
 180. Lindvall, M., J. E. Hardebo, and C. Owman. Barrier mechanisms for neurotransmitter monoamines in the choroid plexus. Acta Physiol. Scand. 108: 215–221, 1980
 181. Lindvall, M., and C. Owman. Autonomic nerves in the mammalian choroid plexus and their influence on the formation of cerebrospinal fluid. J. Cereb. Blood Flow Metab. 1: 245–266, 1981
 182. Lindvall, O., and A. Björklund. The glyoxylic acid fluorescence histochemical method: a detailed account of the methodology for the visualization of central catecholamine neurons. Histochemistry 39: 97–127, 1974
 183. Lindvall, O., and A. Björklund. Organization of catecholamine neurons in the rat central nervous system. In: Handbook of Psychopharmacology. Chemical Pathways in the Brain, edited by S. D. Iversen and S. H. Snyder. New York: Plenum, 1978, vol. 9, p. 139–231.
 184. Ljung, B. Physiological patterns of neuroeffector control mechanisms. In: Vascular Neuroeffector Mechanisms, edited by J. A. Bevan, G. Burnstock, B. Johansson, R. A. Maxwell, and O. A. Nedergaard. Basel: Karger, 1976, p. 143–155.
 185. Loizou, L. A. Uptake of monoamines into central neurones and the blood‐brain barrier in the infant rat. Br. J. Pharmacol. 40: 800–813, 1970
 186. Lorén, I., J. Alumets, R. Håkanson, and F. Sundler. Immunoreactive pancreatic polypeptide (PP) occurs in the central and peripheral nervous system: preliminary immunocytochemical observations. Cell Tissue Res. 200: 179–186, 1979
 187. Lundberg, J. M. Evidence for coexistence of vasoactive intestinal polypeptide (VIP) and acetylcholine in neurons of cat exocrine glands. Morphological, biochemical and functional studies. Acta Physiol. Scand. Suppl. 406: 1–57, 1981
 188. MacKenzie, E. T., J. McCulloch, M. O'Keane, J. D. Pickard, and A. M. Harper. Cerebral circulation and norepinephrine: relevance of the blood‐brain barrier. Am. J. Physiol. 231: 483–488, 1976
 189. MacKenzie, E. T., J. McCulloch, J. D. Pickard, and A. M. Harper. Cerebral circulatory and metabolic effects of endogenous noradrenaline. In: Blood Flow and Metabolism in the Brain, edited by M. Harper, B. Jennett, D. Miller, and J. Rowan. Edinburgh: Churchill Livingstone, 1975, p. 1. 28–1.32.
 190. MacKenzie, E. T., A. P. McGeorge, D. I. Graham, W. Fitch, L. Edvinsson, and A. M. Harper. Effects of increasing arterial pressure on cerebral blood flow in the baboon: influence of the sympathetic nervous system. Pfluegers Arch. 378: 189–195, 1979
 191. MacKenzie, E. T., J. D. Pickard, and J. E. Hardebo. Blood‐brain barrier, amines and the cerebral circulation: problems and approaches. In: Neurogenic Control of the Brain Circulation, edited by C. Owman and L. Edvinsson. Oxford, UK: Pergamon, 1977, p. 231–243. (Wenner‐Gren Center Int. Symp. Ser., vol. 30.)
 192. MacKenzie, E. T., M. Stewart, A. R. Young, and A. M. Harper. Cerebrovascular actions of serotonin. Adv. Neurol. 20: 77–83, 1978
 193. Matsuo, Y. Integration of hormonal and neural control of gastrointestinal secretion. Asian Med. J. 19: 612–629, 1976
 194. McCubbin, J. W., Y. Kaneko, and I. H. Page. Inhibition of neurogenic vasoconstriction by serotonin. Circ. Res. 11: 74–83, 1962
 195. McCulloch, J., H. E. Savaki, and W. Angerson. Regional water permeability in the CNS of conscious rats: effects of hypercapnia and locus coeruleus lesions. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 509–516. (Dev. Neurosci. Ser., vol. 14.)
 196. McGrath, M. A. 5‐Hydroxytryptamine and neurotransmitter release in canine blood vessels. Inhibition by low and augmentation by high concentrations. Circ. Res. 41: 428–435, 1977
 197. McGrath, M. A., and J. T. Shepherd. Inhibition of adrenergic neurotransmission in canine vascular smooth muscle by histamine. Mediation by H2‐receptors. Circ. Res. 39: 566–573, 1976
 198. McGrath, M. A., and P. M. Vanhoutte. Vasodilatation caused by peripheral inhibition of adrenergic neurotransmission. In: Mechanism of Vasodilatation, edited by P. M. Vanhoutte and I. Leusen. Basel: Karger, 1978, p. 248–257.
 199. McKee, J. C., M. J. Denn, and H. L. Stone. Neurogenic cerebral vasodilatation from electrical stimulation of the cerebellum in the monkey. Stroke 7: 179–186, 1976
 200. McKenna, O. A., and E. T. Angelakos. Acetylcholinesterase‐containing nerve fibres in the canine kidney. Circ. Res. 23: 645–651, 1968
 201. McNay, J. L., and L. J. Goldberg. Comparison of the effects of dopamine, isoproterenol, norepinephrine and bradykinin on canine renal and femoral blood flow. J. Pharmacol. Exp. Ther. 151: 23–31, 1966
 202. Mecca, T. E., and R. C. Webb. Serotonin and vasodilatation. Bibl. Cardiol. 38: 81–90, 1984
 203. Miura, M., and D. J. Reis. Cerebellum: a pressor response elicited from the fastigial nucleus and its efferent pathway in brainstem. Brain Res. 13: 595–599, 1969
 204. Miura, M., and D. J. Reis. A blood pressure response from fastigial nucleus and its relay pathway in brainstem. Am. J. Physiol. 219: 1330–1336, 1970
 205. Monnier, M. Les centres végétatifs bulbaires. Arch. Int. Physiol. 49: 455–463, 1939
 206. Mueller, S. M., and D. B. Rusterholz. “Trophic” influence of sympathetic nerves on the peripheral and cerebral vasculature. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 317–325. (Dev. Neurosci. Ser., vol. 14.)
 207. Myers, H. A., E. A. Schenk, and C. R. Honig. Ganglion cells in arterioles of skeletal muscle: role in sympathetic vasodilation. Am. J. Physiol. 229: 126–138, 1975
 208. Nelson, E., and M. Rennels. Neuromuscular contacts in intracranial arteries of the cat. Science 167: 301–302, 1969
 209. Nielsen, K. C., and C. Owman. Adrenergic innervation of pial arteries related to the circle of Willis in the cat. Brain Res. 6: 773–776, 1967
 210. Nielsen, K. C., C. Owman, and B. Sporrong. Ultrastructure of the autonomic innervation apparatus in the main pial arteries of rats and cats. Brain Res. 27: 25–32, 1971
 211. Norberg, K.‐A., and B. Hamberger. The sympathetic adrenergic neuron. Some characteristics revealed by histochemical studies on the intraneuronal distribution of the transmitter. Acta Physiol. Scand. Suppl. 238: 1–42, 1964
 212. Oldendorf, W. H. Brain uptake of radiolabeled amino acids, amines, and hexoses after arterial injection. Am. J. Physiol. 221: 1629–1639, 1971
 213. Olson, L., M. Álund, and K. A. Norberg. Fluorescence‐microscopical demonstration of a population of gastro‐intestinal nerve fibres with a selective affinity for quinacrine. Cell Tissue Res. 171: 407–423, 1976
 214. Otsuka, M., S. Konishi, M. Yanagisawa, A. Tsunoo, and H. Akagi. Role of substance P as a sensory transmitter in spinal cord and sympathetic ganglia. In: Substance P in the Nervous System, edited by G. Lawrenson. London: Pitman, 1982, p. 13–34. (Ciba Found. Symp. 91.)
 215. Owman, C. Neurogenic vasodilatation mediated by the autonomic nervous system. Triangle Engl. Ed. 18: 89–99, 1979
 216. Owman, C. Autonomic innervation of blood vessels, with special emphasis on human cerebrovascular nerves and corresponding amine receptors. Gen. Pharmacol. 14: 17–20, 1983
 217. Owman, C., S. Aronson, G. Gennser, and N.‐O. Sjöberg. Histochemical and pharmacological evidence of amine mechanisms in human fetal vascular shunts. In: Fetal Pharmacology, edited by L. O. Boréus. New York: Raven, 1973, p. 179–191.
 218. Owman, C., and L. Edvinsson (editors). Neurogenic Control of the Brain Circulation. Oxford, UK: Pergamon, 1977 (Wenner‐Gren Cent. Int. Symp. Ser., vol. 30.)
 219. Owman, C., and L. Edvinsson. Histochemical and pharmacological approach to the investigation of neurotransmitters, with particular regard to the cerebrovascular bed. In: Neurogenic Control of the Brain Circulation, edited by C. Owman and L. Edvinsson. Oxford, UK: Pergamon, 1977, p. 15–38. (Wenner‐Gren Cent. Int. Symp. Ser., vol. 30.)
 220. Owman, C., L. Edvinsson, and J. E. Hardebo. Pharmacological in vitro analysis of amine‐mediated vasomotor functions in the intracranial and extracranial vascular beds. Blood Vessels 15: 128–147, 1978
 221. Owman, C., L. Edvinsson, and J. E. Hardebo. Amine mechanisms and contractile properties of the cerebral microvascular endothelium. In: Vascular Neuroeffector Mechanisms, edited by J. A. Bevan, T. Godfraind, R. A. Maxwell, and P. M. Vanhoutte. New York: Raven, 1980, p. 277–290.
 222. Owman, C., L. Edvinsson, J. E. Hardebo, U. Gröschel‐Stewart, K. Unsicker, and B. Walles. Immunohistochemical demonstration of actin and myosin in brain capillaries. Acta Neurol. Scand. Suppl. 64: 384–385, 1977
 223. Owman, C., L. Edvinsson, J. E. Hardebo, B. Larsson, B. Nilsson, and T. Skärby. Parasympathomimetic effects on brain circulation and metabolism. In: Cerebral Circulation and Neurotransmitters, edited by A. Bès and G. Géraud. Amsterdam: Excerpta Med., 1980, p. 191–198. (Int. Congr. Ser. 507.)
 224. Owman, C., L. Edvinsson, M. Lindvall, and N.‐O. Sjöberg. Influence of external light conditions on norepinephrine levels in organs innervated by sympathetic nerves from different levels. Brain Res. Bull. 9: 777–779, 1982
 225. Owman, C., and B. Falck. Functional approach to the histochemistry of catecholamines in peripheral sympathetic nerves. In: Golgi Centennial Symposium: Perspectives in Neurobiology, edited by M. Santini. New York: Raven, 1975, p. 415–436.
 226. Owman, C., and J. E. Hardebo. Mechanisms of cerebral vasodilatation: amines, peptides, and the blood‐brain barrier. In: Vasodilatation, edited by P. M. Vanhoutte and I. Leusen. New York: Raven, 1981, p. 159–179.
 227. Owman, C., and M. Lindvall. Sympathetic nervous control of CSF production in normal and hydrocephalic animals, and its relationship to cerebral blood volume and intracranial pressure. In: Intracranial Pressure V, edited by S. Ishii, H. Nagai, and M. Brock. Berlin: Springer‐Verlag, 1983, p. 10–28.
 228. Palkovits, M. The anatomy of central cardiovascular neurons. In: Central Adrenaline Neurons: Basic Aspects and Their Role in Cardiovascular Disease, edited by K. Fuxe, M. Goldstein, B. Hökfelt, and T. Hökfelt. Oxford, UK: Pergamon, 1980, p. 3–17. (Wenner‐Gren Cent. Int. Symp. Ser., vol. 33.)
 229. Palkovits, M. Neuropeptides and biogenic amines in central cardiovascular control mechanisms. In: Perspectives in Cardiovascular Research. Central Nervous System Mechanisms in Hypertension, edited by J. P. Buckley and C. M. Ferrario. New York: Raven, 1981, vol. 6, p. 73–87.
 230. Palkovits, M. Neuropeptides in the central regulation of blood pressure. In: Developments in Cardiovascular Medicine. Pathophysiology of Hypertension, edited by H. Villarreal and M. P. Sambhi. The Hague: Nijhoff, 1984, vol. 30, p. 282–290.
 231. Peerless, S. J., and M. J. Kendall. Experimental cerebral vasospasm. In: Proceedings of the Ninth Princeton Conference on Cerebral Vascular Disease, edited by J. P. Whistnant and B. A. Sandok. New York: Grune & Stratton, 1974, p. 49–58.
 232. Pernow, B. Studies on substance P. Purification, occurrence and biological actions. Acta Physiol. Scand. Suppl. 105: 1–90, 1953
 233. Pernow, B., and S. Rosell. Effect of substance P on blood flow in canine adipose tissue and skeletal muscle. Acta Physiol. Scand. 93: 139–148, 1975
 234. Peroutka, S. J., R. M. Lebovitz, and S. H. Snyder. Two distinct central serotonin receptors with different physiological function. Science 212: 827–829, 1981
 235. Raichle, M. E., R. L. Grubb, Jr., and J. O. Eichling. Neural and hormonal regulation of brain water permeability. In: Neurogenic Control of the Brain Circulation, edited by C. Owman and L. Edvinsson. Oxford, UK: Pergamon, 1977, p. 465–470. (Wenner‐Gren Cent. Int. Symp. Ser., vol. 30.)
 236. Raichle, M. E., B. K. Hartman, J. O. Eichling, and L. G. Sharpe. Central noradrenergic regulation of cerebral blood flow and vascular permeability. Proc. Natl. Acad. Sci. USA 72: 3726–3730, 1975
 237. Rapoport, S. I. Blood‐Brain Barrier in Physiology and Medicine. New York: Raven, 1976
 238. Raynor, R. B., and J. G. McMurtry. Prevention of serotonin‐producing cerebral vasospasm. J. Neurosurg. 20: 94–96, 1963
 239. Reis, D. J., C. Iadecola, E. MacKenzie, M. Mori, M. Nakai, and L. W. Tucker. Primary and metabolically coupled cerebrovascular dilation elicited by stimulation of two intrinsic systems of brain. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 475–484. (Dev. Neurosci. Ser., vol. 14.)
 240. Rennels, M. L., and E. Nelson. Capillary innervation in the mammalian central nervous system: an electron microscopic demonstration. Am. J. Anat. 144: 233–241, 1975
 241. Robinson, R. G., F. E. Bloom, and E. L. F. Battenberg. A fluorescent histochemical study of changes in noradrenergic neurons following experimental cerebral infarction in the rat. Brain Res. 132: 259–272, 1977
 242. Robinson, R. G., W. J. Shoemaker, M. Schlumpf, T. Valk, and F. E. Bloom. Effect of experimental infarction in rat brain on catecholamines and behaviour. Nature London 255: 332–334.
 243. Robison, G. A., R. W. Butcher, and E. W. Sutherland. Cyclic AMP. New York: Academic, 1971
 244. Rosell, S., E. Burcher, D. Chang, and K. Folkers. Cardiovascular and metabolic actions of neurotensin and (Glu4)‐neurotensin. Acta Physiol. Scand. 98: 484–491, 1976
 245. Rosenblueth, A., and W. B. Cannon. The chemical mediation of sympathetic vasodilator nerve impulses. Am. J. Physiol. 112: 33–40, 1935
 246. Ryan, M. J., K. E. Clark, and M. J. Brody. Neurogenic and mechanical control of canine uterine vascular resistance. Am. J. Physiol. 227: 547–555, 1974
 247. Sadoshima, S., D. Busija, M. Brody, and D. Heistad. Protection against stroke by sympathetic nerves. In: Cerebral Blood Flow: Effect of Nerves and Neurotransmitters, edited by D. D. Heistad and M. L. Marcus. Amsterdam: Elsevier/North‐Holland, 1982, p. 309–315. (Dev. Neurosci. Ser., vol. 14.)
 248. Said, S. I., and V. Mutt. Polypeptide with broad biological activity: isolation from small intestine. Science 169: 1217–1218, 1970
 249. Said, S. I., and V. Mutt. Potent peripheral and splanchnic vasodilator peptide from normal gut. Nature London 225: 863–865, 1970
 250. Said, S. I., and V. Mutt. Isolation from porcine‐intestinal wall of a vasoactive octacosapeptide related to secretin and to glucagon. Eur. J. Biochem. 28: 199–204, 1972
 251. Schenk, E. A., and A. El‐Badawi. Dual innervation of arteries and arterioles. A histochemical study. Z. Zellforsch. Mikrosk. Anat. 91: 170–177, 1968
 252. Schmidt, C. F., and J. P. Hendrix. The action of chemical substances on cerebral blood vessels. Res. Publ. Assoc. Res. Nerv. Ment. Dis. 18: 229–276, 1937
 253. Schuelke, D. M., A. L. Mark, P. G. Schmid, and J. W. Eckstein. Coronary vasodilatation produced by dopamine after adrenergic blockade. J. Pharmacol. Exp. Ther. 176: 320–327, 1971
 254. Schultzberg, M., T. Hökfelt, G. Nilsson, L. Terenius, J. F. Rehfeld, M. Brown, R. Elde, M. Goldstein, and S. Said. Distribution of peptide‐ and catecholamine‐containing neurons in the gastro‐intestinal tract of rat and guinea‐pig: immunohistochemical studies with antisera to substance P, vasoactive intestinal polypeptide, enkephalins, somatostatin, gastrin/cholecystokinin, neurotensin and dopamine‐β‐hydroxylase. Neuroscience 5: 689–744, 1980
 255. Sercombe, R., P. Aubineau, L. Edvinsson, C. Grivas, H. Mamo, C. Owman, E. Pinard, and J. Seylaz. CBF changes by sympathetic nerve stimulation and denervation: correlation with the degree of sympathetic innervation of cerebral arterioles. In: Blood Flow and Metabolism in the Brain, edited by A. M. Harper, W. B. Jennett, J. D. Miller, and J. O. Rowan. Edinburgh: Churchill Livingstone, 1975, p. 2. 25–2.29.
 256. Sercombe, R., P. Aubineau, L. Edvinsson, H. Mamo, C. Owman, E. Pinard, and J. Seylaz. Neurogenic influence on local cerebral blood flow. Effect of catecholamines or sympathetic stimulation as correlated with the sympathetic innervation. Neurology 25: 954–963, 1975
 257. Sercombe, R., P. Aubineau, L. Edvinsson, H. Mamo, C. Owman, and J. Seylaz. Pharmacological evidence in vitro and in vivo for functional beta1 receptors in the cerebral circulation. Eur. J. Physiol. 368: 241–244, 1977
 258. Shimazu, K., F. Gotoh, S. Nakajima, S. Komatsumoto, and M. Ichijo. Demonstration of autonomic action potential from cerebral artery. Acta Neurol. Scand. Suppl. 72: 98–99, 1979
 259. Simpson, F. O., and C. E. Devine. The fine structure of autonomic neuromuscular contacts in arterioles of sheep renal cortex. J. Anat. 100: 127–137, 1966
 260. Sladek, J. R., Jr., and A. Björklund(editors). Monoamine Transmitter Histochemistry. A Twenty Year Commemoration. Brain Res. Bull. 9: 1–829, 1982
 261. Sokoloff, L. The action of drugs on the cerebral circulation. Pharmacol. Rev. 11: 1–85, 1959
 262. Sokoloff, L. The radioactive deoxyglucose method. Theory, procedure, and applications for the measurement of local glucose utilization in the central nervous system. Adv. Neurochem. 4: 1–82, 1982
 263. Steinbusch, H. W. M., and A. H. Mulder. Immunohistochemical localization of histamine in neurons and mast cells in the rat brain. In: Handbook of Chemical Neuroanatomy, edited by A. Björklund, T. Hökfelt, and M. Kuhar. Amsterdam: Elsevier/North‐Holland, 1984, p. 126–140.
 264. Steinbusch, H. W. M., A. A. J. Verhofstad, and H. W. J. Joosten. Localization of serotonin in the central nervous system by immunohistochemistry: description of a specific and sensitive technique and some applications. Neuroscience 3: 811–819, 1978
 265. Sternberger, L. A. Immunocytochemistry. Englewood Cliffs, NJ: Prentice‐Hall, 1974
 266. Su, C., J. A. Bevan, and G. Burnstock. [3H]‐adenosine triphosphate: release during stimulation of enteric nerves. Science 173: 336–338, 1971
 267. Sundler, F., E. Moghimzadeh, R. Håkanson, M. Ekelund, and P. Emson. Nerve fibers in the gut and pancreas of the rat displaying neuropeptide‐Y immunoreactivity. Intrinsic and extrinsic origin. Cell Tissue Res. 230: 487–493, 1983
 268. Sutherland, E. W., and G. A. Robison. The role of cyclic‐3′,5′‐AMP in responses to catecholamines and other hormones. Pharmacol. Rev. 18: 145–161, 1966
 269. Svendgaard, N.‐A., L. Edvinsson, C. Owman, and C. Sahlin. Increased sensitivity of the basilar artery to norepinephrine and 5‐hydroxytryptamine following experimental subarachnoid hemorrhage. Surg. Neurol. 8: 191–195, 1977
 270. Swanson, L. W., M. A. Connelly, and B. K. Hartman. Ultrastructural evidence for central monoaminergic innervation of blood vessels in the paraventricular nucleus of the hypothalamus. Brain Res. 136: 166–173, 1977
 271. Tindall, G. T., and J. C. Greenfield. The effects of intraarterial histamine on blood flow in the internal and external carotid artery of man. Stroke 4: 46–49, 1973
 272. Uddman, R., J. Alumets, L. Edvinsson, R. Håkanson, and F. Sundler. VIP nerve fibres around peripheral blood vessels. Acta Physiol. Scand. 112: 65–70, 1981
 273. Uddman, R., L. Edvinsson, R. Håkanson, C. Owman, and F. Sundler. Immunohistochemical demonstration of APP (avian pancreatic polypeptide)‐immunoreactive nerve fibres around cerebral blood vessels. Brain Res. Bull. 9: 715–718, 1982
 274. Uddman, R., L. Edvinsson, C. Owman, and F. Sundler. Perivascular substance P: occurrence and distribution in mammalian pial vessels. J. Cereb. Blood Flow Metab. 1: 227–232, 1981
 275. Uddman, R., L. Edvinsson, C. Owman, and F. Sundler. Nerve fibres containing gastrin‐releasing peptide (GRP) around pial vessels. J. Cereb. Blood Flow Metab. 3: 386–390, 1983
 276. Uddman, R., O. Ninoyu, and F. Sundler. Adrenergic and peptidergic innervation of cochlear blood vessels. Acta Oto‐Rhino‐Laryngol. Belg. 236: 7–14, 1982
 277. Uvnäs, B. Cholinergic vasodilator nerves. Federation Proc. 25: 1618–1622, 1966
 278. Van Nueten, J. M., P. A. J. Janssen, J. Van Beek, R. Xhonneux, T. J. Verbeuren, and P. M. Vanhoutte. Vascular effects of ketanserin (R 41 468), a novel antagonist of 5‐HT2 serotonergic receptors. J. Pharmacol. Exp. Ther. 218: 217–230, 1981
 279. Vasquez, J., and M. J. Purves. Studies on the dilator pathway to cerebral blood vessels. In: Neurogenic Control of the Brain Circulation, edited by C. Owman and L. Edvinsson. Oxford, UK: Pergamon, 1977, p. 59–73. (Wenner‐Gren Cent. Int. Symp. Ser., vol. 30.)
 280. Vatner, S. F., L. L. Priano, J. D. Rutherford, and W. T. Manders. Sympathetic regulation of the cerebral circulation by the carotid chemoreceptor reflex. Am. J. Physol. 238 (Heart Circ. Physiol. 7): H594–H598, 1980
 281. Verhofstad, A. A. J., H. W. M. Steinbusch, B. Penke, J. Varga, and H. W. J. Joosten. Use of antibodies to norepinephrine and epinephrine in immunohistochemistry. In: Advances in Biochemical Psychopharmacology. Histochemistry and Cell Biology of Autonomic Neurons, SIF Cells, and Paraneurons, edited by O. Eränkö, S. Soinila, and H. Päivärinta. New York: Raven, 1980, vol. 25, p. 185–193.
 282. Verity, M. A. Morphologic studies of the vascular neuroeffector apparatus. In: Physiology and Pharmacology of Vascular Neuroeffector Systems, edited by J. A. Bevan, R. F. Furchgott, R. A. Maxwell, and A. P. Somlyo. Basel: Karger, 1971, p. 2–12.
 283. Viveros, O. H., D. G. Garlick, and E. M. Renkin. Sympathetic beta adrenergic vasodilatation in skeletal muscle of the dog. Am. J. Physiol. 215: 1218–1225, 1968
 284. Walsh, J. A. Antagonism by methysergide of vascular effects of 5‐hydroxytryptamine in man. Br. J. Pharmacol. 30: 518–530, 1967
 285. Wang, S. C., and S. W. Ranson. Autonomic responses to electrical stimulation of the lower brain stem. J. Comp. Neurol. 71: 437–455, 1939
 286. Watters, J. W. The effect of bradykinin and histamine on cerebral arteries of monkeys. Radiology 98: 299–304, 1971
 287. Wei, E. P., H. A. Kontos, and S. I. Said. Mechanism of action of vasoactive intestinal polypeptide on cerebral arterioles. Am. J. Physiol. 239 (Heart Circ. Physiol. 8): H765–H768, 1980
 288. Wilkins, R. H. Cerebral Arterial Spasm. Baltimore, MD: Williams & Wilkins, 1980
 289. Willems, J. L., and M. G. Bogaert. Ganglionic inhibition by dopamine and neurogenic vasodilatation. In: Mechanism of Vasodilatation, edited by P. M. Vanhoutte and I. Leusen. Basel: Karger, 1978, p. 241–247.
 290. Williams, L. T., and R. L. Lefkowitz. Receptor Binding Studies in Adrenergic Pharmacology. New York: Raven, 1978
 291. Willis, E., B. Ottesen, G. Wagner, F. Sundler, and J. Fahrenkrug. Vasoactive intestinal polypeptide (VIP) as a possible neurotransmitter involved in penile erection. Acta Physiol. Scand. 113: 545–547, 1981
 292. Wilson, D. A., J. T. O'Neil, S. I. Said, and R. J. Traystman. Vasoactive intestinal polypeptide and the canine cerebral circulation. Circ. Res. 48: 138–148, 1981
 293. Woolley, D. W., and E. Shaw. Antimetabolites of serotonin. J. Biol. Chem. 203: 69–79, 1953
 294. Wurtman, R. J., J. Axelrod, and D. E. Kelly. The Pineal. New York: Academic, 1968
 295. Wurtman, R. J., and N. T. Zervas. Monoamine neurotransmitters and the pathophysiology of stroke and central nervous system trauma. J. Neurosurg. 40: 34–36, 1974
 296. Yeh, B. K., J. L. McNay, and L. I. Goldberg. Attenuation of dopamine renal and mesenteric vasodilation by haloperidol: evidence for a specific dopamine receptor. J. Pharmacol. Exp. Ther. 168: 303–309, 1969

Contact Editor

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

* Required Field

How to Cite

Christer Owman. Neurogenic Control of the Vascular System: Focus on Cerebral Circulation. Compr Physiol 2011, Supplement 4: Handbook of Physiology, The Nervous System, Intrinsic Regulatory Systems of the Brain: 525-580. First published in print 1986. doi: 10.1002/cphy.cp010410